1
|
Shovlin S, Young LS, Varešlija D. Hormonal and neuronal interactions shaping the brain metastatic microenvironment. Cancer Lett 2025; 624:217739. [PMID: 40288563 DOI: 10.1016/j.canlet.2025.217739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Metastatic progression drives the majority of cancer-related fatalities, and involvement of the central nervous system (CNS) poses especially formidable challenges to patients and clinicians. Brain metastases (BrM), commonly originate from lung, breast and melanoma cancers, and carry disproportionately poor outcomes. Although therapeutic advances have extended survival for many extracranial tumors, BrM incidence continues to climb-underscoring critical knowledge gaps in understanding the unique biology of tumor colonization in the CNS. While definitive evidence remains limited, a growing focus on cancer neuroscience-especially regarding hormone dependent cancer cells in the brain-has begun to reveal that factors normally regulated by sex steroids and neurosteroids may similarly influence the specialized metastatic microenvironment in the CNS. Steroid hormones can permeate the blood-brain barrier (BBB) or be synthesized de novo by astrocytes and other CNS-resident cells, potentially influencing processes such as inflammation, synaptic plasticity, and immune surveillance. However, how these hormonal pathways are co-opted by disseminated cancer cells remains unclear. Here, we review the complex hormonal landscape of the adult brain and examine how neuroendocrine-immune interactions, often regulated by sex hormones, may support metastatic growth. We discuss the interplay between systemic hormones, local steroidogenesis, and tumor adaptation to identify novel therapeutic opportunities.
Collapse
Affiliation(s)
- Stephen Shovlin
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Leonie S Young
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland; Beaumont RCSI Cancer Centre, Beaumont Hospital, Dublin, Ireland.
| | - Damir Varešlija
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland; Beaumont RCSI Cancer Centre, Beaumont Hospital, Dublin, Ireland.
| |
Collapse
|
2
|
Ma W, Oliveira-Nunes MC, Xu K, Kossenkov A, Reiner BC, Crist RC, Hayden J, Chen Q. Type I interferon response in astrocytes promotes brain metastasis by enhancing monocytic myeloid cell recruitment. Nat Commun 2023; 14:2632. [PMID: 37149684 PMCID: PMC10163863 DOI: 10.1038/s41467-023-38252-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 04/20/2023] [Indexed: 05/08/2023] Open
Abstract
Cancer metastasis to the brain is a significant clinical problem. Metastasis is the consequence of favorable interactions between invaded cancer cells and the microenvironment. Here, we demonstrate that cancer-activated astrocytes create a sustained low-level activated type I interferon (IFN) microenvironment in brain metastatic lesions. We further confirm that the IFN response in astrocytes facilitates brain metastasis. Mechanistically, IFN signaling in astrocytes activates C-C Motif Chemokine Ligand 2 (CCL2) production, which further increases the recruitment of monocytic myeloid cells. The correlation between CCL2 and monocytic myeloid cells is confirmed in clinical brain metastasis samples. Lastly, genetically or pharmacologically inhibiting C-C Motif Chemokine Receptor 2 (CCR2) reduces brain metastases. Our study clarifies a pro-metastatic effect of type I IFN in the brain even though IFN response has been considered to have anti-tumor effects. Moreover, this work expands our understandings on the interactions between cancer-activated astrocytes and immune cells in brain metastasis.
Collapse
Affiliation(s)
- Weili Ma
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Maria Cecília Oliveira-Nunes
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, 19104, USA
- Carisma Therapeutics, Philadelphia, PA, 19104, USA
| | - Ke Xu
- MD/PhD Program, Boston University School of Medicine, Boston, MA, 02215, USA
| | - Andrew Kossenkov
- Gene Expression & Regulation Program, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Benjamin C Reiner
- Department of Psychiatry, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Richard C Crist
- Department of Psychiatry, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - James Hayden
- Imaging Shared Resource, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Qing Chen
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, 19104, USA.
| |
Collapse
|
3
|
Spampinato SF, Merlo S, Costantino G, Sano Y, Kanda T, Sortino MA. Decreased Astrocytic CCL2 Accounts for BAF-312 Effect on PBMCs Transendothelial Migration Through a Blood Brain Barrier in Vitro Model. J Neuroimmune Pharmacol 2022; 17:427-436. [PMID: 34599741 PMCID: PMC9810569 DOI: 10.1007/s11481-021-10016-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/20/2021] [Indexed: 01/13/2023]
Abstract
Disruption of the blood brain barrier (BBB) is a common event in several neurological diseases and in particular, in multiple sclerosis (MS), it contributes to the infiltration of the central nervous system by peripheral inflammatory cells. Sphingosine-1-phosphate (S1P) is a bioactive molecule with pleiotropic effects. Agonists of S1P receptors such as fingolimod and siponimod (BAF-312) are in clinical practice for MS and have been shown to preserve BBB function in inflammatory conditions. Using an in vitro BBB model of endothelial-astrocytes co-culture exposed to an inflammatory insult (tumor necrosis factor-α and interferon-γ; T&I), we show that BAF-312 reduced the migration of peripheral blood mononuclear cells (PBMCs) through the endothelial layer, only in the presence of astrocytes. This effect was accompanied by decreased expression of the adhesion molecule ICAM-1. BAF-312 also reduced the activation of astrocytes, by controlling NF-kB and NLRP3 induction and preventing the increase of proinflammatory cytokine and chemokines. Reduction of CCL2 by BAF-312 may be responsible for the observed effects and, accordingly, addition of exogenous CCL2 was able to counteract BAF-312 effects and rescued T&I responses on PBMC migration, ICAM-1 expression and astrocyte activation. The present results further point out BAF-312 effects on BBB properties, suggesting also the key role of astrocytes in mediating drug effects on endothelial function.
Collapse
Affiliation(s)
- Simona F. Spampinato
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Sara Merlo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giuseppe Costantino
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy ,Ph.D. Program in Neuroscience and Education, DISCUM, University of Foggia, 71121 Foggia, Italy
| | - Yasuteru Sano
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Takashi Kanda
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Maria Angela Sortino
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| |
Collapse
|
4
|
Hajal C, Shin Y, Li L, Serrano JC, Jacks T, Kamm RD. The CCL2-CCR2 astrocyte-cancer cell axis in tumor extravasation at the brain. SCIENCE ADVANCES 2021; 7:eabg8139. [PMID: 34162553 PMCID: PMC8221620 DOI: 10.1126/sciadv.abg8139] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/11/2021] [Indexed: 05/12/2023]
Abstract
Although brain metastases are common in cancer patients, little is known about the mechanisms of cancer extravasation across the blood-brain barrier (BBB), a key step in the metastatic cascade that regulates the entry of cancer cells into the brain parenchyma. Here, we show, in a three-dimensional in vitro BBB microvascular model, that astrocytes promote cancer cell transmigration via their secretion of C-C motif chemokine ligand 2 (CCL2). We found that this chemokine, produced primarily by astrocytes, promoted the chemotaxis and chemokinesis of cancer cells via their C-C chemokine receptor type 2 (CCR2), with no notable changes in vascular permeability. These findings were validated in vivo, where CCR2-deficient cancer cells exhibited significantly reduced rates of arrest and transmigration in mouse brain capillaries. Our results reveal that the CCL2-CCR2 astrocyte-cancer cell axis plays a fundamental role in extravasation and, consequently, metastasis to the brain.
Collapse
Affiliation(s)
- Cynthia Hajal
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yoojin Shin
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Leanne Li
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jean Carlos Serrano
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Tyler Jacks
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Roger D Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
5
|
Yang J, Wang L, Xu Z, Wu L, Liu B, Wang J, Tian D, Xiong X, Chen Q. Integrated Analysis to Evaluate the Prognostic Value of Signature mRNAs in Glioblastoma Multiforme. Front Genet 2020; 11:253. [PMID: 32296458 PMCID: PMC7136556 DOI: 10.3389/fgene.2020.00253] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 03/02/2020] [Indexed: 12/13/2022] Open
Abstract
Background Gliomas are the most common intracranial tumors and are classified as I-IV. Among them, glioblastoma multiforme (GBM) is the most common invasive glioma with a poor prognosis. New molecular biomarkers that can predict clinical outcomes in GBM patients must be identified, which will help comprehend their pathogenesis and supply personalized treatment. Our research revealed four powerful survival indicators in GBM by reanalyzing microarray data and genetic sequencing data in public databases. Moreover, it unraveled new potential therapeutic targets which could help improve the survival time and quality of life of GBM patients. Materials and Methods To identify prognostic signatures in GBMs, we analyzed the gene profiling data of GBM and standard brain samples from the Gene Expression Omnibus, including four datasets and RNA sequencing data from The Cancer Genome Atlas (TCGA) containing 152 glioblastoma tissues. We performed the differential analysis, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, weighted gene co-expression network analysis (WGCNA) and Cox regression analysis. Results After differential analysis in GSE12657, GSE15824, GSE42656 and GSE50161, overlapping differentially expressed genes were identified. We identified 110 up-regulated DEGs and 75 down-regulated DEGs in the GBM samples. Significantly enriched subclasses of the GO classification of these genes included mitotic sister chromatid separation, mitotic nuclear division and so on. In KEGG pathway analysis, the most abundant terms were ECM-receptor interaction and protein digestion and absorption. WGCNA analysis was performed on these 185 DEGs in 152 glioblastoma samples obtained from TCGA, and gene co-expression networks were constructed. We then performed a multivariate Cox analysis and established a Cox proportional hazards regression model using the top 20 genes significantly correlated with survival time. We identified a four-protein prognostic signature that could divide patients into high-risk and low-risk groups. Increased expression of SLC12A5, CCL2, IGFBP2, and PDPN was associated with increased risk scores. Finally, the K-M curves confirmed that these genes could be used as independent predictors of survival in patients with glioblastoma. Conclusion Our analytical study identified a set of potential biomarkers that could predict survival and may contribute to successful treatment of GBM patients.
Collapse
Affiliation(s)
- Ji'an Yang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Long Wang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhou Xu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Liquan Wu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Baohui Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Junmin Wang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Daofeng Tian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
6
|
Contreras-Zárate MJ, Cittelly DM. Sex steroid hormone function in the brain niche: Implications for brain metastatic colonization and progression. Cancer Rep (Hoboken) 2020; 5:e1241. [PMID: 33350105 PMCID: PMC8022872 DOI: 10.1002/cnr2.1241] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 01/04/2020] [Accepted: 01/30/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND While sex hormones and their receptors play well-known roles in progression of primary tumors through direct action on sex steroid hormone-responsive cancer cells, emerging evidence suggest that hormones also play important roles in metastatic progression by modulating the tumor microenvironment. Estrogens and androgens synthesized in gonads and within the brain influence memory, behavior, and outcomes of brain pathologies. Yet, their impact on brain metastatic colonization and progression is just beginning to be explored. RECENT FINDINGS Estradiol and testosterone cross the blood-brain barrier and are synthesized de novo in astrocytes and other cells within the adult brain. Circulating and brain-synthesized estrogens have been shown to promote brain metastatic colonization of tumors lacking estrogen receptors (ERs), through mechanisms involving the upregulation of growth factors and neurotrophins in ER+ reactive astrocytes. In this review, we discuss additional mechanisms by which hormones may influence brain metastases, through modulation of brain endothelial cells, astrocytes, and microglia. CONCLUSION A greater understanding of hormone-brain-tumor interactions may shed further light on the mechanisms underlying the adaptation of cancer cells to the brain niche, and provide therapeutic alternatives modulating the brain metastatic niche.
Collapse
Affiliation(s)
| | - Diana M Cittelly
- Department of Pathology, University of Colorado Denver, Aurora, Colorado
| |
Collapse
|
7
|
Li L, Ni L, Heary RF, Elkabes S. Astroglial TLR9 antagonism promotes chemotaxis and alternative activation of macrophages via modulation of astrocyte-derived signals: implications for spinal cord injury. J Neuroinflammation 2020; 17:73. [PMID: 32098620 PMCID: PMC7041103 DOI: 10.1186/s12974-020-01748-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 02/13/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The recruitment of immune system cells into the central nervous system (CNS) has a profound effect on the outcomes of injury and disease. Glia-derived chemoattractants, including chemokines, play a pivotal role in this process. In addition, cytokines and chemokines influence the phenotype of infiltrating immune cells. Depending on the stimuli present in the local milieu, infiltrating macrophages acquire the classically activated M1 or alternatively activated M2 phenotypes. The polarization of macrophages into detrimental M1 versus beneficial M2 phenotypes significantly influences CNS pathophysiology. Earlier studies indicated that a toll-like receptor 9 (TLR9) antagonist modulates astrocyte-derived cytokine and chemokine release. However, it is not known whether these molecular changes affect astrocyte-induced chemotaxis and polarization of macrophages. The present studies were undertaken to address these issues. METHODS The chemotaxis and polarization of mouse peritoneal macrophages by spinal cord astrocytes were evaluated in a Transwell co-culture system. Arrays and ELISA were utilized to quantify chemokines in the conditioned medium (CM) of pure astrocyte cultures. Immunostaining for M1- and M2-specific markers characterized the macrophage phenotype. The percentage of M2 macrophages at the glial scar was determined by stereological approaches in mice sustaining a mid-thoracic spinal cord contusion injury (SCI) and intrathecally treated with oligodeoxynucleotide 2088 (ODN 2088), the TLR9 antagonist. Statistical analyses used two-tailed independent-sample t-test and one-way analysis of variance (ANOVA) followed by Tukey's post hoc test. A p value < 0.05 was considered to be statistically significant. RESULTS ODN 2088-treated astrocytes significantly increased the chemotaxis of peritoneal macrophages via release of chemokine (C-C motif) ligand 1 (CCL1). Vehicle-treated astrocytes polarized macrophages into the M2 phenotype and ODN 2088-treated astrocytes promoted further M2 polarization. Reduced CCL2 and CCL9 release by astrocytes in response to ODN 2088 facilitated the acquisition of the M2 phenotype, suggesting that CCL2 and CCL9 are negative regulators of M2 polarization. The percentage of M2 macrophages at the glial scar was higher in mice sustaining a SCI and receiving ODN 2088 treatment as compared to vehicle-treated injured controls. CONCLUSIONS TLR9 antagonism could create a favorable environment during SCI by supporting M2 macrophage polarization and chemotaxis via modulation of astrocyte-to-macrophage signals.
Collapse
Affiliation(s)
- Lun Li
- Reynolds Family Spine Laboratory, Department of Neurosurgery, New Jersey Medical School, Rutgers, The State University of New Jersey, 205 South Orange Avenue, F-1204, Newark, NJ 07103 USA
| | - Li Ni
- Reynolds Family Spine Laboratory, Department of Neurosurgery, New Jersey Medical School, Rutgers, The State University of New Jersey, 205 South Orange Avenue, F-1204, Newark, NJ 07103 USA
| | - Robert F. Heary
- Reynolds Family Spine Laboratory, Department of Neurosurgery, New Jersey Medical School, Rutgers, The State University of New Jersey, 205 South Orange Avenue, F-1204, Newark, NJ 07103 USA
| | - Stella Elkabes
- Reynolds Family Spine Laboratory, Department of Neurosurgery, New Jersey Medical School, Rutgers, The State University of New Jersey, 205 South Orange Avenue, F-1204, Newark, NJ 07103 USA
| |
Collapse
|
8
|
Astrocytes in multiple sclerosis and experimental autoimmune encephalomyelitis: Star-shaped cells illuminating the darkness of CNS autoimmunity. Brain Behav Immun 2019; 80:10-24. [PMID: 31125711 DOI: 10.1016/j.bbi.2019.05.029] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/16/2019] [Accepted: 05/20/2019] [Indexed: 12/18/2022] Open
Abstract
Neuropathology in the human autoimmune disease multiple sclerosis (MS) is considered to be mediated by autoreactive leukocytes, such as T cells, B cells, and macrophages. However, the inflammation and tissue damage in MS and its animal model experimental autoimmune encephalomyelitis (EAE) is also critically regulated by astrocytes, the most abundant cell population in the central nervous system (CNS). Under physiological conditions, astrocytes are integral to the development and function of the CNS, whereas in CNS autoimmunity, astrocytes influence the pathogenesis, progression, and recovery of the diseases. In this review, we summarize recent advances in astrocytic functions in the context of MS and EAE, which are categorized into two opposite aspects, one being detrimental and the other beneficial. Inhibition of the detrimental functions and/or enhancement of the beneficial functions of astrocytes might be favorable for the treatment of MS.
Collapse
|
9
|
Díaz LR, Saavedra-López E, Romarate L, Mitxitorena I, Casanova PV, Cribaro GP, Gallego JM, Pérez-Vallés A, Forteza-Vila J, Alfaro-Cervello C, García-Verdugo JM, Barcia C, Barcia C. Imbalance of immunological synapse-kinapse states reflects tumor escape to immunity in glioblastoma. JCI Insight 2018; 3:120757. [PMID: 30232280 DOI: 10.1172/jci.insight.120757] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 08/17/2018] [Indexed: 12/19/2022] Open
Abstract
Since the proper activation of T cells requires the physical interaction with target cells through the formation of immunological synapses (IS), an alteration at this level could be a reason why tumors escape the immune response. As part of their life cycle, it is thought that T cells alternate between a static phase, the IS, and a dynamic phase, the immunological kinapse (IK), depending on high or low antigen sensing. Our investigation performed in tissue samples of human glioma shows that T cells are able to establish synapsing interactions not only with glioma tumorigenic cells, but also with stromal myeloid cells. Particularly, the IS displaying a T cell receptor-rich (TCR-rich) central supramolecular activation cluster (cSMAC) is preferentially established with stromal cells, as opposed to malignant cells. Conversely, T cells in the malignant areas showed distinct morphometric parameters compared with nonneoplastic tissue - the former characterized by an elongated shape, well-suited to kinaptic dynamics. Importantly, high-resolution 3-dimensional analyses demonstrated the existence of bona-fide IK preferentially arranged in malignant areas of the tumor. This imbalance of IS/IK states between these 2 microenvironments reveals the low antigenic sensing of T cells when patrolling tumorigenic cells and reflects the immunoevasive environment of the tumor.
Collapse
Affiliation(s)
- Laura R Díaz
- Department of Biochemistry and Molecular Biology, School of Medicine, and.,Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Barcelona, Spain
| | - Elena Saavedra-López
- Department of Biochemistry and Molecular Biology, School of Medicine, and.,Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Barcelona, Spain
| | - Leire Romarate
- Department of Biochemistry and Molecular Biology, School of Medicine, and.,Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Barcelona, Spain
| | - Izaskun Mitxitorena
- Department of Biochemistry and Molecular Biology, School of Medicine, and.,Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Barcelona, Spain
| | - Paola V Casanova
- Department of Biochemistry and Molecular Biology, School of Medicine, and.,Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Barcelona, Spain
| | - George P Cribaro
- Department of Biochemistry and Molecular Biology, School of Medicine, and.,Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Barcelona, Spain
| | | | - Ana Pérez-Vallés
- Department of Pathology, Valencia General Hospital, Valencia, Spain
| | - Jerónimo Forteza-Vila
- Unidad Mixta CIPF/UCV de Investigación Oncológica, Instituto Valenciano de Patología, Universidad Católica de Valencia, Valencia, Spain
| | - Clara Alfaro-Cervello
- Laboratory of Comparative Neurobiology, Instituto Cavanilles, Universitat de València, CIBERNED, Valencia, Spain
| | - José M García-Verdugo
- Laboratory of Comparative Neurobiology, Instituto Cavanilles, Universitat de València, CIBERNED, Valencia, Spain
| | | | - Carlos Barcia
- Department of Biochemistry and Molecular Biology, School of Medicine, and.,Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Barcelona, Spain
| |
Collapse
|
10
|
Jiang X, Ganesan P, Rengarajan T, Choi DK, Arulselvan P. Cellular phenotypes as inflammatory mediators in Parkinson's disease: Interventional targets and role of natural products. Biomed Pharmacother 2018; 106:1052-1062. [PMID: 30119171 DOI: 10.1016/j.biopha.2018.06.162] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 06/26/2018] [Accepted: 06/27/2018] [Indexed: 12/15/2022] Open
Abstract
Pathogenesis of Parkinson's disease (PD) is undoubtedly a multifactorial phenomenon, with diverse etiological agents. Pro-inflammatory mediators act as a skew that directs disease progression during neurodegenerative diseases. Understanding the dynamics of inflammation and inflammatory mediators in preventing or reducing disease progression has recently gained much attention. Inflammatory neuro-degeneration is regulated via cytokines, chemokines, lipid mediators and immune cell subsets; however, individual cellular phenotypes in the Central Nervous System (CNS) acts in diverse ways whose persistent activation leads to unresolving inflammation often causing unfavorable outcomes in neurodegenerative disease like PD. Specifically, activation of cellular phenotypes like astrocytes, microglia, activation of peripheral immune cells requires different activation signals and agents like (cytokines, misfolded protein aggregates, infectious agents, pesticides like organophosphates, etc.,). However, what is unknown is how the different cellular phenotypes respond uniquely and the role of the factors they secrete alters the signal cascades in the complex neuron-microglial connections in the CNS. Hence, understanding the role of cellular phenotypes and the inflammatory mediators, the cross talk among the signals and their receptors can help us to identify the potential therapeutic target using natural products. In this review we have tried to put together the role of cellular phenotypes as a skew that favors PD progression and we have also discussed how the lack of experimental approaches and challenges that affects understanding the cellular targets that can be used against natural derivatives in alleviating PD pathophysiology. Together, this review will provide the better insights into the role of cellular phenotypes of neuroinflammation, inflammatory mediators and the orchestrating factors of inflammation and how they can be targeted in a more specific way that can be used in the clinical management of PD.
Collapse
Affiliation(s)
- Xu Jiang
- Department of Neurology, Shenzhen Shajing Affiliated Hospital of Guangzhou Medical University, 3 Shajing St, Baoan Qu, Shenzhen Shi, Guangdong Sheng, 518104, China.
| | - Palanivel Ganesan
- Nanotechnology Research Center and Department of Applied Life Science, College of Biomedical and Health Science, Konkuk University, Chungju, 380-701, Republic of Korea; Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju, 380-701, Republic of Korea.
| | - Thamaraiselvan Rengarajan
- Scigen Research and Innovation Pvt. Ltd., Periyar Technology Business Incubator, Periyar Nagar, Thanjavur, 613403, India.
| | - Dong-Kug Choi
- Nanotechnology Research Center and Department of Applied Life Science, College of Biomedical and Health Science, Konkuk University, Chungju, 380-701, Republic of Korea; Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju, 380-701, Republic of Korea.
| | - Palanisamy Arulselvan
- Scigen Research and Innovation Pvt. Ltd., Periyar Technology Business Incubator, Periyar Nagar, Thanjavur, 613403, India; Muthayammal Centre for Advanced Research, Muthayammal College of Arts and Science, Rasipuram, Namakkal, Tamilnadu, 637408, India.
| |
Collapse
|
11
|
Bozic I, Tesovic K, Laketa D, Adzic M, Jakovljevic M, Bjelobaba I, Savic D, Nedeljkovic N, Pekovic S, Lavrnja I. Voltage Gated Potassium Channel Kv1.3 Is Upregulated on Activated Astrocytes in Experimental Autoimmune Encephalomyelitis. Neurochem Res 2018; 43:1020-1034. [PMID: 29574670 DOI: 10.1007/s11064-018-2509-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/13/2018] [Accepted: 03/17/2018] [Indexed: 12/13/2022]
Abstract
Kv1.3 is a voltage gated potassium channel that has been implicated in pathophysiology of multiple sclerosis (MS). In the present study we investigated temporal and cellular expression pattern of this channel in the lumbar part of spinal cords of animals with experimental autoimmune encephalomyelitis (EAE), animal model of MS. EAE was actively induced in female Dark Agouti rats. Expression of Kv1.3 was analyzed at different time points of disease progression, at the onset, peak and end of EAE. We here show that Kv1.3 increased by several folds at the peak of EAE at both gene and protein level. Double immunofluorescence analyses demonstrated localization of Kv1.3 on activated microglia, macrophages, and reactive astrocytes around inflammatory lesions. In vitro experiments showed that pharmacological block of Kv1.3 in activated astrocytes suppresses the expression of proinflammatory mediators, suggesting a role of this channel in inflammation. Our results support the hypothesis that Kv1.3 may be a therapeutic target of interest for MS and add astrocytes to the list of cells whose activation would be suppressed by inhibiting Kv1.3 in inflammatory conditions.
Collapse
Affiliation(s)
- Iva Bozic
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Blvd Despota Stefana 142, 11060, Belgrade, Serbia.
| | - Katarina Tesovic
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Blvd Despota Stefana 142, 11060, Belgrade, Serbia
| | - Danijela Laketa
- Institute of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Marija Adzic
- Institute of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Marija Jakovljevic
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Blvd Despota Stefana 142, 11060, Belgrade, Serbia
| | - Ivana Bjelobaba
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Blvd Despota Stefana 142, 11060, Belgrade, Serbia
| | - Danijela Savic
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Blvd Despota Stefana 142, 11060, Belgrade, Serbia
| | - Nadezda Nedeljkovic
- Institute of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Sanja Pekovic
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Blvd Despota Stefana 142, 11060, Belgrade, Serbia
| | - Irena Lavrnja
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Blvd Despota Stefana 142, 11060, Belgrade, Serbia
| |
Collapse
|
12
|
Zhang G, Wang J, Yao G, Shi B. Downregulation of CCL2 induced by the upregulation of microRNA-206 is associated with the severity of HEV71 encephalitis. Mol Med Rep 2017; 16:4620-4626. [PMID: 28765968 PMCID: PMC5647018 DOI: 10.3892/mmr.2017.7142] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 02/20/2017] [Indexed: 12/25/2022] Open
Abstract
MicroRNAs (miRNAs) have been investigated widely as key regulators of gene expression in different diseases by affecting the miRNA‑mediated regulatory function. Human enterovirus 71 (HEV71) can cause a series of human diseases, including encephalitis. Chemokine (C‑C motif) ligand 2 (CCL2) is one of the important genes involved in regulating inflammation. However, the mechanisms underlying HEV71 encephalitis mediated by CCL2 remain to be elucidated. In the present study, reverse transcription‑quantitative polymerase chain reaction analysis was used to determine the expression level of miR‑206 and the mRNA expression of CCL2 in samples. Western blot analysis was used to detect the protein levels of CCL2. A luciferase assay was used to verify the miR‑206 target site in CCL2. A CCK‑8 assay and flow cytometry were used to determine cell proliferation and apoptosis. The results demonstrated that miR‑206 was downregulated in severe HEV71 encephalitis. Using bioinformatics analysis, miR‑206 was predicted to target the human CCL2 3'‑untranslated region (3'‑UTR). A dual‑luciferase assay demonstrated that miR‑206 downregulated the expression of CCL2 by directly targeting its 3'‑UTR, whereas CCL2 3'‑UTR mutations completely eliminated its interaction with miR‑206. The expression levels of miR‑206 and CCL2 were inversely correlated in cerebrospinal fluid. The expression of exogenous miRNA, which mimicked miR‑206 miRNA, decreased the protein and mRNA levels of CCL2, whereas the suppression of endogenous miR‑206 resulted in an increase of the protein and mRNA levels of CCL2. The present study also found that miR‑206 promoted NPC cell proliferation and reduced the apoptosis of NPC cells via CCL2. The mechanism is likely to involve suppression of the expression of miR‑206 and upregulation of the expression of CCL2, important in regulating the progress of HEV71 encephalitis. In conclusion, miR‑206 may be useful in the prognosis and treatment of HEV71 encephalitis.
Collapse
Affiliation(s)
- Guangyou Zhang
- Department of Pediatrics, Taian Central Hospital, Taian, Shandong 271000, P.R. China
| | - Jiwen Wang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Guo Yao
- Department of Pediatrics, Taian Central Hospital, Taian, Shandong 271000, P.R. China
| | - Baohai Shi
- Department of Pediatrics, Taian Central Hospital, Taian, Shandong 271000, P.R. China
| |
Collapse
|
13
|
Spielman LJ, Estaki M, Ghosh S, Gibson DL, Klegeris A. The effects of voluntary wheel running on neuroinflammatory status: Role of monocyte chemoattractant protein-1. Mol Cell Neurosci 2017; 79:93-102. [DOI: 10.1016/j.mcn.2016.12.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 12/30/2016] [Indexed: 12/17/2022] Open
|
14
|
Filipello F, Pozzi D, Proietti M, Romagnani A, Mazzitelli S, Matteoli M, Verderio C, Grassi F. Ectonucleotidase activity and immunosuppression in astrocyte-CD4 T cell bidirectional signaling. Oncotarget 2017; 7:5143-56. [PMID: 26784253 PMCID: PMC4868677 DOI: 10.18632/oncotarget.6914] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 01/01/2016] [Indexed: 12/04/2022] Open
Abstract
Astrocytes play a crucial role in neuroinflammation as part of the glia limitans, which regulates infiltration of the brain parenchyma by leukocytes. The signaling pathways and molecular events, which result from the interaction of activated T cells with astrocytes are poorly defined. Here we show that astrocytes promote the expression and enzymatic activity of CD39 and CD73 ectonucleotidases in recently activated CD4 cells by a contact dependent mechanism that is independent of T cell receptor interaction with class II major histocompatibility complex (MHC). Transforming growth factor-β (TGF-β) is robustly upregulated and sufficient to promote ectonucleotidases expression. T cell adhesion to astrocyte results in differentiation to an immunosuppressive phenotype defined by expression of the transcription factor Rorγt, which characterizes the CD4 T helper 17 subset. CD39 activity in T cells in turn inhibits spontaneous calcium oscillations in astrocytes that correlated with enhanced and reduced transcription of CCL2 chemokine and Sonic hedgehog (Shh), respectively. We hypothesize this TCR-independent interaction promote an immunosuppressive program in T cells to control possible brain injury by deregulated T cell activation during neuroinflammation. On the other hand, the increased secretion of CCL2 with concomitant reduction of Shh might promote leukocytes extravasation into the brain parenchyma.
Collapse
Affiliation(s)
- Fabia Filipello
- Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center, Rozzano, Italy
| | - Davide Pozzi
- Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center, Rozzano, Italy
| | - Michele Proietti
- Institute for Research in Biomedicine, Bellinzona, Switzerland.,Center of Chronic Immunodeficiency, University Medical Center, Freiburg, Germany
| | - Andrea Romagnani
- Institute for Research in Biomedicine, Bellinzona, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Sonia Mazzitelli
- Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center, Rozzano, Italy.,Hertie Institute for Clinical Brain Research, University of Tubingen, Department of Cellular Neurology, Tubingen, Germany
| | - Michela Matteoli
- Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center, Rozzano, Italy.,CNR Institute of Neuroscience, Milano, Italy
| | - Claudia Verderio
- Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center, Rozzano, Italy.,CNR Institute of Neuroscience, Milano, Italy
| | - Fabio Grassi
- Institute for Research in Biomedicine, Bellinzona, Switzerland.,Department of Medical Biotechnology and Translational Medicine, University of Milan, Istituto Nazionale di Genetica Molecolare, Milan, Italy
| |
Collapse
|
15
|
Le Thuc O, Cansell C, Bourourou M, Denis RG, Stobbe K, Devaux N, Guyon A, Cazareth J, Heurteaux C, Rostène W, Luquet S, Blondeau N, Nahon JL, Rovère C. Central CCL2 signaling onto MCH neurons mediates metabolic and behavioral adaptation to inflammation. EMBO Rep 2016; 17:1738-1752. [PMID: 27733491 DOI: 10.15252/embr.201541499] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 08/25/2016] [Accepted: 08/30/2016] [Indexed: 12/30/2022] Open
Abstract
Sickness behavior defines the endocrine, autonomic, behavioral, and metabolic responses associated with infection. While inflammatory responses were suggested to be instrumental in the loss of appetite and body weight, the molecular underpinning remains unknown. Here, we show that systemic or central lipopolysaccharide (LPS) injection results in specific hypothalamic changes characterized by a precocious increase in the chemokine ligand 2 (CCL2) followed by an increase in pro-inflammatory cytokines and a decrease in the orexigenic neuropeptide melanin-concentrating hormone (MCH). We therefore hypothesized that CCL2 could be the central relay for the loss in body weight induced by the inflammatory signal LPS. We find that central delivery of CCL2 promotes neuroinflammation and the decrease in MCH and body weight. MCH neurons express CCL2 receptor and respond to CCL2 by decreasing both electrical activity and MCH release. Pharmacological or genetic inhibition of CCL2 signaling opposes the response to LPS at both molecular and physiologic levels. We conclude that CCL2 signaling onto MCH neurons represents a core mechanism that relays peripheral inflammation to sickness behavior.
Collapse
Affiliation(s)
- Ophélia Le Thuc
- Université Côte d'Azur, Nice, France.,CNRS, IPMC, Sophia Antipolis, France
| | - Céline Cansell
- Université Côte d'Azur, Nice, France.,CNRS, IPMC, Sophia Antipolis, France
| | - Miled Bourourou
- Université Côte d'Azur, Nice, France.,CNRS, IPMC, Sophia Antipolis, France
| | - Raphaël Gp Denis
- Univ Paris Diderot Sorbonne Paris Cité Unité de Biologie Fonctionnelle et Adaptative CNRS UMR 8251, Paris, France
| | - Katharina Stobbe
- Université Côte d'Azur, Nice, France.,CNRS, IPMC, Sophia Antipolis, France
| | - Nadège Devaux
- Université Côte d'Azur, Nice, France.,CNRS, IPMC, Sophia Antipolis, France
| | - Alice Guyon
- Université Côte d'Azur, Nice, France.,CNRS, IPMC, Sophia Antipolis, France
| | - Julie Cazareth
- Université Côte d'Azur, Nice, France.,CNRS, IPMC, Sophia Antipolis, France
| | | | - William Rostène
- Institut de la Vision UMRS 968-Université Pierre et Marie Curie, Paris, France
| | - Serge Luquet
- Univ Paris Diderot Sorbonne Paris Cité Unité de Biologie Fonctionnelle et Adaptative CNRS UMR 8251, Paris, France
| | - Nicolas Blondeau
- Université Côte d'Azur, Nice, France.,CNRS, IPMC, Sophia Antipolis, France
| | - Jean-Louis Nahon
- Université Côte d'Azur, Nice, France .,CNRS, IPMC, Sophia Antipolis, France.,Station de Primatologie UPS846 CNRS, Rousset-sur-Arc, France
| | - Carole Rovère
- Université Côte d'Azur, Nice, France .,CNRS, IPMC, Sophia Antipolis, France
| |
Collapse
|
16
|
Du C, Pan P, Jiang Y, Zhang Q, Bao J, Liu C. Microarray data analysis to identify crucial genes regulated by CEBPB in human SNB19 glioma cells. World J Surg Oncol 2016; 14:258. [PMID: 27716259 PMCID: PMC5054626 DOI: 10.1186/s12957-016-0997-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 08/30/2016] [Indexed: 01/18/2023] Open
Abstract
Background Glioma is one of the most common primary malignancies in the brain or spine. The transcription factor (TF) CCAAT/enhancer binding protein beta (CEBPB) is important for maintaining the tumor initiating capacity and invasion ability. To investigate the regulation mechanism of CEBPB in glioma, microarray data GSE47352 was analyzed. Methods GSE47352 was downloaded from Gene Expression Omnibus, including three samples of SNB19 human glioma cells transduced with non-target control small hairpin RNA (shRNA) lentiviral vectors for 72 h (normal glioma cells) and three samples of SNB19 human glioma cells transduced with CEBPB shRNA lentiviral vectors for 72 h (CEBPB-silenced glioma cells). The differentially expressed genes (DEGs) were screened using limma package and then annotated. Afterwards, the Database for Annotation, Visualization, and Integrated Discovery (DAVID) software was applied to perform enrichment analysis for the DEGs. Furthermore, the protein-protein interaction (PPI) network and transcriptional regulatory network were constructed using Cytoscape software. Results Total 529 DEGs were identified in the normal glioma cells compared with the CEBPB-silenced glioma cells, including 336 up-regulated and 193 down-regulated genes. The significantly enriched pathways included chemokine signaling pathway (which involved CCL2), focal adhesion (which involved THBS1 and THBS2), TGF-beta signaling pathway (which involved THBS1, THBS2, SMAD5, and SMAD6) and chronic myeloid leukemia (which involved TGFBR2 and CCND1). In the PPI network, CCND1 (degree = 29) and CCL2 (degree = 12) were hub nodes. Additionally, CEBPB and TCF12 might function in glioma through targeting others (CEBPB → TCF12, CEBPB → TGFBR2, and TCF12 → TGFBR2). Conclusions CEBPB might act in glioma by regulating CCL2, CCND1, THBS1, THBS2, SMAD5, SMAD6, TGFBR2, and TCF12.
Collapse
Affiliation(s)
- Chenghua Du
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia University for the Nationalities, Huolinhe Street No.1742, Tongliao, Inner Mongolia, 028007, China.
| | - Pan Pan
- Department of Hepatology, Tongliao City Hospital for Infectious Diseases, Tongliao, Inner Mongolia, 028007, China
| | - Yan Jiang
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia University for the Nationalities, Huolinhe Street No.1742, Tongliao, Inner Mongolia, 028007, China
| | - Qiuli Zhang
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia University for the Nationalities, Huolinhe Street No.1742, Tongliao, Inner Mongolia, 028007, China
| | - Jinsuo Bao
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia University for the Nationalities, Huolinhe Street No.1742, Tongliao, Inner Mongolia, 028007, China
| | - Chang Liu
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia University for the Nationalities, Huolinhe Street No.1742, Tongliao, Inner Mongolia, 028007, China
| |
Collapse
|
17
|
Patel C, Xu Z, Shosha E, Xing J, Lucas R, Caldwell RW, Caldwell RB, Narayanan SP. Treatment with polyamine oxidase inhibitor reduces microglial activation and limits vascular injury in ischemic retinopathy. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1862:1628-39. [PMID: 27239699 PMCID: PMC5091072 DOI: 10.1016/j.bbadis.2016.05.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 04/26/2016] [Accepted: 05/26/2016] [Indexed: 12/15/2022]
Abstract
Retinal vascular injury is a major cause of vision impairment in ischemic retinopathies. Insults such as hyperoxia, oxidative stress and inflammation contribute to this pathology. Previously, we showed that hyperoxia-induced retinal neurodegeneration is associated with increased polyamine oxidation. Here, we are studying the involvement of polyamine oxidases in hyperoxia-induced injury and death of retinal vascular endothelial cells. New-born C57BL6/J mice were exposed to hyperoxia (70% O2) from postnatal day (P) 7 to 12 and were treated with the polyamine oxidase inhibitor MDL 72527 or vehicle starting at P6. Mice were sacrificed after different durations of hyperoxia and their retinas were analyzed to determine the effects on vascular injury, microglial cell activation, and inflammatory cytokine profiling. The results of this analysis showed that MDL 72527 treatment significantly reduced hyperoxia-induced retinal vascular injury and enhanced vascular sprouting as compared with the vehicle controls. These protective effects were correlated with significant decreases in microglial activation as well as levels of inflammatory cytokines and chemokines. In order to model the effects of polyamine oxidation in causing microglial activation in vitro, studies were performed using rat brain microvascular endothelial cells treated with conditioned-medium from rat retinal microglia stimulated with hydrogen peroxide. Conditioned-medium from activated microglial cultures induced cell stress signals and cell death in microvascular endothelial cells. These studies demonstrate the involvement of polyamine oxidases in hyperoxia-induced retinal vascular injury and retinal inflammation in ischemic retinopathy, through mechanisms involving cross-talk between endothelial cells and resident retinal microglia.
Collapse
Affiliation(s)
- C Patel
- Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA; Vascular Biology Center, Augusta University, Augusta, GA 30912, USA; Department of Occupational Therapy, College of Allied Health Sciences, Augusta University, Augusta, GA 30912, USA.
| | - Z Xu
- Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA; Vascular Biology Center, Augusta University, Augusta, GA 30912, USA.
| | - E Shosha
- Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA; Vascular Biology Center, Augusta University, Augusta, GA 30912, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA.
| | - J Xing
- Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA; Vascular Biology Center, Augusta University, Augusta, GA 30912, USA.
| | - R Lucas
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA; Department of Pharmacology & Toxicology, Augusta University, Augusta, GA 30912, USA.
| | - R W Caldwell
- Department of Pharmacology & Toxicology, Augusta University, Augusta, GA 30912, USA.
| | - R B Caldwell
- Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA; Vascular Biology Center, Augusta University, Augusta, GA 30912, USA; Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA 30912, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA.
| | - S P Narayanan
- Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA; Vascular Biology Center, Augusta University, Augusta, GA 30912, USA; Department of Occupational Therapy, College of Allied Health Sciences, Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
18
|
Chang AL, Miska J, Wainwright DA, Dey M, Rivetta CV, Yu D, Kanojia D, Pituch KC, Qiao J, Pytel P, Han Y, Wu M, Zhang L, Horbinski CM, Ahmed AU, Lesniak MS. CCL2 Produced by the Glioma Microenvironment Is Essential for the Recruitment of Regulatory T Cells and Myeloid-Derived Suppressor Cells. Cancer Res 2016; 76:5671-5682. [PMID: 27530322 DOI: 10.1158/0008-5472.can-16-0144] [Citation(s) in RCA: 461] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 07/07/2016] [Indexed: 12/15/2022]
Abstract
In many aggressive cancers, such as glioblastoma multiforme, progression is enabled by local immunosuppression driven by the accumulation of regulatory T cells (Treg) and myeloid-derived suppressor cells (MDSC). However, the mechanistic details of how Tregs and MDSCs are recruited in various tumors are not yet well understood. Here we report that macrophages and microglia within the glioma microenvironment produce CCL2, a chemokine that is critical for recruiting both CCR4+ Treg and CCR2+Ly-6C+ monocytic MDSCs in this disease setting. In murine gliomas, we established novel roles for tumor-derived CCL20 and osteoprotegerin in inducing CCL2 production from macrophages and microglia. Tumors grown in CCL2-deficient mice failed to maximally accrue Tregs and monocytic MDSCs. In mixed-bone marrow chimera assays, we found that CCR4-deficient Treg and CCR2-deficient monocytic MDSCs were defective in glioma accumulation. Furthermore, administration of a small-molecule antagonist of CCR4 improved median survival in the model. In clinical specimens of glioblastoma multiforme, elevated levels of CCL2 expression correlated with reduced overall survival of patients. Finally, we found that CD163-positive infiltrating macrophages were a major source of CCL2 in glioblastoma multiforme patients. Collectively, our findings show how glioma cells influence the tumor microenvironment to recruit potent effectors of immunosuppression that drive progression. Cancer Res; 76(19); 5671-82. ©2016 AACR.
Collapse
Affiliation(s)
- Alan L Chang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois. Committee on Cancer Biology, The University of Chicago, Chicago, Illinois
| | - Jason Miska
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Derek A Wainwright
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Mahua Dey
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, Indiana. Section of Neurosurgery, Department of Surgery, The University of Chicago Hospitals, Chicago, Illinois
| | - Claudia V Rivetta
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Dou Yu
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Deepak Kanojia
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Katarzyna C Pituch
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Jian Qiao
- Section of Neurosurgery, Department of Surgery, The University of Chicago Hospitals, Chicago, Illinois. Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Peter Pytel
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | - Yu Han
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Meijing Wu
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Lingjiao Zhang
- Section of Neurosurgery, Department of Surgery, The University of Chicago Hospitals, Chicago, Illinois
| | - Craig M Horbinski
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois. Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Atique U Ahmed
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Maciej S Lesniak
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.
| |
Collapse
|
19
|
Ni J, Xie S, Ramkissoon SH, Luu V, Sun Y, Bandopadhayay P, Beroukhim R, Roberts TM, Stiles CD, Segal RA, Ligon KL, Hahn WC, Zhao JJ. Tyrosine receptor kinase B is a drug target in astrocytomas. Neuro Oncol 2016; 19:22-30. [PMID: 27402815 DOI: 10.1093/neuonc/now139] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Astrocytomas are the most common primary human brain tumors. Receptor tyrosine kinases (RTKs), including tyrosine receptor kinase B (TrkB, also known as tropomyosin-related kinase B; encoded by neurotrophic tyrosine kinase receptor type 2 [NTRK2]), are frequently mutated by rearrangement/fusion in high-grade and low-grade astrocytomas. We found that activated TrkB can contribute to the development of astrocytoma and might serve as a therapeutic target in this tumor type. METHODS To identify RTKs capable of inducing astrocytoma formation, a library of human tyrosine kinases was screened for the ability to transform murine Ink4a-/-/Arf-/- astrocytes. Orthotopic allograft studies were conducted to evaluate the effects of RTKs on the development of astrocytoma. Since TrkB was identified as a driver of astrocytoma formation, the effect of the Trk inhibitors AZD1480 and RXDX-101 was assessed in astrocytoma cells expressing activated TrkB. RNA sequencing, real-time PCR, western blotting, and enzyme-linked immunosorbent assays were conducted to characterize NTRK2 in astrocytomas. RESULTS Activated TrkB cooperated with Ink4a/Arf loss to induce the formation of astrocytomas through a mechanism mediated by activation of signal transducer and activator of transcription 3 (STAT3). TrkB activation positively correlated with Ccl2 expression. TrkB-induced astrocytomas remained dependent on TrkB signaling for survival, highlighting a role of NTRK2 as an addictive oncogene. Furthermore, the QKI-NTRK2 fusion associated with human astrocytoma transformed Ink4a-/-/Arf-/- astrocytes, and this process was also mediated via STAT3 signaling. CONCLUSIONS Our findings provide evidence that constitutively activated NTRK2 alleles, notably the human tumor-associated QKI-NTRK2 fusion, can cooperate with Ink4a/Arf loss to drive astrocytoma formation. Therefore, we propose NTRK2 as a potential therapeutic target in the subset of astrocytoma patients defined by QKI-NTRK2 fusion.
Collapse
Affiliation(s)
- Jing Ni
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| | - Shaozhen Xie
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| | - Shakti H Ramkissoon
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| | - Victor Luu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| | - Yu Sun
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| | - Pratiti Bandopadhayay
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| | - Rameen Beroukhim
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| | - Thomas M Roberts
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| | - Charles D Stiles
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| | - Rosalind A Segal
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| | - Keith L Ligon
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| | - William C Hahn
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| | - Jean J Zhao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| |
Collapse
|
20
|
CCL2 mediates the circadian response to low dose endotoxin. Neuropharmacology 2016; 108:373-81. [PMID: 27178133 DOI: 10.1016/j.neuropharm.2016.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 05/07/2016] [Accepted: 05/09/2016] [Indexed: 11/23/2022]
Abstract
The mammalian circadian system is mainly originated in a master oscillator located in the suprachiasmatic nuclei (SCN) in the hypothalamus. Previous reports from our and other groups have shown that the SCN are sensitive to systemic immune activation during the early night, through a mechanism that relies on the action of proinflammatory factors within this structure. Chemokine (C-C motif) ligand 2 (CCL2) is induced in the brain upon peripheral immune activation, and it has been shown to modulate neuronal physiology. In the present work we tested whether CCL2 might be involved in the response of the circadian clock to peripheral endotoxin administration. The CCL2 receptor, C-C chemokine receptor type 2 (CCR2), was detected in the SCN of mice, with higher levels of expression during the early night, when the clock is sensitive to immune activation. Ccl2 was induced in the SCN upon intraperitoneal lipopolysaccharide (LPS) administration. Furthermore, mice receiving an intracerebroventricular (Icv) administration of a CCL2 synthesis inhibitor (Bindarit), showed a reduction LPS-induced circadian phase changes and Icv delivery of CCL2 led to phase delays in the circadian clock. In addition, we tested the possibility that CCL2 might also be involved in the photic regulation of the clock. Icv administration of Bindarit did not modify the effects of light pulses on the circadian clock. In summary, we found that CCL2, acting at the SCN level is important for the circadian effects of immune activation.
Collapse
|
21
|
Interaction of astrocytes and T cells in physiological and pathological conditions. Brain Res 2015; 1623:63-73. [PMID: 25813828 DOI: 10.1016/j.brainres.2015.03.026] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 03/10/2015] [Accepted: 03/12/2015] [Indexed: 12/24/2022]
Abstract
The central nervous system (CNS) has long been recognized as a site of 'immune privilege' because of the existence of the blood brain barrier (BBB) which presumably isolates CNS from the peripheral immunosurveillance. Different from the peripheral organs, CNS is unique in response to all forms of CNS injury and disease which is mainly mediated by resident microglia and astrocyte. There is increasing evidence that immune cells are not only involved in neuroinflammation process but also the maintenance of CNS homeostasis. T cells, an important immune cell population, are involved in the pathogenesis of some neurological diseases by inducing either innate or adaptive immune responses. Astrocytes, which are the most abundant cell type in the CNS, maintain the integrity of BBB and actively participate in the initiation and progression of neurological diseases. Surprisingly, how astrocytes and T cells interact and the consequences of their interaction are not clear. In this review we briefly summarized T cells diversity and astrocyte function. Then, we examined the evidence for the astrocytes and T cells interaction under physiological and pathological conditions including ischemic stroke, multiple sclerosis, viral infection, and Alzheimer's disease. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
|
22
|
Conditional ablation of astroglial CCL2 suppresses CNS accumulation of M1 macrophages and preserves axons in mice with MOG peptide EAE. J Neurosci 2014; 34:8175-85. [PMID: 24920622 DOI: 10.1523/jneurosci.1137-14.2014] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Current multiple sclerosis (MS) therapies only partially prevent chronically worsening neurological deficits, which are largely attributable to progressive loss of CNS axons. Prior studies of experimental autoimmune encephalomyelitis (EAE) induced in C57BL/6 mice by immunization with myelin oligodendrocyte glycoprotein peptide 35-55 (MOG peptide), a model of MS, documented continued axon loss for months after acute CNS inflammatory infiltrates had subsided, and massive astroglial induction of CCL2 (MCP-1), a chemokine for CCR2(+) monocytes. We now report that conditional deletion of astroglial CCL2 significantly decreases CNS accumulation of classically activated (M1) monocyte-derived macrophages and microglial expression of M1 markers during the initial CNS inflammatory phase of MOG peptide EAE, reduces the acute and long-term severity of clinical deficits and slows the progression of spinal cord axon loss. In addition, lack of astroglial-derived CCL2 results in increased accumulation of Th17 cells within the CNS in these mice, but also in greater confinement of CD4(+) lymphocytes to CNS perivascular spaces. These findings suggest that therapies designed to inhibit astroglial CCL2-driven trafficking of monocyte-derived macrophages to the CNS during acute MS exacerbations have the potential to significantly reduce CNS axon loss and slow progression of neurological deficits.
Collapse
|
23
|
Kim RY, Hoffman AS, Itoh N, Ao Y, Spence R, Sofroniew MV, Voskuhl RR. Astrocyte CCL2 sustains immune cell infiltration in chronic experimental autoimmune encephalomyelitis. J Neuroimmunol 2014; 274:53-61. [PMID: 25005117 DOI: 10.1016/j.jneuroim.2014.06.009] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 06/09/2014] [Accepted: 06/17/2014] [Indexed: 02/09/2023]
Abstract
Chemokine (C-C motif) ligand 2 (CCL2), initially identified as monocyte chemoattractant protein-1 (MCP-1), recruits immune cells to the central nervous system (CNS) during autoimmune inflammation. CCL2 can be expressed by multiple cell types, but which cells are responsible for CCL2 function during acute and chronic phases of autoimmune disease is not known. We determined the role of CCL2 in astrocytes in vivo during experimental autoimmune encephalomyelitis (EAE) by using Cre-loxP gene deletion. Mice with a conditional gene deletion of CCL2 from astrocytes had less severe EAE late in disease while having a similar incidence and severity of disease at onset as compared to wild type (WT) control littermates. EAE mice devoid of CCL2 in astrocytes had less macrophage and T cell inflammation in the white matter of the spinal cord and less diffuse activation of astrocytes and microglia in both white and gray matter as well as less axonal loss and demyelination, compared to WT littermates. These findings demonstrate that CCL2 in astrocytes plays an important role in the continued recruitment of immune cells and activation of glial cells in the CNS during chronic EAE, thereby suggesting a novel cell specific target for neuroprotective treatments of chronic neuroinflammatory diseases.
Collapse
Affiliation(s)
- Roy Y Kim
- Molecular Cellular and Integrative Physiology Interdepartmental Ph.D. Program, University of California, Los Angeles; Multiple Sclerosis Program, Department of Neurology, University of California, Los Angeles
| | - Alexandria S Hoffman
- Multiple Sclerosis Program, Department of Neurology, University of California, Los Angeles
| | - Noriko Itoh
- Multiple Sclerosis Program, Department of Neurology, University of California, Los Angeles
| | - Yan Ao
- Department of Neurobiology, University of California, Los Angeles
| | - Rory Spence
- Multiple Sclerosis Program, Department of Neurology, University of California, Los Angeles
| | | | - Rhonda R Voskuhl
- Multiple Sclerosis Program, Department of Neurology, University of California, Los Angeles.
| |
Collapse
|
24
|
Neuroantibody biomarkers: links and challenges in environmental neurodegeneration and autoimmunity. Autoimmune Dis 2014; 2014:340875. [PMID: 25045531 PMCID: PMC4090524 DOI: 10.1155/2014/340875] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 05/18/2014] [Accepted: 05/19/2014] [Indexed: 11/18/2022] Open
Abstract
The majority of neurodegenerative (ND) and autoimmune diseases (AID) remain idiopathic. The contribution of environmental chemicals to the development of these disorders has become of great interest in recent years. A convergence of mechanism between of ND and AID development has also emerged. In the case of ND, including neurotoxicity, the focus of this review, work over the last two decade in the realm of biomarker development, indicates that the immune response provides a venue whereby humoral immunity, in the form of autoantibodies to nervous system specific proteins, or neuroantibodies (NAb), may provide, once validated, a sensitive high throughput surrogate biomarker of effect with the potential of predicting outcome in absence of overt neurotoxicity/neurodegeneration. In addition, NAb may prove to be a contributor to the progression of the nervous system pathology, as well as biomarker of stage and therapeutic efficacy. There is a compelling need for biomarkers of effect in light of the introduction of new chemicals, such as nanoengineered material, where potential neurotoxicity remains to be defined. Furthermore, the convergence of mechanisms associated with ND and AID draws attention to the neglected arena of angiogenesis in defining the link between environment, ND, and AID.
Collapse
|
25
|
Jansson D, Rustenhoven J, Feng S, Hurley D, Oldfield RL, Bergin PS, Mee EW, Faull RLM, Dragunow M. A role for human brain pericytes in neuroinflammation. J Neuroinflammation 2014; 11:104. [PMID: 24920309 PMCID: PMC4105169 DOI: 10.1186/1742-2094-11-104] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 05/19/2014] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Brain inflammation plays a key role in neurological disease. Although much research has been conducted investigating inflammatory events in animal models, potential differences in human brain versus rodent models makes it imperative that we also study these phenomena in human cells and tissue. METHODS Primary human brain cell cultures were generated from biopsy tissue of patients undergoing surgery for drug-resistant epilepsy. Cells were treated with pro-inflammatory compounds IFNγ, TNFα, IL-1β, and LPS, and chemokines IP-10 and MCP-1 were measured by immunocytochemistry, western blot, and qRT-PCR. Microarray analysis was also performed on late passage cultures treated with vehicle or IFNγ and IL-1β. RESULTS Early passage human brain cell cultures were a mixture of microglia, astrocytes, fibroblasts and pericytes. Later passage cultures contained proliferating fibroblasts and pericytes only. Under basal culture conditions all cell types showed cytoplasmic NFκB indicating that they were in a non-activated state. Expression of IP-10 and MCP-1 were significantly increased in response to pro-inflammatory stimuli. The two chemokines were expressed in mixed cultures as well as cultures of fibroblasts and pericytes only. The expression of IP-10 and MCP-1 were regulated at the mRNA and protein level, and both were secreted into cell culture media. NFκB nuclear translocation was also detected in response to pro-inflammatory cues (except IFNγ) in all cell types. Microarray analysis of brain pericytes also revealed widespread changes in gene expression in response to the combination of IFNγ and IL-1β treatment including interleukins, chemokines, cellular adhesion molecules and much more. CONCLUSIONS Adult human brain cells are sensitive to cytokine challenge. As expected 'classical' brain immune cells, such as microglia and astrocytes, responded to cytokine challenge but of even more interest, brain pericytes also responded to such challenge with a rich repertoire of gene expression. Immune activation of brain pericytes may play an important role in communicating inflammatory signals to and within the brain interior and may also be involved in blood brain barrier (BBB) disruption . Targeting brain pericytes, as well as microglia and astrocytes, may provide novel opportunities for reducing brain inflammation and maintaining BBB function and brain homeostasis in human brain disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Mike Dragunow
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 85 Park Road, Auckland 1023, New Zealand.
| |
Collapse
|
26
|
Guo MF, Meng J, Li YH, Yu JZ, Liu CY, Feng L, Yang WF, Li JL, Feng QJ, Xiao BG, Ma CG. The inhibition of Rho kinase blocks cell migration and accumulation possibly by challenging inflammatory cytokines and chemokines on astrocytes. J Neurol Sci 2014; 343:69-75. [PMID: 24952673 DOI: 10.1016/j.jns.2014.05.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 04/17/2014] [Accepted: 05/14/2014] [Indexed: 12/21/2022]
Abstract
Multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE), are autoimmune diseases characterized by the immune-mediated demyelination and neurodegeneration of the CNS. Our previous studies showed that Rho kinase inhibitor Fasudil can delay onset, and ameliorate severity of EAE, accompanied by the improvement in myelination and the inhibition of inflammatory responses in the CNS. In this study, we found that Fasudil inhibited the migration of T cells indirectly by affecting the production of inflammatory factors and the expression of chemokines in astrocytes functions, indicating that Fasudil treatment reduced inflammatory cytokines such as TNF-α and IL-6, reactive oxygen species (NO) and chemokines like MIP-3α (CCL-20), RANTES (CCL5), MIP-1α (CCL-3) and MCP-1 (CCL2) in vitro, and blocked the chemotaxis of reactive mononuclear cells in EAE mice. Further studies found that Fasudil treatment reduced the infiltration and accumulation of pathogenic T cells into the CNS. Astrocytes expressing GFAP and CCL-20 were inhibited in Fasudil-treated EAE compared with control mice. These results demonstrate that Fasudil alleviates the pathogenesis of EAE possibly by blocking astrocyte-derived chemokine-mediated migration of inflammatory macrophages and pathogenic T cells, and might be used to treat MS.
Collapse
Affiliation(s)
- Min-Fang Guo
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong, China
| | - Jian Meng
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong, China
| | - Yan-Hua Li
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong, China
| | - Jie-Zhong Yu
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong, China
| | - Chun-Yun Liu
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong, China
| | - Ling Feng
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong, China
| | - Wan-Fang Yang
- Department of Encephalopathy and National Major Clinical Department of Ministry of Health, Third Hospital, Department of Neurology, Shanxi University of Traditional Chinese Medicine, Taiyuan, China
| | - Jun-Lian Li
- Department of Encephalopathy and National Major Clinical Department of Ministry of Health, Third Hospital, Department of Neurology, Shanxi University of Traditional Chinese Medicine, Taiyuan, China
| | - Qian-Jin Feng
- Department of Encephalopathy and National Major Clinical Department of Ministry of Health, Third Hospital, Department of Neurology, Shanxi University of Traditional Chinese Medicine, Taiyuan, China
| | - Bao-Guo Xiao
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.
| | - Cun-Gen Ma
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong, China; Department of Encephalopathy and National Major Clinical Department of Ministry of Health, Third Hospital, Department of Neurology, Shanxi University of Traditional Chinese Medicine, Taiyuan, China.
| |
Collapse
|
27
|
Paul D, Ge S, Lemire Y, Jellison ER, Serwanski DR, Ruddle NH, Pachter JS. Cell-selective knockout and 3D confocal image analysis reveals separate roles for astrocyte-and endothelial-derived CCL2 in neuroinflammation. J Neuroinflammation 2014; 11:10. [PMID: 24444311 PMCID: PMC3906899 DOI: 10.1186/1742-2094-11-10] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 12/28/2013] [Indexed: 12/30/2022] Open
Abstract
Background Expression of chemokine CCL2 in the normal central nervous system (CNS) is nearly undetectable, but is significantly upregulated and drives neuroinflammation during experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis which is considered a contributing factor in the human disease. As astrocytes and brain microvascular endothelial cells (BMEC) forming the blood–brain barrier (BBB) are sources of CCL2 in EAE and other neuroinflammatory conditions, it is unclear if one or both CCL2 pools are critical to disease and by what mechanism(s). Methods Mice with selective CCL2 gene knockout (KO) in astrocytes (Astro KO) or endothelial cells (Endo KO) were used to evaluate the respective contributions of these sources to neuroinflammation, i.e., clinical disease progression, BBB damage, and parenchymal leukocyte invasion in a myelin oligodendrocyte glycoprotein peptide (MOG35-55)-induced EAE model. High-resolution 3-dimensional (3D) immunofluorescence confocal microscopy and colloidal gold immuno-electron microscopy were employed to confirm sites of CCL2 expression, and 3D immunofluorescence confocal microscopy utilized to assess inflammatory responses along the CNS microvasculature. Results Cell-selective loss of CCL2 immunoreactivity was demonstrated in the respective KO mice. Compared to wild-type (WT) mice, Astro KO mice showed reduced EAE severity but similar onset, while Endo KO mice displayed near normal severity but significantly delayed onset. Neither of the KO mice showed deficits in T cell proliferation, or IL-17 and IFN-γ production, following MOG35-55 exposure in vitro, or altered MOG-major histocompatibility complex class II tetramer binding. 3D confocal imaging further revealed distinct actions of the two CCL2 pools in the CNS. Astro KOs lacked the CNS leukocyte penetration and disrupted immunostaining of CLN-5 at the BBB seen during early EAE in WT mice, while Endo KOs uniquely displayed leukocytes stalled in the microvascular lumen. Conclusions These results point to astrocyte and endothelial pools of CCL2 each regulating different stages of neuroinflammation in EAE, and carry implications for drug delivery in neuroinflammatory disease.
Collapse
Affiliation(s)
| | - Shujun Ge
- Department of Cell Biology, Blood-brain Barrier Laboratory, 263 Farmington Ave,, Farmington CT 06030, USA.
| | | | | | | | | | | |
Collapse
|
28
|
Miljković D, Spasojević I. Multiple sclerosis: molecular mechanisms and therapeutic opportunities. Antioxid Redox Signal 2013; 19:2286-334. [PMID: 23473637 PMCID: PMC3869544 DOI: 10.1089/ars.2012.5068] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Revised: 02/09/2012] [Accepted: 03/09/2013] [Indexed: 12/15/2022]
Abstract
The pathophysiology of multiple sclerosis (MS) involves several components: redox, inflammatory/autoimmune, vascular, and neurodegenerative. All of them are supported by the intertwined lines of evidence, and none of them should be written off. However, the exact mechanisms of MS initiation, its development, and progression are still elusive, despite the impressive pace by which the data on MS are accumulating. In this review, we will try to integrate the current facts and concepts, focusing on the role of redox changes and various reactive species in MS. Knowing the schedule of initial changes in pathogenic factors and the key turning points, as well as understanding the redox processes involved in MS pathogenesis is the way to enable MS prevention, early treatment, and the development of therapies that target specific pathophysiological components of the heterogeneous mechanisms of MS, which could alleviate the symptoms and hopefully stop MS. Pertinent to this, we will outline (i) redox processes involved in MS initiation; (ii) the role of reactive species in inflammation; (iii) prooxidative changes responsible for neurodegeneration; and (iv) the potential of antioxidative therapy.
Collapse
Affiliation(s)
- Djordje Miljković
- Department of Immunology, Institute for Biological Research “Siniša Stanković,” University of Belgrade, Belgrade, Serbia
| | - Ivan Spasojević
- Life Sciences Department, Institute for Multidisciplinary Research, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
29
|
Brambilla R, Morton PD, Ashbaugh JJ, Karmally S, Lambertsen KL, Bethea JR. Astrocytes play a key role in EAE pathophysiology by orchestrating in the CNS the inflammatory response of resident and peripheral immune cells and by suppressing remyelination. Glia 2013; 62:452-67. [DOI: 10.1002/glia.22616] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 11/21/2013] [Accepted: 11/21/2013] [Indexed: 12/19/2022]
Affiliation(s)
- Roberta Brambilla
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, Miller School of Medicine; University of Miami; Miami Florida
- The Neuroscience Program, Miller School of Medicine; University of Miami; Miami Florida
| | - Paul D. Morton
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, Miller School of Medicine; University of Miami; Miami Florida
- The Neuroscience Program, Miller School of Medicine; University of Miami; Miami Florida
| | - Jessica Jopek Ashbaugh
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, Miller School of Medicine; University of Miami; Miami Florida
- Department of Microbiology and Immunology, Miller School of Medicine; University of Miami; Miami Florida
| | - Shaffiat Karmally
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, Miller School of Medicine; University of Miami; Miami Florida
| | - Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine; University of Southern Denmark; Odense Denmark
| | - John R. Bethea
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, Miller School of Medicine; University of Miami; Miami Florida
- The Neuroscience Program, Miller School of Medicine; University of Miami; Miami Florida
- Department of Microbiology and Immunology, Miller School of Medicine; University of Miami; Miami Florida
| |
Collapse
|
30
|
Vasco C, Canazza A, Rizzo A, Mossa A, Corsini E, Silvani A, Fariselli L, Salmaggi A, Ciusani E. Circulating T regulatory cells migration and phenotype in glioblastoma patients: an in vitro study. J Neurooncol 2013; 115:353-63. [DOI: 10.1007/s11060-013-1236-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 08/25/2013] [Indexed: 12/20/2022]
|
31
|
Estrogen mediates neuroprotection and anti-inflammatory effects during EAE through ERα signaling on astrocytes but not through ERβ signaling on astrocytes or neurons. J Neurosci 2013; 33:10924-33. [PMID: 23804112 DOI: 10.1523/jneurosci.0886-13.2013] [Citation(s) in RCA: 146] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Estrogens can signal through either estrogen receptor α (ERα) or β (ERβ) to ameliorate experimental autoimmune encephalomyelitis (EAE), the most widely used mouse model of multiple sclerosis (MS). Cellular targets of estrogen-mediated neuroprotection are still being elucidated. Previously, we demonstrated that ERα on astrocytes, but not neurons, was critical for ERα ligand-mediated neuroprotection in EAE, including decreased T-cell and macrophage inflammation and decreased axonal loss. Here, we determined whether ERβ on astrocytes or neurons could mediate neuroprotection in EAE, by selectively removing ERβ from either of these cell types using Cre-loxP gene deletion. Our results demonstrated that, even though ERβ ligand treatment was neuroprotective in EAE, this neuroprotection was not mediated through ERβ on either astrocytes or neurons and did not involve a reduction in levels of CNS inflammation. Given the differential neuroprotective and anti-inflammatory effects mediated via ERα versus ERβ on astrocytes, we looked for molecules within astrocytes that were affected by signaling through ERα, but not ERβ. We found that ERα ligand treatment, but not ERβ ligand treatment, decreased expression of the chemokines CCL2 and CCL7 by astrocytes in EAE. Together, our data show that neuroprotection in EAE mediated via ERβ signaling does not require ERβ on either astrocytes or neurons, whereas neuroprotection in EAE mediated via ERα signaling requires ERα on astrocytes and reduces astrocyte expression of proinflammatory chemokines. These findings reveal important cellular differences in the neuroprotective mechanisms of estrogen signaling through ERα and ERβ in EAE.
Collapse
|
32
|
Barcia C. Glial-mediated inflammation underlying parkinsonism. SCIENTIFICA 2013; 2013:357805. [PMID: 24278772 PMCID: PMC3820356 DOI: 10.1155/2013/357805] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 06/13/2013] [Indexed: 06/02/2023]
Abstract
The interest in studying neuroimmune interactions is increasing in the scientific community, and for many researchers, immunity is becoming a crucial factor in the understanding of the physiology of the normal brain as well as the biology underlying neurodegenerative diseases. Mounting data over the last two decades point toward immune and inflammatory alterations as important mediators of the progressive dopaminergic degeneration in Parkinson's disease. The purpose of this review is to address, under a historical perspective, as well as in the light of recent reports, the glial-mediated inflammatory and immune responses that occur in Parkinsonism. In line with this, this review also evaluates and highlights available anti-inflammatory drugs and putative targets for Parkinson's disease therapy for the near future.
Collapse
Affiliation(s)
- Carlos Barcia
- Department of Biochemistry and Molecular Biology, Institute of Neuroscience & School of Medicine, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193 Barcelona, Spain
| |
Collapse
|
33
|
Hicks DJ, Núñez A, Banyard AC, Williams A, Ortiz-Pelaez A, Fooks AR, Johnson N. Differential chemokine responses in the murine brain following lyssavirus infection. J Comp Pathol 2013; 149:446-62. [PMID: 23746482 DOI: 10.1016/j.jcpa.2013.04.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 02/20/2013] [Accepted: 04/06/2013] [Indexed: 12/17/2022]
Abstract
The hallmark of lyssavirus infection is lethal encephalomyelitis. Previous studies have reported distinct lyssavirus isolate-related differences in severity of cellular recruitment into the encephalon in a murine model of infection following peripheral inoculation with rabies virus (RABV) and European bat lyssavirus (EBLV)-1 and -2. In order to understand the role of chemokines in this process, comparative studies of the chemokine pattern, distribution and production in response to infection with these lyssaviruses were undertaken. Expression of CCL2, CCL5 and CXCL10 was observed throughout the murine brain with a distinct caudal bias in distribution, similar to both inflammatory changes and virus antigen distribution. CCL2 immunolabelling was localized to neuronal and astroglial populations. CCL5 immunolabelling was only detected in the astroglia, while CXCL10 labelling, although present in the astroglia, was more prominent in neurons. Isolate-dependent differences in the amount of chemokine immunolabelling in specific brain regions and chemokine production by neurons in vitro were observed, with a greater expression of CCL5 in vivo and CXCL10 production in vitro after EBLV infection. Additionally, strong positive associations between chemokine immunolabelling and perivascular cuffing and, to a lesser extent, virus antigen score were also observed. These differences in chemokine expression may explain the variation in severity of encephalitic changes observed in animals infected with different lyssavirus isolates.
Collapse
Affiliation(s)
- D J Hicks
- Pathology Unit, Department of Specialist Scientific Support, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
34
|
Barcia C, Mitxitorena I, Carrillo-de Sauvage MA, Gallego JM, Pérez-Vallés A, Barcia C. Imaging the microanatomy of astrocyte-T-cell interactions in immune-mediated inflammation. Front Cell Neurosci 2013; 7:58. [PMID: 23641198 PMCID: PMC3639405 DOI: 10.3389/fncel.2013.00058] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 04/15/2013] [Indexed: 11/19/2022] Open
Abstract
The role of astrocytes in the immune-mediated inflammatory response in the brain is more prominent than previously thought. Astrocytes become reactive in response to neuro-inflammatory stimuli through multiple pathways, contributing significantly to the machinery that modifies the parenchymal environment. In particular, astrocytic signaling induces the establishment of critical relationships with infiltrating blood cells, such as lymphocytes, which is a fundamental process for an effective immune response. The interaction between astrocytes and T-cells involves complex modifications to both cell types, which undergo micro-anatomical changes and the redistribution of their binding and secretory domains. These modifications are critical for different immunological responses, such as for the effectiveness of the T-cell response, for the specific infiltration of these cells and their homing in the brain parenchyma, and for their correct apposition with antigen-presenting cells (APCs) to form immunological synapses (ISs). In this article, we review the current knowledge of the interactions between T-cells and astrocytes in the context of immune-mediated inflammation in the brain, based on the micro-anatomical imaging of these appositions by high-resolution confocal microscopy and three-dimensional rendering. The study of these dynamic interactions using detailed technical approaches contributes to understanding the function of astrocytes in inflammatory responses and paves the way for new therapeutic strategies.
Collapse
Affiliation(s)
- Carlos Barcia
- Department of Neurosurgery, Hospital General Universitari de València València, Spain
| | | | | | | | | | | |
Collapse
|
35
|
O'Brien ER, Howarth C, Sibson NR. The role of astrocytes in CNS tumors: pre-clinical models and novel imaging approaches. Front Cell Neurosci 2013; 7:40. [PMID: 23596394 PMCID: PMC3627137 DOI: 10.3389/fncel.2013.00040] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 03/26/2013] [Indexed: 02/06/2023] Open
Abstract
Brain metastasis is a significant clinical problem, yet the mechanisms governing tumor cell extravasation across the blood-brain barrier (BBB) and CNS colonization are unclear. Astrocytes are increasingly implicated in the pathogenesis of brain metastasis but in vitro work suggests both tumoricidal and tumor-promoting roles for astrocyte-derived molecules. Also, the involvement of astrogliosis in primary brain tumor progression is under much investigation. However, translation of in vitro findings into in vivo and clinical settings has not been realized. Increasingly sophisticated resources, such as transgenic models and imaging technologies aimed at astrocyte-specific markers, will enable better characterization of astrocyte function in CNS tumors. Techniques such as bioluminescence and in vivo fluorescent cell labeling have potential for understanding the real-time responses of astrocytes to tumor burden. Transgenic models targeting signaling pathways involved in the astrocytic response also hold great promise, allowing translation of in vitro mechanistic findings into pre-clinical models. The challenging nature of in vivo CNS work has slowed progress in this area. Nonetheless, there has been a surge of interest in generating pre-clinical models, yielding insights into cell extravasation across the BBB, as well as immune cell recruitment to the parenchyma. While the function of astrocytes in the tumor microenvironment is still unknown, the relationship between astrogliosis and tumor growth is evident. Here, we review the role of astrogliosis in both primary and secondary brain tumors and outline the potential for the use of novel imaging modalities in research and clinical settings. These imaging approaches have the potential to enhance our understanding of the local host response to tumor progression in the brain, as well as providing new, more sensitive diagnostic imaging methods.
Collapse
Affiliation(s)
- Emma R. O'Brien
- Department of Oncology, CR-UK/MRC Gray Institute for Radiation Oncology and Biology, Churchill Hospital, University of OxfordOxford, UK
| | | | | |
Collapse
|