1
|
Syage AR, Pachow C, Murray KM, Henningfield C, Fernandez K, Du A, Cheng Y, Olivarria G, Kawauchi S, MacGregor GR, Green KN, Lane TE. Cystatin F attenuates neuroinflammation and demyelination following murine coronavirus infection of the central nervous system. J Neuroinflammation 2024; 21:157. [PMID: 38879499 PMCID: PMC11179388 DOI: 10.1186/s12974-024-03153-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 06/12/2024] [Indexed: 06/19/2024] Open
Abstract
BACKGROUND Cystatin F is a secreted lysosomal cysteine protease inhibitor that has been implicated in affecting the severity of demyelination and enhancing remyelination in pre-clinical models of immune-mediated demyelination. How cystatin F impacts neurologic disease severity following viral infection of the central nervous system (CNS) has not been well characterized and was the focus of this study. We used cystatin F null-mutant mice (Cst7-/-) with a well-established model of murine coronavirus-induced neurologic disease to evaluate the contributions of cystatin F in host defense, demyelination and remyelination. METHODS Wildtype controls and Cst7-/- mice were intracranially (i.c.) infected with a sublethal dose of the neurotropic JHM strain of mouse hepatitis virus (JHMV), with disease progression and survival monitored daily. Viral plaque assays and qPCR were used to assess viral levels in CNS. Immune cell infiltration into the CNS and immune cell activation were determined by flow cytometry and 10X genomics chromium 3' single cell RNA sequencing (scRNA-seq). Spinal cord demyelination was determined by luxol fast blue (LFB) and Hematoxylin/Eosin (H&E) staining and axonal damage assessed by immunohistochemical staining for SMI-32. Remyelination was evaluated by electron microscopy (EM) and calculation of g-ratios. RESULTS JHMV-infected Cst7-/- mice were able to control viral replication within the CNS, indicating that cystatin F is not essential for an effective Th1 anti-viral immune response. Infiltration of T cells into the spinal cords of JHMV-infected Cst7-/- mice was increased compared to infected controls, and this correlated with increased axonal damage and demyelination associated with impaired remyelination. Single-cell RNA-seq of CD45 + cells enriched from spinal cords of infected Cst7-/- and control mice revealed enhanced expression of transcripts encoding T cell chemoattractants, Cxcl9 and Cxcl10, combined with elevated expression of interferon-g (Ifng) and perforin (Prf1) transcripts in CD8 + T cells from Cst7-/- mice compared to controls. CONCLUSIONS Cystatin F is not required for immune-mediated control of JHMV replication within the CNS. However, JHMV-infected Cst7-/- mice exhibited more severe clinical disease associated with increased demyelination and impaired remyelination. The increase in disease severity was associated with elevated expression of T cell chemoattractant chemokines, concurrent with increased neuroinflammation. These findings support the idea that cystatin F influences expression of proinflammatory gene expression impacting neuroinflammation, T cell activation and/or glia cell responses ultimately impacting neuroinflammation and neurologic disease.
Collapse
Affiliation(s)
- Amber R Syage
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine, 92697, USA
| | - Collin Pachow
- Department of Molecular Biology & Biochemistry, School of Biological Sciences, University of California, Irvine, 92697, USA
| | - Kaitlin M Murray
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine, 92697, USA
| | - Caden Henningfield
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine, 92697, USA
| | - Kellie Fernandez
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine, 92697, USA
| | - Annie Du
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine, 92697, USA
| | - Yuting Cheng
- Department of Molecular Biology & Biochemistry, School of Biological Sciences, University of California, Irvine, 92697, USA
| | - Gema Olivarria
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine, 92697, USA
| | - Shimako Kawauchi
- Transgenic Mouse Facility, ULAR, Office of Research, University of California, Irvine, 92697, USA
| | - Grant R MacGregor
- Department of Developmental & Cell Biology, University of California, Irvine, 92697, USA
| | - Kim N Green
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine, 92697, USA
| | - Thomas E Lane
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine, 92697, USA.
- Center for Virus Research, University of California, Irvine, 92697, USA.
| |
Collapse
|
2
|
Setlhare B, Letsoalo M, Nkabinde SA, Nkabinde M, Mzobe G, Mtshali A, Parveen S, Ngcobo S, Invernizzi L, Maharaj V, Ngcobo M, Gqaleni N. An in vitro study to elucidate the effects of product Nkabinde on immune response in peripheral blood mononuclear cells of healthy donors. Front Pharmacol 2024; 15:1308913. [PMID: 38533263 PMCID: PMC10963514 DOI: 10.3389/fphar.2024.1308913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 02/26/2024] [Indexed: 03/28/2024] Open
Abstract
Introduction: A significant number of the South African population still rely on traditional medicines (TM) as their primary healthcare due to their belief in their holistic healing and immune-boosting properties. However, little to no scientific data is available on the effects of most TM products on cytokine and cellular biomarkers of the immune response. Here, we evaluated the impact of traditional medicine [Product Nkabinde (PN)] in inducing cellular and cytokine biomarkers of inflammation in peripheral blood mononuclear cells (PBMCs) from eight healthy volunteers. Methods: PN was supplied by a local Traditional Health Practitioner (THP). The IC50 (half maximum concentration) of the standardized extract on isolated PBMCs was established using the cell viability assay over 24 h of incubation. Luminex and flow cytometry assays were used to measure cytokine and cellular levels in PBMCs stimulated with PN and/or PHA over 24, 48, and 72 h, respectively. Results: The IC50 concentration of PN in treated PBMCs was established at 325.3 μg/mL. In the cellular activation assay, the percentages of CD38-HLA-DR + on total CD4+ T cells were significantly increased in PBMCs stimulated with PN compared to unstimulated controls after 24 h (p = 0.008). PN significantly induced the production of anti-inflammatory IL-10 (p = 0.041); proinflammatory cytokines IL-1α (p = 0.003), TNF-α (p < 0.0001); and chemokine MIP-1β (p = 0.046) compared to the unstimulated control after 24 h. At 48 h incubation, the production of proinflammatory cytokines IL-1α (p = 0.034) and TNF-α (p = 0.011) were significantly induced following treatment with PN. Conclusion: We conclude that the PN possesses in vitro immunomodulatory properties that may influence immune and inflammatory responses. More studies using PN are needed to further understand key parameters mediating induction, expression, and regulation of the immune response in the context of pathogen-associated infections.
Collapse
Affiliation(s)
- Boitumelo Setlhare
- Discipline of Traditional Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Marothi Letsoalo
- Centre for Aids Programme of Research in South Africa (CAPRISA), Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Siphathimandla Authority Nkabinde
- Nelson R. Mandela School of Medicine, Doris Duke Medical Research Institute, Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Magugu Nkabinde
- Nelson R. Mandela School of Medicine, Doris Duke Medical Research Institute, Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Gugulethu Mzobe
- Discipline of Traditional Medicine, University of KwaZulu-Natal, Durban, South Africa
- Centre for Aids Programme of Research in South Africa (CAPRISA), Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Andile Mtshali
- Discipline of Traditional Medicine, University of KwaZulu-Natal, Durban, South Africa
- Centre for Aids Programme of Research in South Africa (CAPRISA), Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Sobia Parveen
- Discipline of Traditional Medicine, University of KwaZulu-Natal, Durban, South Africa
- Centre for Aids Programme of Research in South Africa (CAPRISA), Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Samukelisiwe Ngcobo
- Discipline of Traditional Medicine, University of KwaZulu-Natal, Durban, South Africa
- Centre for Aids Programme of Research in South Africa (CAPRISA), Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Luke Invernizzi
- Discipline of Traditional Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Vinesh Maharaj
- Discipline of Traditional Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Mlungisi Ngcobo
- Discipline of Traditional Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Nceba Gqaleni
- Discipline of Traditional Medicine, University of KwaZulu-Natal, Durban, South Africa
- Nelson R. Mandela School of Medicine, Doris Duke Medical Research Institute, Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
3
|
Cheng Y, Javonillo DI, Pachow C, Scarfone VM, Fernandez K, Walsh CM, Green KN, Lane TE. Ablation of microglia following infection of the central nervous system with a neurotropic murine coronavirus infection leads to increased demyelination and impaired remyelination. J Neuroimmunol 2023; 381:578133. [PMID: 37352687 PMCID: PMC11840753 DOI: 10.1016/j.jneuroim.2023.578133] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/12/2023] [Accepted: 06/15/2023] [Indexed: 06/25/2023]
Abstract
Intracranial inoculation of susceptible mice with a glial-tropic strain of mouse hepatitis virus (JHMV), a murine coronavirus, results in an acute encephalomyelitis followed by viral persistence in white matter tracts accompanied by chronic neuroinflammation and demyelination. Microglia are the resident immune cell of the central nervous system (CNS) and are considered important in regulating events associated with neuroinflammation as well as influencing both white matter damage and remyelination. To better understand mechanisms by which microglia contribute to these immune-mediated events, JHMV-infected mice with established demyelination were treated with the small molecular inhibitor of colony stimulating factor 1 receptor (CSF1R), PLX5622, to deplete microglia. Treatment with PLX5622 did not affect viral replication within the CNS yet the severity of demyelination was increased and remyelination impaired compared to control mice. Gene expression analysis revealed that targeting microglia resulted in altered expression of genes associated with immune cell activation and phagocytosis of myelin debris. These findings indicate that microglia are not critical in viral surveillance in persistently JHMV-infected mice yet restrict white matter damage and remyelination, in part, by influencing phagocytosis of myelin debris.
Collapse
Affiliation(s)
- Yuting Cheng
- Department of Molecular Biology & Biochemistry, School of Biological Sciences, University of California, Irvine 92697, USA
| | - Dominic Ibarra Javonillo
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine 92697, USA
| | - Collin Pachow
- Department of Molecular Biology & Biochemistry, School of Biological Sciences, University of California, Irvine 92697, USA
| | - Vanessa M Scarfone
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine 92697, USA
| | - Kellie Fernandez
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine 92697, USA
| | - Craig M Walsh
- Department of Molecular Biology & Biochemistry, School of Biological Sciences, University of California, Irvine 92697, USA
| | - Kim N Green
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine 92697, USA
| | - Thomas E Lane
- Department of Molecular Biology & Biochemistry, School of Biological Sciences, University of California, Irvine 92697, USA; Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine 92697, USA; Center for Virus Research, University of California, Irvine 92697, USA.
| |
Collapse
|
4
|
Olivarria G, Lane TE. Evaluating the role of chemokines and chemokine receptors involved in coronavirus infection. Expert Rev Clin Immunol 2022; 18:57-66. [PMID: 34964406 PMCID: PMC8851429 DOI: 10.1080/1744666x.2022.2017282] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/08/2021] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Coronaviruses are a large family of positive-stranded nonsegmented RNA viruses with genomes of 26-32 kilobases in length. Human coronaviruses are commonly associated with mild respiratory illness; however, the past three decades have seen the emergence of severe acute respiratory coronavirus (SARS-CoV), middle eastern respiratory coronavirus (MERS-CoV), and SARS-CoV-2 which is the etiologic agent for COVID-19. Severe forms of COVID-19 include acute respiratory distress syndrome (ARDS) associated with cytokine release syndrome that can culminate in multiorgan failure and death. Among the proinflammatory factors associated with severe COVID-19 are the chemokines CCL2, CCL3, CXCL8, and CXCL10. Infection of susceptible mice with murine coronaviruses, such as mouse hepatitis virus (MHV), elicits a similar chemokine response profile as observed in COVID-19 patients and these in vivo models have been informative and show that targeting chemokines reduces the severity of inflammation in target organs. AREAS COVERED PubMed was used using keywords: Chemokines and coronaviruses; Chemokines and mouse hepatitis virus; Chemokines and COVID-19. Clinicaltrials.gov was used using keywords: COVID-19 and chemokines; COVID-19 and cytokines; COVID-19 and neutrophil. EXPERT OPINION Chemokines and chemokine receptors are clinically relevant therapeutic targets for reducing coronavirus-induced inflammation.
Collapse
Affiliation(s)
- Gema Olivarria
- Department of Neurobiology & Behavior, University of California, Irvine 92697
| | - Thomas E. Lane
- Department of Neurobiology & Behavior, University of California, Irvine 92697
- Department of Molecular Biology & Behavior, School of Biological Sciences, University of California, Irvine 92697
- Center for Virus Research, University of California, Irvine 92697
| |
Collapse
|
5
|
Nickoloff-Bybel EA, Festa L, Meucci O, Gaskill PJ. Co-receptor signaling in the pathogenesis of neuroHIV. Retrovirology 2021; 18:24. [PMID: 34429135 PMCID: PMC8385912 DOI: 10.1186/s12977-021-00569-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/11/2021] [Indexed: 12/13/2022] Open
Abstract
The HIV co-receptors, CCR5 and CXCR4, are necessary for HIV entry into target cells, interacting with the HIV envelope protein, gp120, to initiate several signaling cascades thought to be important to the entry process. Co-receptor signaling may also promote the development of neuroHIV by contributing to both persistent neuroinflammation and indirect neurotoxicity. But despite the critical importance of CXCR4 and CCR5 signaling to HIV pathogenesis, there is only one therapeutic (the CCR5 inhibitor Maraviroc) that targets these receptors. Moreover, our understanding of co-receptor signaling in the specific context of neuroHIV is relatively poor. Research into co-receptor signaling has largely stalled in the past decade, possibly owing to the complexity of the signaling cascades and functions mediated by these receptors. Examining the many signaling pathways triggered by co-receptor activation has been challenging due to the lack of specific molecular tools targeting many of the proteins involved in these pathways and the wide array of model systems used across these experiments. Studies examining the impact of co-receptor signaling on HIV neuropathogenesis often show activation of multiple overlapping pathways by similar stimuli, leading to contradictory data on the effects of co-receptor activation. To address this, we will broadly review HIV infection and neuropathogenesis, examine different co-receptor mediated signaling pathways and functions, then discuss the HIV mediated signaling and the differences between activation induced by HIV and cognate ligands. We will assess the specific effects of co-receptor activation on neuropathogenesis, focusing on neuroinflammation. We will also explore how the use of substances of abuse, which are highly prevalent in people living with HIV, can exacerbate the neuropathogenic effects of co-receptor signaling. Finally, we will discuss the current state of therapeutics targeting co-receptors, highlighting challenges the field has faced and areas in which research into co-receptor signaling would yield the most therapeutic benefit in the context of HIV infection. This discussion will provide a comprehensive overview of what is known and what remains to be explored in regard to co-receptor signaling and HIV infection, and will emphasize the potential value of HIV co-receptors as a target for future therapeutic development. ![]()
Collapse
Affiliation(s)
- E A Nickoloff-Bybel
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - L Festa
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 S. 40th Street, Philadelphia, PA, 19104, USA
| | - O Meucci
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA.,Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - P J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA.
| |
Collapse
|
6
|
Grant AH, Estrada A, Ayala-Marin YM, Alvidrez-Camacho AY, Rodriguez G, Robles-Escajeda E, Cadena-Medina DA, Rodriguez AC, Kirken RA. The Many Faces of JAKs and STATs Within the COVID-19 Storm. Front Immunol 2021; 12:690477. [PMID: 34326843 PMCID: PMC8313986 DOI: 10.3389/fimmu.2021.690477] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/28/2021] [Indexed: 12/12/2022] Open
Abstract
The positive-sense single stranded RNA virus, Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), resulted in a global pandemic with horrendous health and economic consequences not seen in a century. At a finer scale, immunologically, many of these devastating effects by SARS-CoV-2 can be traced to a "cytokine storm" resulting in the simultaneous activation of Janus Kinases (JAKs) and Signal Transducers and Activators of Transcription (STAT) proteins downstream of the many cytokine receptor families triggered by elevated cytokines found in Coronavirus Disease 2019 (COVID-19). In this report, cytokines found in the storm are discussed in relation to the JAK-STAT pathway in response to SARS-CoV-2 and the lessons learned from RNA viruses and previous Coronaviruses (CoVs). Therapeutic strategies to counteract the SARS-CoV-2 mediated storm are discussed with an emphasis on cell signaling and JAK inhibition.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Robert A. Kirken
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, United States
| |
Collapse
|
7
|
Syage AR, Ekiz HA, Skinner DD, Stone C, O'Connell RM, Lane TE. Single-Cell RNA Sequencing Reveals the Diversity of the Immunological Landscape following Central Nervous System Infection by a Murine Coronavirus. J Virol 2020; 94:e01295-20. [PMID: 32999036 PMCID: PMC7925182 DOI: 10.1128/jvi.01295-20] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/16/2020] [Indexed: 01/12/2023] Open
Abstract
Intracranial (i.c.) infection of susceptible C57BL/6 mice with the neurotropic JHM strain of mouse hepatitis virus (JHMV) (a member of the Coronaviridae family) results in acute encephalomyelitis and viral persistence associated with an immune-mediated demyelinating disease. The present study was undertaken to better understand the molecular pathways evoked during innate and adaptive immune responses as well as the chronic demyelinating stage of disease in response to JHMV infection of the central nervous system (CNS). Using single-cell RNA sequencing analysis (scRNAseq) on flow-sorted CD45-positive (CD45+) cells enriched from brains and spinal cords of experimental mice, we demonstrate the heterogeneity of the immune response as determined by the presence of unique molecular signatures and pathways involved in effective antiviral host defense. Furthermore, we identify potential genes involved in contributing to demyelination as well as remyelination being expressed by both microglia and macrophages. Collectively, these findings emphasize the diversity of the immune responses and molecular networks at defined stages following viral infection of the CNS.IMPORTANCE Understanding the immunological mechanisms contributing to both host defense and disease following viral infection of the CNS is of critical importance given the increasing number of viruses that are capable of infecting and replicating within the nervous system. With this in mind, the present study was undertaken to evaluate the molecular signatures of immune cells within the CNS at defined times following infection with a neuroadapted murine coronavirus using scRNAseq. This approach has revealed that the immunological landscape is diverse, with numerous immune cell subsets expressing distinct mRNA expression profiles that are, in part, dictated by the stage of infection. In addition, these findings reveal new insight into cellular pathways contributing to control of viral replication as well as to neurologic disease.
Collapse
Affiliation(s)
- Amber R Syage
- Division of Microbiology & Immunology, Department of Pathology, School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - H Atakan Ekiz
- Division of Microbiology & Immunology, Department of Pathology, School of Medicine, University of Utah, Salt Lake City, Utah, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Dominic D Skinner
- Division of Microbiology & Immunology, Department of Pathology, School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Colleen Stone
- Division of Microbiology & Immunology, Department of Pathology, School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Ryan M O'Connell
- Division of Microbiology & Immunology, Department of Pathology, School of Medicine, University of Utah, Salt Lake City, Utah, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Thomas E Lane
- Division of Microbiology & Immunology, Department of Pathology, School of Medicine, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
8
|
Mangale V, Syage AR, Ekiz HA, Skinner DD, Cheng Y, Stone CL, Brown RM, O'Connell RM, Green KN, Lane TE. Microglia influence host defense, disease, and repair following murine coronavirus infection of the central nervous system. Glia 2020; 68:2345-2360. [PMID: 32449994 PMCID: PMC7280614 DOI: 10.1002/glia.23844] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/03/2020] [Accepted: 05/04/2020] [Indexed: 12/24/2022]
Abstract
The present study examines functional contributions of microglia in host defense, demyelination, and remyelination following infection of susceptible mice with a neurotropic coronavirus. Treatment with PLX5622, an inhibitor of colony stimulating factor 1 receptor (CSF1R) that efficiently depletes microglia, prior to infection of the central nervous system (CNS) with the neurotropic JHM strain of mouse hepatitis virus (JHMV) resulted in increased mortality compared with control mice that correlated with impaired control of viral replication. Single cell RNA sequencing (scRNASeq) of CD45+ cells isolated from the CNS revealed that PLX5622 treatment resulted in muted CD4+ T cell activation profile that was associated with decreased expression of transcripts encoding MHC class II and CD86 in macrophages but not dendritic cells. Evaluation of spinal cord demyelination revealed a marked increase in white matter damage in PLX5622-treated mice that corresponded with elevated expression of transcripts encoding disease-associated proteins Osteopontin (Spp1), Apolipoprotein E (Apoe), and Triggering receptor expressed on myeloid cells 2 (Trem2) that were enriched within macrophages. In addition, PLX5622 treatment dampened expression of Cystatin F (Cst7), Insulin growth factor 1 (Igf1), and lipoprotein lipase (Lpl) within macrophage populations which have been implicated in promoting repair of damaged nerve tissue and this was associated with impaired remyelination. Collectively, these findings argue that microglia tailor the CNS microenvironment to enhance control of coronavirus replication as well as dampen the severity of demyelination and influence repair.
Collapse
Affiliation(s)
- Vrushali Mangale
- Division of Microbiology & Immunology, Department of PathologyUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Amber R. Syage
- Department of Neurobiology & Behavior, School of Biological SciencesUniversity of CaliforniaIrvineCaliforniaUSA
| | - H. Atakan Ekiz
- Division of Microbiology & Immunology, Department of PathologyUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Dominic D. Skinner
- Division of Microbiology & Immunology, Department of PathologyUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Yuting Cheng
- Department of Neurobiology & Behavior, School of Biological SciencesUniversity of CaliforniaIrvineCaliforniaUSA
| | - Colleen L. Stone
- Division of Microbiology & Immunology, Department of PathologyUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - R. Marshall Brown
- Division of Microbiology & Immunology, Department of PathologyUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Ryan M. O'Connell
- Division of Microbiology & Immunology, Department of PathologyUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Kim N. Green
- Department of Neurobiology & Behavior, School of Biological SciencesUniversity of CaliforniaIrvineCaliforniaUSA
| | - Thomas E. Lane
- Department of Neurobiology & Behavior, School of Biological SciencesUniversity of CaliforniaIrvineCaliforniaUSA
| |
Collapse
|
9
|
Ellwanger JH, Kulmann-Leal B, Kaminski VDL, Rodrigues AG, Bragatte MADS, Chies JAB. Beyond HIV infection: Neglected and varied impacts of CCR5 and CCR5Δ32 on viral diseases. Virus Res 2020; 286:198040. [PMID: 32479976 PMCID: PMC7260533 DOI: 10.1016/j.virusres.2020.198040] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/27/2020] [Accepted: 05/27/2020] [Indexed: 12/18/2022]
Abstract
CCR5 regulates multiple cell types (e.g., T regulatory and Natural Killer cells) and immune responses. The effects of CCR5, CCR5Δ32 (variant associated with reduced CCR5 expression) and CCR5 antagonists vary between infections. CCR5 affects the pathogenesis of flaviviruses, especially in the brain. The genetic variant CCR5Δ32 increases the risk of symptomatic West Nile virus infection. The triad “CCR5, extracellular vesicles and infections” is an emerging topic.
The interactions between chemokine receptors and their ligands may affect susceptibility to infectious diseases as well as their clinical manifestations. These interactions mediate both the traffic of inflammatory cells and virus-associated immune responses. In the context of viral infections, the human C-C chemokine receptor type 5 (CCR5) receives great attention from the scientific community due to its role as an HIV-1 co-receptor. The genetic variant CCR5Δ32 (32 base-pair deletion in CCR5 gene) impairs CCR5 expression on the cell surface and is associated with protection against HIV infection in homozygous individuals. Also, the genetic variant CCR5Δ32 modifies the CCR5-mediated inflammatory responses in various conditions, such as inflammatory and infectious diseases. CCR5 antagonists mimic, at least in part, the natural effects of the CCR5Δ32 in humans, which explains the growing interest in the potential benefits of using CCR5 modulators for the treatment of different diseases. Nevertheless, beyond HIV infection, understanding the effects of the CCR5Δ32 variant in multiple viral infections is essential to shed light on the potential effects of the CCR5 modulators from a broader perspective. In this context, this review discusses the involvement of CCR5 and the effects of the CCR5Δ32 in human infections caused by the following pathogens: West Nile virus, Influenza virus, Human papillomavirus, Hepatitis B virus, Hepatitis C virus, Poliovirus, Dengue virus, Human cytomegalovirus, Crimean-Congo hemorrhagic fever virus, Enterovirus, Japanese encephalitis virus, and Hantavirus. Subsequently, this review addresses the impacts of CCR5 gene editing and CCR5 modulation on health and viral diseases. Also, this article connects recent findings regarding extracellular vesicles (e.g., exosomes), viruses, and CCR5. Neglected and emerging topics in “CCR5 research” are briefly described, with focus on Rocio virus, Zika virus, Epstein-Barr virus, and Rhinovirus. Finally, the potential influence of CCR5 on the immune responses to coronaviruses is discussed.
Collapse
Affiliation(s)
- Joel Henrique Ellwanger
- Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil; Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Bruna Kulmann-Leal
- Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil; Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Valéria de Lima Kaminski
- Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil; Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil; Programa de Pós-Graduação em Biotecnologia, Laboratório de Imunologia Aplicada, Instituto de Ciência e Tecnologia - ICT, Universidade Federal de São Paulo - UNIFESP, São José dos Campos, São Paulo, Brazil
| | - Andressa Gonçalves Rodrigues
- Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Marcelo Alves de Souza Bragatte
- Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil; Núcleo de Bioinformática do Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - José Artur Bogo Chies
- Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil; Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil.
| |
Collapse
|
10
|
Li T, Zhu J. Entanglement of CCR5 and Alzheimer's Disease. Front Aging Neurosci 2019; 11:209. [PMID: 31447666 PMCID: PMC6692443 DOI: 10.3389/fnagi.2019.00209] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 07/24/2019] [Indexed: 12/22/2022] Open
Abstract
Although the mechanisms of Alzheimer's disease are diverse and unclear, the past 20 years have witnessed the unprecedented development of the AD inflammation theory. As a key inflammatory receptor family, the C-C chemokine receptor family is a remarkable participant in the cause of Alzheimer's disease; of this family, CCR5 is the most widely studied. CCR5 is an essential entrance when HIV infects immune cells and is also involved in other inflammatory and immune activities. New evidence on the inevitably intertwined link between Alzheimer's disease and CCR5 indicates that CCR5 accelerates the development of Alzheimer's disease, and few studies disputed it. The role of CCR5 in Alzheimer's disease remains elusive. However, as the research progresses, this intricate relationship will gradually be uncovered.
Collapse
Affiliation(s)
- Tianwen Li
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, Shanghai, China
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianhong Zhu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, Shanghai, China
- Institutes of Brain Science, Shanghai, China
| |
Collapse
|
11
|
Brown DG, Soto R, Yandamuri S, Stone C, Dickey L, Gomes-Neto JC, Pastuzyn ED, Bell R, Petersen C, Buhrke K, Fujinami RS, O'Connell RM, Stephens WZ, Shepherd JD, Lane TE, Round JL. The microbiota protects from viral-induced neurologic damage through microglia-intrinsic TLR signaling. eLife 2019; 8:e47117. [PMID: 31309928 PMCID: PMC6634972 DOI: 10.7554/elife.47117] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 06/10/2019] [Indexed: 12/30/2022] Open
Abstract
Symbiotic microbes impact the function and development of the central nervous system (CNS); however, little is known about the contribution of the microbiota during viral-induced neurologic damage. We identify that commensals aid in host defense following infection with a neurotropic virus through enhancing microglia function. Germfree mice or animals that receive antibiotics are unable to control viral replication within the brain leading to increased paralysis. Microglia derived from germfree or antibiotic-treated animals cannot stimulate viral-specific immunity and microglia depletion leads to worsened demyelination. Oral administration of toll-like receptor (TLR) ligands to virally infected germfree mice limits neurologic damage. Homeostatic activation of microglia is dependent on intrinsic signaling through TLR4, as disruption of TLR4 within microglia, but not the entire CNS (excluding microglia), leads to increased viral-induced clinical disease. This work demonstrates that gut immune-stimulatory products can influence microglia function to prevent CNS damage following viral infection.
Collapse
Affiliation(s)
- D Garrett Brown
- Department of Pathology, Division of Microbiology and ImmunologyUniversity of Utah School of MedicineSalt Lake CityUnited States
| | - Raymond Soto
- Department of Pathology, Division of Microbiology and ImmunologyUniversity of Utah School of MedicineSalt Lake CityUnited States
| | - Soumya Yandamuri
- Department of Pathology, Division of Microbiology and ImmunologyUniversity of Utah School of MedicineSalt Lake CityUnited States
| | - Colleen Stone
- Department of Pathology, Division of Microbiology and ImmunologyUniversity of Utah School of MedicineSalt Lake CityUnited States
| | - Laura Dickey
- Department of Pathology, Division of Microbiology and ImmunologyUniversity of Utah School of MedicineSalt Lake CityUnited States
| | - Joao Carlos Gomes-Neto
- Department of Pathology, Division of Microbiology and ImmunologyUniversity of Utah School of MedicineSalt Lake CityUnited States
| | - Elissa D Pastuzyn
- Department of NeurobiologyUniversity of Utah School of MedicineSalt Lake CityUnited States
| | - Rickesha Bell
- Department of Pathology, Division of Microbiology and ImmunologyUniversity of Utah School of MedicineSalt Lake CityUnited States
| | - Charisse Petersen
- Department of Pathology, Division of Microbiology and ImmunologyUniversity of Utah School of MedicineSalt Lake CityUnited States
| | - Kaitlin Buhrke
- Department of Pathology, Division of Microbiology and ImmunologyUniversity of Utah School of MedicineSalt Lake CityUnited States
| | - Robert S Fujinami
- Department of Pathology, Division of Microbiology and ImmunologyUniversity of Utah School of MedicineSalt Lake CityUnited States
| | - Ryan M O'Connell
- Department of Pathology, Division of Microbiology and ImmunologyUniversity of Utah School of MedicineSalt Lake CityUnited States
| | - W Zac Stephens
- Department of Pathology, Division of Microbiology and ImmunologyUniversity of Utah School of MedicineSalt Lake CityUnited States
| | - Jason D Shepherd
- Department of NeurobiologyUniversity of Utah School of MedicineSalt Lake CityUnited States
| | - Thomas E Lane
- Department of Pathology, Division of Microbiology and ImmunologyUniversity of Utah School of MedicineSalt Lake CityUnited States
| | - June L Round
- Department of Pathology, Division of Microbiology and ImmunologyUniversity of Utah School of MedicineSalt Lake CityUnited States
| |
Collapse
|
12
|
Cheng Y, Skinner DD, Lane TE. Innate Immune Responses and Viral-Induced Neurologic Disease. J Clin Med 2018; 8:jcm8010003. [PMID: 30577473 PMCID: PMC6352557 DOI: 10.3390/jcm8010003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/12/2018] [Accepted: 12/18/2018] [Indexed: 12/19/2022] Open
Abstract
Multiple sclerosis (MS) is a disease of the central nervous system (CNS) characterized by chronic neuroinflammation, axonal damage, and demyelination. Cellular components of the adaptive immune response are viewed as important in initiating formation of demyelinating lesions in MS patients. This notion is supported by preclinical animal models, genome-wide association studies (GWAS), as well as approved disease modifying therapies (DMTs) that suppress clinical relapse and are designed to impede infiltration of activated lymphocytes into the CNS. Nonetheless, emerging evidence demonstrates that the innate immune response e.g., neutrophils can amplify white matter damage through a variety of different mechanisms. Indeed, using a model of coronavirus-induced neurologic disease, we have demonstrated that sustained neutrophil infiltration into the CNS of infected animals correlates with increased demyelination. This brief review highlights recent evidence arguing that targeting the innate immune response may offer new therapeutic avenues for treatment of demyelinating disease including MS.
Collapse
Affiliation(s)
- Yuting Cheng
- Division of Microbiology & Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.
| | - Dominic D Skinner
- Division of Microbiology & Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.
| | - Thomas E Lane
- Division of Microbiology & Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.
| |
Collapse
|
13
|
Dickey LL, Hanley TM, Huffaker TB, Ramstead AG, O'Connell RM, Lane TE. MicroRNA 155 and viral-induced neuroinflammation. J Neuroimmunol 2017; 308:17-24. [PMID: 28139244 DOI: 10.1016/j.jneuroim.2017.01.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 01/11/2017] [Accepted: 01/23/2017] [Indexed: 02/07/2023]
Abstract
MicroRNA (miRNA) regulation of gene expression is becoming an increasingly recognized mechanism by which host immune responses are governed following microbial infection. miRNAs are short, non-coding RNAs that repress translation of target genes, and have been implicated in a number of activities that modulate host immune responses, including the regulation of immune cell proliferation, survival, expansion, differentiation, migration, polarization, and effector function. This review highlights several examples in which mammalian-encoded miR-155 influences immune responses following viral infection of the CNS.
Collapse
Affiliation(s)
- Laura L Dickey
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, United States.
| | - Timothy M Hanley
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, United States.
| | - Thomas B Huffaker
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, United States.
| | - Andrew G Ramstead
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, United States.
| | - Ryan M O'Connell
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, United States.
| | - Thomas E Lane
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, United States.
| |
Collapse
|
14
|
Pan WK, Zhang YF, Yu H, Gao Y, Zheng BJ, Li P, Xie C, Ge X. Identifying key genes associated with Hirschsprung's disease based on bioinformatics analysis of RNA-sequencing data. World J Pediatr 2017; 13:267-273. [PMID: 28120235 PMCID: PMC7091079 DOI: 10.1007/s12519-017-0002-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 08/25/2016] [Indexed: 01/17/2023]
Abstract
BACKGROUND Hirschsprung's disease (HSCR) is a type of megacolon induced by deficiency or dysfunction of ganglion cells in the distal intestine and is associated with developmental disorders of the enteric nervous system. To explore the mechanisms of HSCR, we analyzed the RNA-sequencing data of the expansion and the narrow segments of colon tissues separated from children with HSCR. METHODS RNA-sequencing of the expansion segments and the narrow segments of colon tissues isolated from children with HSCR was performed. After differentially expressed genes (DEGs) were identified using the edgeR package in R, functional and pathway enrichment analyses of DEGs were carried out using DAVID software. To further screen the key genes, protein-protein interaction (PPI) network and module analyses were conducted separately using Cytoscape software. RESULTS A total of 117 DEGs were identified in the expansion segment samples, including 47 up-regulated and 70 down-regulated genes. Functional enrichment analysis suggested that FOS and DUSP1 were implicated in response to endogenous stimulus. In the PPI network analysis, FOS (degree=20), EGR1 (degree=16), ATF3 (degree=9), NOS1 (degree=8), CCL5 (degree=8), DUSP1 (degree=7), CXCL3 (degree=6), VIP (degree=6), FOSB (degree=5), and NOS2 (degree=4) had higher degrees, which could interact with other genes. In addition, two significant modules (module 1 and module 2) were identified from the PPI network. CONCLUSIONS Several genes (including FOS, EGR1, ATF3, NOS1, CCL5, DUSP1, CXCL3, VIP, FOSB, and NOS2) might be involved in the development of HSCR through their effect on the nervous system.
Collapse
Affiliation(s)
- Wei-Kang Pan
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004 China
| | - Ya-Fei Zhang
- Department of Endoscopy, Shaanxi Nuclear Industry 215 Hospital, Xianyang, 712000 China
| | - Hui Yu
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004 China
| | - Ya Gao
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China.
| | - Bai-Jun Zheng
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004 China
| | - Peng Li
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004 China
| | - Chong Xie
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004 China
| | - Xin Ge
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004 China
| |
Collapse
|
15
|
Hutton AJ, Polak ME, Spalluto CM, Wallington JC, Pickard C, Staples KJ, Warner JA, Wilkinson TMA. Human Lung Fibroblasts Present Bacterial Antigens to Autologous Lung Th Cells. THE JOURNAL OF IMMUNOLOGY 2016; 198:110-118. [PMID: 27895174 DOI: 10.4049/jimmunol.1600602] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 10/28/2016] [Indexed: 11/19/2022]
Abstract
Lung fibroblasts are key structural cells that reside in the submucosa where they are in contact with large numbers of CD4+ Th cells. During severe viral infection and chronic inflammation, the submucosa is susceptible to bacterial invasion by lung microbiota such as nontypeable Haemophilus influenzae (NTHi). Given their proximity in tissue, we hypothesized that human lung fibroblasts play an important role in modulating Th cell responses to NTHi. We demonstrate that fibroblasts express the critical CD4+ T cell Ag-presentation molecule HLA-DR within the human lung, and that this expression can be recapitulated in vitro in response to IFN-γ. Furthermore, we observed that cultured lung fibroblasts could internalize live NTHi. Although unable to express CD80 and CD86 in response to stimulation, fibroblasts expressed the costimulatory molecules 4-1BBL, OX-40L, and CD70, all of which are related to memory T cell activation and maintenance. CD4+ T cells isolated from the lung were predominantly (mean 97.5%) CD45RO+ memory cells. Finally, cultured fibroblasts activated IFN-γ and IL-17A cytokine production by autologous, NTHi-specific lung CD4+ T cells, and cytokine production was inhibited by a HLA-DR blocking Ab. These results indicate a novel role for human lung fibroblasts in contributing to responses against bacterial infection through activation of bacteria-specific CD4+ T cells.
Collapse
Affiliation(s)
- Andrew J Hutton
- Clinical and Experimental Sciences Unit, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom; and
| | - Marta E Polak
- Clinical and Experimental Sciences Unit, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom; and
| | - C Mirella Spalluto
- Clinical and Experimental Sciences Unit, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom; and
| | - Joshua C Wallington
- Clinical and Experimental Sciences Unit, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom; and
| | - Chris Pickard
- Clinical and Experimental Sciences Unit, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom; and
| | - Karl J Staples
- Clinical and Experimental Sciences Unit, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom; and
| | - Jane A Warner
- Clinical and Experimental Sciences Unit, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom; and
| | - Tom M A Wilkinson
- Clinical and Experimental Sciences Unit, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom; and.,National Institute for Health Research Southampton Respiratory Biomedical Research Unit, Southampton General Hospital, Southampton SO16 6YD, United Kingdom
| |
Collapse
|
16
|
Savarin C, Bergmann CC, Hinton DR, Stohlman SA. Differential Regulation of Self-reactive CD4 + T Cells in Cervical Lymph Nodes and Central Nervous System during Viral Encephalomyelitis. Front Immunol 2016; 7:370. [PMID: 27708643 PMCID: PMC5030268 DOI: 10.3389/fimmu.2016.00370] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/07/2016] [Indexed: 12/12/2022] Open
Abstract
Viral infections have long been implicated as triggers of autoimmune diseases, including multiple sclerosis (MS), a central nervous system (CNS) inflammatory demyelinating disorder. Epitope spreading, molecular mimicry, cryptic antigen, and bystander activation have been implicated as mechanisms responsible for activating self-reactive (SR) immune cells, ultimately leading to organ-specific autoimmune disease. Taking advantage of coronavirus JHM strain of mouse hepatitis virus (JHMV)-induced demyelination, this study demonstrates that the host also mounts counteractive measures to specifically limit expansion of endogenous SR T cells. In this model, immune-mediated demyelination is associated with induction of SR T cells after viral control. However, their decline during persisting infection, despite ongoing demyelination, suggests an active control mechanism. Antigen-specific IL-10-secreting CD4+ T cells (Tr1) and Foxp3+ regulatory T cells (Tregs), both known to control autoimmunity and induced following JHMV infection, were assessed for their relative in vivo suppressive function of SR T cells. Ablation of Foxp3+ Tregs in chronically infected DEREG mice significantly increased SR CD4+ T cells within cervical lymph nodes (CLN), albeit without affecting their numbers or activation within the CNS compared to controls. In contrast, infected IL-27 receptor deficient (IL-27R-/-) mice, characterized by a drastic reduction of Tr1 cells, revealed that SR CD4+ T cells in CLN remained unchanged but were specifically increased within the CNS. These results suggest that distinct Treg subsets limit SR T cells in the draining lymph nodes and CNS to maximize suppression of SR T-cell-mediated autoimmune pathology. The JHMV model is thus valuable to decipher tissue-specific mechanisms preventing autoimmunity.
Collapse
Affiliation(s)
- Carine Savarin
- Department of Neurosciences, NC-30, Lerner Research Institute, Cleveland Clinic Foundation , Cleveland, OH , USA
| | - Cornelia C Bergmann
- Department of Neurosciences, NC-30, Lerner Research Institute, Cleveland Clinic Foundation , Cleveland, OH , USA
| | - David R Hinton
- Department of Pathology, Keck School of Medicine, University of Southern California , Los Angeles, CA , USA
| | - Stephen A Stohlman
- Department of Neurosciences, NC-30, Lerner Research Institute, Cleveland Clinic Foundation , Cleveland, OH , USA
| |
Collapse
|
17
|
Dickey LL, Worne CL, Glover JL, Lane TE, O’Connell RM. MicroRNA-155 enhances T cell trafficking and antiviral effector function in a model of coronavirus-induced neurologic disease. J Neuroinflammation 2016; 13:240. [PMID: 27604627 PMCID: PMC5015201 DOI: 10.1186/s12974-016-0699-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 08/20/2016] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) are noncoding RNAs that modulate cellular gene expression, primarily at the post-transcriptional level. We sought to examine the functional role of miR-155 in a model of viral-induced neuroinflammation. METHODS Acute encephalomyelitis and immune-mediated demyelination were induced by intracranial injection with the neurotropic JHM strain of mouse hepatitis virus (JHMV) into C57BL/6 miR-155 (+/+) wildtype (WT) mice or miR-155 (-/-) mice. Morbidity and mortality, viral load and immune cell accumulation in the CNS, and spinal cord demyelination were assessed at defined points post-infection. T cells harvested from infected mice were used to examine cytolytic activity, cytokine activity, and expression of certain chemokine receptors. To determine the impact of miR-155 on trafficking, T cells from infected WT or miR-155 (-/-) mice were adoptively transferred into RAG1 (-/-) mice, and T cell accumulation into the CNS was assessed using flow cytometry. Statistical significance was determined using the Mantel-Cox log-rank test or Student's T tests. RESULTS Compared to WT mice, JHMV-infected miR-155 (-/-) mice developed exacerbated disease concomitant with increased morbidity/mortality and an inability to control viral replication within the CNS. In corroboration with increased susceptibility to disease, miR-155 (-/-) mice had diminished CD8(+) T cell responses in terms of numbers, cytolytic activity, IFN-γ secretion, and homing to the CNS that corresponded with reduced expression of the chemokine receptor CXCR3. Both IFN-γ secretion and trafficking were impaired in miR-155 (-/-) , virus-specific CD4(+) T cells; however, expression of the chemokine homing receptors analyzed on CD4(+) cells was not affected. Except for very early during infection, there were not significant differences in macrophage infiltration into the CNS between WT and miR-155 (-/-) JHMV-infected mice, and the severity of demyelination was similar at 14 days p.i. between WT and miR-155 (-/-) JHMV-infected mice. CONCLUSIONS These findings support a novel role for miR-155 in host defense in a model of viral-induced encephalomyelitis. Specifically, miR-155 enhances antiviral T cell responses including cytokine secretion, cytolytic activity, and homing to the CNS in response to viral infection. Further, miR-155 can play either a host-protective or host-damaging role during neuroinflammation depending on the disease trigger.
Collapse
Affiliation(s)
- Laura L. Dickey
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112 USA
| | - Colleen L. Worne
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112 USA
| | - Jessica L. Glover
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112 USA
| | - Thomas E. Lane
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112 USA
| | - Ryan M. O’Connell
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112 USA
| |
Collapse
|
18
|
Huber AK, Duncker PC, Irani DN. Immune responses to non-tumor antigens in the central nervous system. Front Oncol 2014; 4:328. [PMID: 25431758 PMCID: PMC4230036 DOI: 10.3389/fonc.2014.00328] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 10/30/2014] [Indexed: 12/16/2022] Open
Abstract
The central nervous system (CNS), once viewed as an immune-privileged site protected by the blood-brain barrier (BBB), is now known to be a dynamic immunological environment through which immune cells migrate to prevent and respond to events such as localized infection. During these responses, endogenous glial cells, including astrocytes and microglia, become highly reactive and may secrete inflammatory mediators that regulate BBB permeability and recruit additional circulating immune cells. Here, we discuss the various roles played by astrocytes, microglia, and infiltrating immune cells during host immunity to non-tumor antigens in the CNS, focusing first on bacterial and viral infections, and then turning to responses directed against self-antigens in the setting of CNS autoimmunity.
Collapse
Affiliation(s)
- Amanda K Huber
- Department of Neurology, University of Michigan Medical School , Ann Arbor, MI , USA
| | - Patrick C Duncker
- Department of Neurology, University of Michigan Medical School , Ann Arbor, MI , USA
| | - David N Irani
- Department of Neurology, University of Michigan Medical School , Ann Arbor, MI , USA
| |
Collapse
|
19
|
Libbey JE, Lane TE, Fujinami RS. Axonal pathology and demyelination in viral models of multiple sclerosis. DISCOVERY MEDICINE 2014; 18:79-89. [PMID: 25091490 PMCID: PMC4371782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Multiple sclerosis (MS) is an immune-mediated inflammatory demyelinating disease of the central nervous system (CNS). Monozygotic twin studies suggest that while there is a genetic contribution, genetics alone cannot be the sole determining factor in the development of MS. As the rates of MS are increasing, particularly among women, environmental factors such as viral infections are coming to the foreground as potential agents in triggering disease in genetically susceptible individuals. This review highlights pathological aspects related to two pre-clinical viral models for MS; data are consistent between these two models as experimental infection of susceptible mice can induce axonal degeneration associated with demyelination. These data are consistent with observations in MS that axonal damage or Wallerian degeneration is occurring within the CNS contributing to the disability and disease severity. Such early damage, where axonal damage is primary to secondary demyelination, could set the stage for more extensive immune mediated demyelination arising later.
Collapse
Affiliation(s)
- Jane E Libbey
- Department of Pathology, University of Utah, 15 North Medical Drive East, 2600A EEJMRB, Salt Lake City, UT 84112, USA
| | | | | |
Collapse
|
20
|
Hosking MP, Lane TE. ELR(+) chemokine signaling in host defense and disease in a viral model of central nervous system disease. Front Cell Neurosci 2014; 8:165. [PMID: 24987333 PMCID: PMC4060560 DOI: 10.3389/fncel.2014.00165] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 05/31/2014] [Indexed: 11/23/2022] Open
Abstract
Intracranial infection of the neurotropic JHM strain of mouse hepatitis virus (JHMV) into the central nervous system (CNS) of susceptible strains of mice results in an acute encephalomyelitis, accompanied by viral replication in glial cells and robust infiltration of virus-specific T cells that contribute to host defense through cytokine secretion and cytolytic activity. Mice surviving the acute stage of disease develop an immune-mediated demyelinating disease, characterized by viral persistence in white matter tracts and a chronic neuroinflammatory response dominated by T cells and macrophages. Chemokines and their corresponding chemokine receptors are dynamically expressed throughout viral infection of the CNS, influencing neuroinflammation by regulating immune cell infltration and glial biology. This review is focused upon the pleiotropic chemokine receptor CXCR2 and its effects upon neutrophils and oligodendrocytes during JHMV infection and a number of other models of CNS inflammation.
Collapse
Affiliation(s)
- Martin P. Hosking
- Department of Molecular Biology and Biochemistry, University of CaliforniaIrvine, CA, USA
| | - Thomas E. Lane
- Department of Pathology, Division of Microbiology and Immunology, School of Medicine, University of UtahSalt Lake City, UT, USA
| |
Collapse
|
21
|
Abstract
UNLABELLED The encephalitic response to viral infection requires local chemokine production and the ensuing recruitment of immune and inflammatory leukocytes. Accordingly, chemokine receptors present themselves as plausible therapeutic targets for drugs aimed at limiting encephalitic responses. However, it remains unclear which chemokines are central to this process and whether leukocyte recruitment is important for limiting viral proliferation and survival in the brain or whether it is predominantly a driver of coincident inflammatory pathogenesis. Here we examine chemokine expression and leukocyte recruitment in the context of avirulent and virulent Semliki Forest virus (SFV) as well as West Nile virus infection and demonstrate rapid and robust expression of a variety of inflammatory CC and CXC chemokines in all models. On this basis, we define a chemokine axis involved in leukocyte recruitment to the encephalitic brain during SFV infection. CXCR3 is the most active; CCR2 is also active but less so, and CCR5 plays only a modest role in leukocyte recruitment. Importantly, inhibition of each of these receptors individually and the resulting suppression of leukocyte recruitment to the infected brain have no effect on viral titer or survival following infection with a virulent SFV strain. In contrast, simultaneous blockade of CXCR3 and CCR2 results in significantly reduced mortality in response to virulent SFV infection. In summary, therefore, our data provide an unprecedented level of insight into chemokine orchestration of leukocyte recruitment in viral encephalitis. Our data also highlight CXCR3 and CCR2 as possible therapeutic targets for limiting inflammatory damage in response to viral infection of the brain. IMPORTANCE Brain inflammation (encephalitis) in response to viral infection can lead to severe illness and even death. This therefore represents an important clinical problem and one that requires the development of new therapeutic approaches. Central to the pathogenesis of encephalitis is the recruitment of inflammatory leukocytes to the infected brain, a process driven by members of the chemokine family. Here we provide an in-depth analysis of the chemokines involved in leukocyte recruitment to the virally infected brain and demonstrate that simultaneous blockade of two of these receptors, namely, CXCR3 and CCR2, does not alter viral titers within the brain but markedly reduces inflammatory leukocyte recruitment and enhances survival in a murine model of lethal viral encephalitis. Our results therefore highlight chemokine receptors as plausible therapeutic targets in treating viral encephalitis.
Collapse
|
22
|
Kapil P, Stohlman SA, Hinton DR, Bergmann CC. PKR mediated regulation of inflammation and IL-10 during viral encephalomyelitis. J Neuroimmunol 2014; 270:1-12. [PMID: 24642385 PMCID: PMC4019976 DOI: 10.1016/j.jneuroim.2014.02.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 02/24/2014] [Accepted: 02/25/2014] [Indexed: 12/21/2022]
Abstract
Double-stranded RNA-dependent protein kinase (PKR) regulates antiviral activity, immune responses, apoptosis and neurotoxicity. Gliatropic coronavirus infection induced PKR activation in infected as well uninfected cells within the central nervous system (CNS). However, PKR deficiency only modestly increased viral replication and did not affect IFN-α/β or IL-1β expression. Despite reduced Il-6, Ccl5, and Cxcl10 mRNA, protein levels remained unaltered. Furthermore, PKR deficiency selectively reduced IL-10 production in CD4, but not CD8 T cells, without affecting CNS pathology. The results demonstrate the ability of PKR to balance neuroinflammation by selectively modulating key cytokines and chemokines in CNS resident and CD4 T cells.
Collapse
Affiliation(s)
- Parul Kapil
- Department of Neurosciences, NC-30, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Stephen A Stohlman
- Department of Neurosciences, NC-30, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - David R Hinton
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Cornelia C Bergmann
- Department of Neurosciences, NC-30, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA.
| |
Collapse
|
23
|
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
|
24
|
Goldeck D, Larbi A, Pellicanó M, Alam I, Zerr I, Schmidt C, Fulop T, Pawelec G. Enhanced Chemokine Receptor Expression on Leukocytes of Patients with Alzheimer's Disease. PLoS One 2013; 8:e66664. [PMID: 23824053 PMCID: PMC3688934 DOI: 10.1371/journal.pone.0066664] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Accepted: 05/08/2013] [Indexed: 01/23/2023] Open
Abstract
Although primarily a neurological complaint, systemic inflammation is present in Alzheimer's Disease, with higher than normal levels of proinflammatory cytokines and chemokines in the periphery as well as the brain. A gradient of these factors may enhance recruitment of activated immune cells into the brain via chemotaxis. Here, we investigated the phenotypes of circulating immune cells in AD patients with multi-colour flow cytometry to determine whether their expression of chemokine receptors is consistent with this hypothesis. In this study, we confirmed our previously reported data on the shift of early- to late-differentiated CD4+ T-cells in AD patients. The percentage of cells expressing CD25, a marker of acute T-cell activation, was higher in patients than in age-matched controls, and percentages of CCR6+ cells were elevated. This chemokine receptor is primarily expressed on pro-inflammatory memory cells and Th17 cells. The proportion of cells expressing CCR4 (expressed on Th2 cells) and CCR5 (Th1 cells and dendritic cells) was also greater in patients, and was more pronounced on CD4+ than CD8+ T-cells. These findings allow a more detailed insight into the systemic immune status of patients with Alzheimer's disease and suggest possible novel targets for immune therapy.
Collapse
Affiliation(s)
- David Goldeck
- Tübingen Ageing and Tumour Immunology Group, Center for Medical Research, University of Tübingen, Tübingen, Germany
- * E-mail:
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research, Biopolis, Singapore, Singapore
| | - Mariavaleria Pellicanó
- Tübingen Ageing and Tumour Immunology Group, Center for Medical Research, University of Tübingen, Tübingen, Germany
| | - Iftikhar Alam
- Tübingen Ageing and Tumour Immunology Group, Center for Medical Research, University of Tübingen, Tübingen, Germany
- Abdul Wali Khan University, Mardan, Pakistan
| | - Inga Zerr
- Clinical Dementia Center, Dept. of Neurology, University Medical School, Georg August University, Göttingen, Germany
| | - Christian Schmidt
- Clinical Dementia Center, Dept. of Neurology, University Medical School, Georg August University, Göttingen, Germany
| | - Tamas Fulop
- Immunology Program, Geriatric Division, Faculty of Medicine, Research Center on Aging, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Graham Pawelec
- Tübingen Ageing and Tumour Immunology Group, Center for Medical Research, University of Tübingen, Tübingen, Germany
| |
Collapse
|
25
|
Weinger JG, Marro BS, Hosking MP, Lane TE. The chemokine receptor CXCR2 and coronavirus-induced neurologic disease. Virology 2013; 435:110-7. [PMID: 23217621 PMCID: PMC3522860 DOI: 10.1016/j.virol.2012.08.049] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 08/31/2012] [Indexed: 12/18/2022]
Abstract
Inoculation with the neurotropic JHM strain of mouse hepatitis virus (MHV) into the central nervous system (CNS) of susceptible strains of mice results in an acute encephalomyelitis in which virus preferentially replicates within glial cells while excluding neurons. Control of viral replication during acute disease is mediated by infiltrating virus-specific T cells via cytokine secretion and cytolytic activity, however sterile immunity is not achieved and virus persists resulting in chronic neuroinflammation associated with demyelination. CXCR2 is a chemokine receptor that upon binding to specific ligands promotes host defense through recruitment of myeloid cells to the CNS as well as protecting oligodendroglia from cytokine-mediated death in response to MHV infection. These findings highlight growing evidence of the diverse and important role of CXCR2 in regulating neuroinflammatory diseases.
Collapse
Affiliation(s)
- Jason G Weinger
- Department of Molecular Biology & Biochemistry, UC Irvine, CA 92697-3900, USA
| | | | | | | |
Collapse
|
26
|
Larena M, Regner M, Lobigs M. The chemokine receptor CCR5, a therapeutic target for HIV/AIDS antagonists, is critical for recovery in a mouse model of Japanese encephalitis. PLoS One 2012; 7:e44834. [PMID: 23028638 PMCID: PMC3448613 DOI: 10.1371/journal.pone.0044834] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 08/09/2012] [Indexed: 11/19/2022] Open
Abstract
Japanese encephalitis is a severe central nervous system (CNS) inflammatory disease caused by the mosquito-borne flavivirus, Japanese encephalitis virus (JEV). In the current study we have investigated the immune responses against JEV in mice lacking expression of the chemokine receptor CCR5, which functions in activation and chemotaxis of leukocytes during infection. We show that CCR5 serves as a host antiviral factor against Japanese encephalitis, with CCR5 deficiency markedly increasing mortality, and viral burden in the CNS. Humoral immune responses, which are essential in recovery from JEV infection, were of similar magnitude in CCR5 sufficient and deficient mice. However, absence of CCR5 resulted in a multifaceted deficiency of cellular immune responses characterized by reduced natural killer and CD8⁺ T cell activity, low splenic cellularity, and impaired trafficking of leukocytes to the brain. Interestingly, adoptive transfer of immune spleen cells, depleted of B lymphocytes, increased resistance of CCR5-deficient recipient mice against JEV regardless of whether the cells were obtained from CCR5-deficient or wild-type donor mice, and only when transferred at one but not at three days post-challenge. This result is consistent with a mechanism by which CCR5 expression enhances lymphocyte activation and thereby promotes host survival in Japanese encephalitis.
Collapse
Affiliation(s)
- Maximilian Larena
- Department of Emerging Pathogens and Vaccines, John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Matthias Regner
- Department of Emerging Pathogens and Vaccines, John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Mario Lobigs
- Department of Emerging Pathogens and Vaccines, John Curtin School of Medical Research, The Australian National University, Canberra, Australia
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Australia
| |
Collapse
|
27
|
Marro BS, Hosking MP, Lane TE. CXCR2 signaling and host defense following coronavirus-induced encephalomyelitis. Future Virol 2012; 7:349-359. [PMID: 22582084 DOI: 10.2217/fvl.12.23] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Inoculation of the neurotropic JHM strain of mouse hepatitis virus (JHMV) into the central nervous system (CNS) of susceptible strains of mice results in wide-spread replication within glial cells accompanied by infiltration of virus-specific T lymphocytes that control virus through cytokine secretion and cytolytic activity. Virus persists within white matter tracts of surviving mice resulting in demyelination that is amplified by inflammatory T cells and macrophages. In response to infection, numerous cytokines/chemokines are secreted by resident cells of the CNS and inflammatory leukocytes that participate in both host defense and disease. Among these are the ELR-positive chemokines that are able to signal through CXC chemokine receptors including CXCR2. Early following JHMV infection, ELR-positive chemokines contribute to host defense by attracting CXCR2-expressing cells including polymorphonuclear cells to the CNS that aid in host defense through increasing the permeability the blood-brain-barrier (BBB). During chronic disease, CXCR2 signaling on oligodendroglia protects these cells from apoptosis and restricts the severity of demyelination. This review covers aspects related to host defense and disease in response to JHMV infection and highlights the different roles of CXCR2 signaling in these processes.
Collapse
Affiliation(s)
- Brett S Marro
- Department of Molecular Biology & Biochemistry, University of California, Irvine 92697-3900
| | | | | |
Collapse
|
28
|
Sorce S, Myburgh R, Krause KH. The chemokine receptor CCR5 in the central nervous system. Prog Neurobiol 2010; 93:297-311. [PMID: 21163326 DOI: 10.1016/j.pneurobio.2010.12.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Revised: 11/29/2010] [Accepted: 12/07/2010] [Indexed: 02/04/2023]
Abstract
The expression and the role of the chemokine receptor CCR5 have been mainly studied in the context of HIV infection. However, this protein is also expressed in the brain, where it can be crucial in determining the outcome in response to different insults. CCR5 expression can be deleterious or protective in controlling the progression of certain infections in the CNS, but it is also emerging that it could play a role in non-infectious diseases. In particular, it appears that, in addition to modulating immune responses, CCR5 can influence neuronal survival. Here, we summarize the present knowledge about the expression of CCR5 in the brain and highlight recent findings suggesting its possible involvement in neuroprotective mechanisms.
Collapse
Affiliation(s)
- Silvia Sorce
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, 1211 Geneva-4, Switzerland
| | | | | |
Collapse
|
29
|
Meucci O. HIV Coreceptors and Their Roles in Leukocyte Trafficking During Neuroinflammatory Diseases. CHEMOKINE RECEPTORS AND NEUROAIDS 2010. [PMCID: PMC7120588 DOI: 10.1007/978-1-4419-0793-6_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Due to the increasing resistance of HIV-1 to antiretroviral therapies, there has been much emphasis on the discovery and development of alternative therapeutics for HIV-1-infected individuals. The chemokine receptors CXCR4 (Bleul et al. 1996a; Feng et al. 1996; Nagasawa et al. 1996; Oberlin et al. 1996) and CCR5 (Alkhatib et al. 1996; Deng et al. 1996; Dragic et al. 1996) were identified as target molecules from the time their role as coreceptors for HIV-1 entry into leukocytes was first discovered 10 years ago. Initial studies focused on the use of the chemokine ligands, or altered derivatives, of CXCR4 and CCR5 to prevent the entrance of HIV-1 into immune cells (Schols 2006). While these studies showed some initial promise, there was evidence of significant caveats to their use, including selection of alternative coreceptor utilizing strains (Marechal et al. 1999; Mosier et al. 1999) and the potential to cause inflammatory side effects. These data prompted the development and study of small molecule inhibitors of CXCR4 and CCR5, which have also been used to examine the roles of these molecules in a variety of inflammatory and infectious diseases.
Collapse
Affiliation(s)
- Olimpia Meucci
- Dept. Pharmacology & Physiology, & Institute of Molecular Medicine, Drexel University College of Medicine, North 15th St. 245, Philadelphia, 19102-1101 USA
| |
Collapse
|
30
|
Abstract
Mouse hepatitis virus (MHV) is a positive-strand RNA virus that causes an acute encephalomyelitis that later resolves into a chronic fulminating demyelinating disease. Cytokine production, chemokine secretion, and immune cell infiltration into the central nervous system are critical to control viral replication during acute infection. Despite potent antiviral T-lymphocyte activity, sterile immunity is not achieved, and MHV chronically persists within oligodendrocytes. Continued infiltration and activation of the immune system, a result of the lingering viral antigen and RNA within oligodendrocytes, lead directly to the development of an immune-mediated demyelination that bears remarkable similarities, both clinically and histologically, to the human demyelinating disease multiple sclerosis. MHV offers a unique model system for studying host defense during acute viral infection and immune-mediated demyelination during chronic infection.
Collapse
Affiliation(s)
- Martin P. Hosking
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697-3900 USA
| | - Thomas E. Lane
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697-3900 USA
- Institute for Immunology, University of California, Irvine, CA 92697-3900 USA
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, CA 92697-3900 USA
| |
Collapse
|
31
|
Jalosinski M, Karolczak K, Mazurek A, Glabinski A. The effects of methylprednisolone and mitoxantrone on CCL5-induced migration of lymphocytes in multiple sclerosis. Acta Neurol Scand 2008; 118:120-5. [PMID: 18336625 DOI: 10.1111/j.1600-0404.2008.00998.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
OBJECTIVES Chemokines are involved in migration of inflammatory cells to the central nervous system (CNS) in multiple sclerosis (MS). The aim of this study was the analysis of the impact of MS treatment on CCL5-induced migration of leukocyte subpopulations. MATERIAL AND METHODS Migration of lymphocytes and monocytes from blood of MS patients treated with methylprednisolone (MP) or mitoxantrone (MTX) was analysed in a chemotaxis chamber. RESULTS CCL5-induced migration of lymphocytes from untreated MS patients was significantly increased over controls. The treatment of MS with MP and MTX reduced this chemotaxis. The plasma level of CCL5 was increased in MS patients before treatment and was also significantly decreased in the treatment of MS with MP and MTX. CONCLUSIONS This observation supports the hypothesis that in MS, chemokine CCL5 may induce migration of leukocytes to the CNS and suggests that treatment of the disease with MP and MTX may reduce this migration.
Collapse
Affiliation(s)
- M Jalosinski
- Department of Experimental and Clinical Neurology, Medical University of Lodz, Lodz, Poland
| | | | | | | |
Collapse
|
32
|
Schaumburg CS, Held KS, Lane TE. Mouse hepatitis virus infection of the CNS: a model for defense, disease, and repair. FRONT BIOSCI-LANDMRK 2008; 13:4393-406. [PMID: 18508518 DOI: 10.2741/3012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Viral infection of the central nervous system (CNS) results in varied outcomes ranging from encephalitis, paralytic poliomyelitis or other serious consequences. One of the principal factors that directs the outcome of infection is the localized innate immune response, which is proceeded by the adaptive immune response against the invading viral pathogen. The role of the immune system is to contain and control the spread of virus within the CNS, and paradoxically, this response may also be pathological. Studies with a neurotropic murine coronavirus, mouse hepatitis virus (MHV) have provided important insights into how the immune system combats neuroinvasive viruses, and have identified molecular and cellular mechanisms contributing to chronic disease in persistently infected mice.
Collapse
Affiliation(s)
- Chris S Schaumburg
- Department of Molecular Biology and Biochemistry, University of California, Irvine 92697-3900, USA
| | | | | |
Collapse
|
33
|
Carr DJ, Wuest T, Ash J. An increase in herpes simplex virus type 1 in the anterior segment of the eye is linked to a deficiency in NK cell infiltration in mice deficient in CXCR3. J Interferon Cytokine Res 2008; 28:245-51. [PMID: 18439102 PMCID: PMC2396780 DOI: 10.1089/jir.2007.0110] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In response to ocular herpes simplex virus type 1 (HSV-1) infection in mice, a rapid induction or increase in the local expression of chemokines, including CXCL10, is found. The present study investigated the role of the receptor for CXCL10, CXCR3, in the host response to corneal HSV-1 infection. Mice deficient in CXCR3 (CXCR3(-/-)) were found to have an increase in infectious virus in the anterior segment of the eye by day 7 postinfection. Coinciding with the increase, selective chemokines, including CCL2, CCL3, CCL5, CXCL9, and CXCL10, were elevated in the anterior segment of the HSV-1-infected CXCR3(-/-) mice. In contrast, there was a time-dependent reduction in the recruitment of natural killer (NK) cells (NK1.1(+)CD3(-)) into the anterior segment of CXCR3(-/-) mice. A reduction in NK cells residing in the anterior segment of mice following antiasialoGM1 antibody treatment resulted in an increase in infectious virus. No other leukocyte populations infiltrating the tissue were modified in the absence of CXCR3. Collectively, the loss of CXCR3 expression specifically reduces NK cell mobilization into the cornea in response to HSV-1.
Collapse
Affiliation(s)
- Daniel J.J. Carr
- Department of Microbiology, Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Todd Wuest
- Department of Microbiology, Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - John Ash
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| |
Collapse
|
34
|
Hickey MJ, Held KS, Baum E, Gao JL, Murphy PM, Lane TE. CCR1 deficiency increases susceptibility to fatal coronavirus infection of the central nervous system. Viral Immunol 2008; 20:599-608. [PMID: 18158733 DOI: 10.1089/vim.2007.0056] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The role of CC chemokine receptor 1 (CCR1) in host defense and disease development was determined in a model of viral-induced neurologic disease. Intracerebral (IC) infection of mice with mouse hepatitis virus (MHV) results in an acute encephalitis followed by a chronic demyelinating disease similar in pathology to the disease multiple sclerosis (MS). No increase in mortality was observed during the acute phase of disease following MHV infection of mice lacking CCR1 (CCR1-/-) as compared to wild-type (CCR1+/+) mice. However, by 21 d post-infection, 74% of CCR1-/- mice had succumbed to death compared to only 32% mortality of CCR1+/+ mice, indicating that chemokine signaling through CCR1 significantly (p <or= 0.04) enhanced survival following IC infection with MHV. Increased mortality in CCR1-/- mice was not associated with increased viral recovery from the CNS, although CCR1 deficiency correlated with reduced T-cell accumulation within the CNS during acute, but not chronic, disease. Despite the reduction in T-cell trafficking into the CNS of CCR1-/- mice during acute disease, components of host defense remained unaltered; T-cell effector functions including cytolytic activity and proliferation and the expression of IFN-gamma within the CNS were not significantly different between CCR1+/+ and CCR1-/- infected mice. In addition, macrophage infiltration into the CNS was unaffected in MHV-infected CCR1-/- mice when compared to CCR1+/+ mice. Furthermore, assessment of neuropathology revealed no difference in the severity of demyelination between CCR1-deficient and wild-type mice. Together, these findings reveal that T-cell and macrophage trafficking are not dependent on CCR1 and highlight an important role for CCR1 signaling in promoting survival during chronic MHV infection.
Collapse
Affiliation(s)
- Michelle J Hickey
- Department of Molecular Biology and Biochemistry, Center for Immunology, University of California, Irvine, California 92697-3900, USA
| | | | | | | | | | | |
Collapse
|
35
|
Kallikourdis M, Andersen KG, Welch KA, Betz AG. Alloantigen-enhanced accumulation of CCR5+ 'effector' regulatory T cells in the gravid uterus. Proc Natl Acad Sci U S A 2007; 104:594-9. [PMID: 17197426 PMCID: PMC1766430 DOI: 10.1073/pnas.0604268104] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2006] [Indexed: 01/16/2023] Open
Abstract
Regulatory T cells play an essential role in preventing fetal rejection by the maternal immune system. Here we show that, based on the expression of CCR5, regulatory T cells can be divided into a highly suppressive CCR5+ and a far less suppressive CCR5- subpopulation, suggesting that the former represent the effector arm of regulatory T cells. Although regulatory T cells from CCR5-/- gene deletion mutants still suppress, they are less effective mediators of maternal-fetal tolerance. The accumulation of CCR5+ regulatory T cells at this site appears to be enhanced by alloantigen. This finding is in stark contrast to the systemic expansion of regulatory T cells during pregnancy, which appears to be alloantigen-independent. The fact that CCR5+ regulatory T cells preferentially accumulate in the gravid uterus and that expression of CCR5 on regulatory T cells can be induced by activation lead us to propose that CCR5 is responsible for the accumulation of those regulatory T cells that have been activated by paternal antigens.
Collapse
MESH Headings
- Animals
- Chemokine CCL4
- Chemokines/genetics
- Chemokines, CC/metabolism
- Female
- Gene Expression
- Immune Tolerance
- Isoantigens/metabolism
- Lymphocyte Activation
- Male
- Maternal-Fetal Exchange/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Models, Immunological
- Pregnancy
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, CCR5/deficiency
- Receptors, CCR5/genetics
- Receptors, CCR5/metabolism
- T-Lymphocytes, Regulatory/cytology
- T-Lymphocytes, Regulatory/immunology
- Uterus/cytology
- Uterus/immunology
Collapse
Affiliation(s)
- Marinos Kallikourdis
- Laboratory of Molecular Biology, Medical Research Council, Hills Road, Cambridge CB2 2QH, United Kingdom
| | - Kristian G. Andersen
- Laboratory of Molecular Biology, Medical Research Council, Hills Road, Cambridge CB2 2QH, United Kingdom
| | - Katie A. Welch
- Laboratory of Molecular Biology, Medical Research Council, Hills Road, Cambridge CB2 2QH, United Kingdom
| | - Alexander G. Betz
- Laboratory of Molecular Biology, Medical Research Council, Hills Road, Cambridge CB2 2QH, United Kingdom
| |
Collapse
|
36
|
Hsieh MF, Lai SL, Chen JP, Sung JM, Lin YL, Wu-Hsieh BA, Gerard C, Luster A, Liao F. Both CXCR3 and CXCL10/IFN-inducible protein 10 are required for resistance to primary infection by dengue virus. THE JOURNAL OF IMMUNOLOGY 2006; 177:1855-63. [PMID: 16849497 DOI: 10.4049/jimmunol.177.3.1855] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We examined the extent to which CXCR3 mediates resistance to dengue infection. Following intracerebral infection with dengue virus, CXCR3-deficient (CXCR3(-/-)) mice showed significantly higher mortality rates than wild-type (WT) mice; moreover, surviving CXCR3(-/-) mice, but not WT mice, often developed severe hind-limb paralysis. The brains of CXCR3(-/-) mice showed higher viral loads than those of WT mice, and quantitative analysis using real-time PCR, flow cytometry, and immunohistochemistry revealed fewer T cells, CD8(+) T cells in particular, in the brains of CXCR3(-/-) mice. This suggests that recruitment of effector T cells to sites of dengue infection was diminished in CXCR3(-/-) mice, which impaired elimination of the virus from the brain and thus increased the likelihood of paralysis and/or death. These results indicate that CXCR3 plays a protective rather than an immunopathological role in dengue virus infection. In studies to identify critical CXCR3 ligands, CXCL10/IFN-inducible protein 10-deficient (CXCL10/IP-10(-/-)) mice infected with dengue virus showed a higher mortality rate than that of the CXCR3(-/-) mice. Although CXCL10/IP-10, CXCL9/monokine induced by IFN-gamma, and CXCL11/IFN-inducible T cell alpha chemoattractant share a single receptor and all three of these chemokines are induced by dengue virus infection, the latter two could not compensate for the absence of CXCL10/IP-10 in this in vivo model. Our results suggest that both CXCR3 and CXCL10/IP-10 contribute to resistance against primary dengue virus infection and that chemokines that are indistinguishable in in vitro assays differ in their activities in vivo.
Collapse
MESH Headings
- Animals
- Brain/immunology
- Brain/metabolism
- Brain/virology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/pathology
- CD4-Positive T-Lymphocytes/virology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- CD8-Positive T-Lymphocytes/virology
- Cell Migration Inhibition
- Cell Movement/genetics
- Cell Movement/immunology
- Chemokine CXCL10
- Chemokines, CXC/deficiency
- Chemokines, CXC/genetics
- Chemokines, CXC/physiology
- Dengue/genetics
- Dengue/immunology
- Dengue/virology
- Dengue Virus/immunology
- Genetic Predisposition to Disease
- Immunity, Innate/genetics
- Inflammation Mediators/metabolism
- Injections, Intraventricular
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, CXCR3
- Receptors, Chemokine/biosynthesis
- Receptors, Chemokine/deficiency
- Receptors, Chemokine/genetics
- Receptors, Chemokine/physiology
- Up-Regulation/immunology
- Viral Load
Collapse
Affiliation(s)
- Ming-Fang Hsieh
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Barcia C, Thomas CE, Curtin JF, King GD, Wawrowsky K, Candolfi M, Xiong WD, Liu C, Kroeger K, Boyer O, Kupiec-Weglinski J, Klatzmann D, Castro MG, Lowenstein PR. In vivo mature immunological synapses forming SMACs mediate clearance of virally infected astrocytes from the brain. J Exp Med 2006; 203:2095-107. [PMID: 16923851 PMCID: PMC1997281 DOI: 10.1084/jem.20060420] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2006] [Accepted: 07/14/2006] [Indexed: 01/21/2023] Open
Abstract
The microanatomy of immune clearance of infected brain cells remains poorly understood. Immunological synapses are essential anatomical structures that channel information exchanges between T cell-antigen-presenting cells (APC) during the priming and effector phases of T cells' function, and during natural killer-target cell interactions. The hallmark of immunological synapses established by T cells is the formation of the supramolecular activation clusters (SMACs), in which adhesion molecules such as leukocyte function-associated antigen 1 segregate to the peripheral domain of the immunological synapse (p-SMAC), which surrounds the T cell receptor-rich or central SMAC (c-SMAC). The inability so far to detect SMAC formation in vivo has cast doubts on its functional relevance. Herein, we demonstrate that the in vivo formation of SMAC at immunological synapses between effector CD8+ T cells and target cells precedes and mediates clearance of virally infected brain astrocytes.
Collapse
Affiliation(s)
- Carlos Barcia
- Board of Governors' Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Stiles LN, Hosking MP, Edwards RA, Strieter RM, Lane TE. Differential roles for CXCR3 in CD4+ and CD8+ T cell trafficking following viral infection of the CNS. Eur J Immunol 2006; 36:613-22. [PMID: 16479546 DOI: 10.1002/eji.200535509] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Lymphocyte infiltration into the central nervous system (CNS) following viral infection represents an important component of host defense and is required for control of viral replication. However, the mechanisms governing inflammation in response to viral infection of the CNS are not well understood. Following intracranial (i.c.) infection of susceptible mice with mouse hepatitis virus (MHV), mice develop an acute encephalomyelitis followed by a chronic demyelinating disease. The CXC chemokine ligand 10 (CXCL10) is expressed following MHV infection and signals T cells to migrate into the CNS. The functional contribution of the CXCL10 receptor CXCR3 in host defense and disease in response to MHV infection was evaluated. The majority of CD4+ and CD8+ T cells infiltrating the CNS following MHV infection express CXCR3. Administration of anti-CXCR3 antibody reduced CD4+ T cell infiltration (p<or=0.05), while CD8+ T cell trafficking was not affected. Anti-CXCR3 treatment during chronic disease correlated with improved motor skills and reduced demyelination. The selective effect of anti-CXCR3 treatment on CD4+ T cells was not the result of either reduced proliferation or modulation in chemokine receptor gene expression. Therefore, CXCR3 signaling has a non-redundant role in T cell subset trafficking in response to viral infection.
Collapse
Affiliation(s)
- Linda N Stiles
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697-3900, USA
| | | | | | | | | |
Collapse
|
39
|
Carr DJ, Ash J, Lane TE, Kuziel WA. Abnormal immune response of CCR5-deficient mice to ocular infection with herpes simplex virus type 1. J Gen Virol 2006; 87:489-499. [PMID: 16476970 PMCID: PMC1479868 DOI: 10.1099/vir.0.81339-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Ocular herpes simplex virus type 1 (HSV-1) infection elicits a strong inflammatory response that is associated with production of the beta chemokines CCL3 and CCL5, which share a common receptor, CCR5. To gain insight into the role of these molecules in ocular immune responses, the corneas of wild-type (WT) and CCR5-deficient (CCR5-/-) mice were infected with HSV-1 and inflammatory parameters were measured. In the absence of CCR5, the early infiltration of neutrophils into the cornea was diminished. Associated with this aberrant leukocyte recruitment, neutrophils in CCR5-/- mice were restricted to the stroma, whereas in WT mice, these cells trafficked to the stroma and epithelial layers of the infected cornea. Virus titres and cytokine/chemokine levels in the infected tissue of these mice were similar for the first 5 days after infection. However, by day 7 post-infection, the CCR5-/- mice showed a significant elevation in the chemokines CCL2, CCL5, CXCL9 and CXCL10 in the trigeminal ganglion and brainstem, as well as a significant increase in virus burden. The increase in chemokine expression was associated with an increase in the infiltration of CD4 and/or CD8 T cells into the trigeminal ganglion and brainstem of CCR5-/- mice. Surprisingly, even though infected CCR5-/- mice were less efficient at controlling the progression of virus replication, there was no difference in mortality. These results suggest that, although CCR5 plays a role in regulating leukocyte trafficking and control of virus burden, compensatory mechanisms are involved in preventing mortality following HSV-1 infection.
Collapse
Affiliation(s)
- Daniel J.J. Carr
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
- Microbiology, and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - John Ash
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Thomas E. Lane
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92037
| | | |
Collapse
|
40
|
Lane TE, Hardison JL, Walsh KB. Functional diversity of chemokines and chemokine receptors in response to viral infection of the central nervous system. Curr Top Microbiol Immunol 2006; 303:1-27. [PMID: 16570854 PMCID: PMC7121733 DOI: 10.1007/978-3-540-33397-5_1] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Encounters with neurotropic viruses result in varied outcomes ranging from encephalitis, paralytic poliomyelitis or other serious consequences to relatively benign infection. One of the principal factors that control the outcome of infection is the localized tissue response and subsequent immune response directed against the invading toxic agent. It is the role of the immune system to contain and control the spread of virus infection in the central nervous system (CNS), and paradoxically, this response may also be pathologic. Chemokines are potent proinflammatory molecules whose expression within virally infected tissues is often associated with protection and/or pathology which correlates with migration and accumulation of immune cells. Indeed, studies with a neurotropic murine coronavirus, mouse hepatitis virus (MHV), have provided important insight into the functional roles of chemokines and chemokine receptors in participating in various aspects of host defense as well as disease development within the CNS. This chapter will highlight recent discoveries that have provided insight into the diverse biologic roles of chemokines and their receptors in coordinating immune responses following viral infection of the CNS.
Collapse
Affiliation(s)
- T E Lane
- Department of Molecular Biology and Biochemistry, University of California, 3205 McGaugh Hall, Irvine, CA 92697-3900, USA.
| | | | | |
Collapse
|
41
|
Hardison JL, Wrightsman RA, Carpenter PM, Kuziel WA, Lane TE, Manning JE. The CC chemokine receptor 5 is important in control of parasite replication and acute cardiac inflammation following infection with Trypanosoma cruzi. Infect Immun 2006; 74:135-43. [PMID: 16368966 PMCID: PMC1346647 DOI: 10.1128/iai.74.1.135-143.2006] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2005] [Revised: 09/13/2005] [Accepted: 09/28/2005] [Indexed: 11/20/2022] Open
Abstract
Infection of susceptible mice with the Colombiana strain of Trypanosoma cruzi results in an orchestrated expression of chemokines and chemokine receptors within the heart that coincides with parasite burden and cellular infiltration. CC chemokine receptor 5 (CCR5) is prominently expressed during both acute and chronic disease, suggesting a role in regulating leukocyte trafficking and accumulation within the heart following T. cruzi infection. To better understand the functional role of CCR5 and its ligands with regard to both host defense and/or disease, CCR5(-/-) mice were infected with T. cruzi, and the disease severity was evaluated. Infected CCR5(-/-) mice develop significantly higher levels of parasitemia (P < or = 0.05) and cardiac parasitism (P < or = 0.01) during acute infection that correlated with reduced survival. Further, we show that CCR5 is essential for directing the migration of macrophages and T cells to the heart early in acute infection with T. cruzi. In addition, data are provided demonstrating that CCR5 does not play an essential role in maintaining inflammation in the heart during chronic infection. Collectively, these studies clearly demonstrate that CCR5 contributes to the control of parasite replication and the development of a protective immune response during acute infection but does not ultimately participate in maintaining a chronic inflammatory response within the heart.
Collapse
Affiliation(s)
- Jenny L Hardison
- Department of Molecular Biology and Biochemistry, 3205 McGaugh Hall, University of California-Irvine, Irvine, CA 92697-3900, USA
| | | | | | | | | | | |
Collapse
|
42
|
Dandekar AA, O'Malley K, Perlman S. Important roles for gamma interferon and NKG2D in gammadelta T-cell-induced demyelination in T-cell receptor beta-deficient mice infected with a coronavirus. J Virol 2005; 79:9388-96. [PMID: 16014902 PMCID: PMC1181615 DOI: 10.1128/jvi.79.15.9388-9396.2005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
gammadelta T cells mediate demyelination in athymic (nude) mice infected with the neurotropic coronavirus mouse hepatitis virus strain JHM. Now, we show that these cells also mediate the same process in mice lacking alphabeta T cells (T-cell receptor beta-deficient [TCRbeta(-/-)] mice) and demyelination is gamma interferon (IFN-gamma) dependent. Most strikingly, our results also show a major role for NKG2D, expressed on gammadelta T cells, in the demyelinating process with in vivo blockade of NKG2D interactions resulting in a 60% reduction in demyelination. NKG2D may serve as a primary recognition receptor or as a costimulatory molecule. We show that NKG2D(+) gammadelta T cells in the JHM-infected central nervous system express the adaptor molecule DAP12 and an NKG2D isoform (NKG2D short), both required for NKG2D to serve as a primary receptor. These results are consistent with models in which gammadelta T cells mediate demyelination using the same effector cytokine, IFN-gamma, as CD8 T cells and do so without a requirement for signaling through the TCR.
Collapse
MESH Headings
- Animals
- CD8-Positive T-Lymphocytes/immunology
- Coronavirus
- Coronavirus Infections/immunology
- Coronavirus Infections/pathology
- Demyelinating Diseases/immunology
- Demyelinating Diseases/pathology
- Interferon-gamma/immunology
- Killer Cells, Natural/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- NK Cell Lectin-Like Receptor Subfamily K
- Receptors, Antigen, T-Cell, alpha-beta/deficiency
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, Immunologic/immunology
- Receptors, Natural Killer Cell
- Spinal Cord/pathology
- Spinal Cord/virology
Collapse
Affiliation(s)
- Ajai A Dandekar
- Interdisciplinary Program in Immunology, Unversity of Iowa, Iowa City, 52242, USA
| | | | | |
Collapse
|
43
|
Ure DR, Lane TE, Liu MT, Rodriguez M. Neutralization of chemokines RANTES and MIG increases virus antigen expression and spinal cord pathology during Theiler's virus infection. Int Immunol 2005; 17:569-79. [PMID: 15824069 PMCID: PMC7108597 DOI: 10.1093/intimm/dxh236] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The role of chemokines during some viral infections is unpredictable because the inflammatory response regulated by these molecules can have two, contrasting effects—viral immunity and immunopathologic injury to host tissues. Using Theiler's virus infection of SJL mice as a model of this type of disease, we have investigated the roles of two chemokines—regulated on activation, normal T cell-expressed and secreted (RANTES) chemokine and monokine induced by IFN-γ (MIG)—by treating mice with antisera that block lymphocyte migration. Control, infected mice showed virus persistence, mild inflammation and a small degree of demyelination in the white matter of the spinal cord at 6 weeks post-infection. Treatment of mice with RANTES antiserum starting at 2 weeks post-infection increased both viral antigen expression and the severity of inflammatory demyelination at 6 weeks post-infection. MIG antiserum increased the spread of virus and the proportion of spinal cord white matter with demyelination. Overall, viral antigen levels correlated strongly with the extent of pathology. At the RNA level, high virus expression was associated with low IL-2 and high IL-10 levels, and RANTES antiserum decreased the IL-2/IL-10 ratio. Our results suggest that RANTES and MIG participate in an immune response that attempts to restrict viral expression while limiting immunopathology and that anti-chemokine treatment poses the risk of exacerbating both conditions in the long term.
Collapse
|
44
|
Abstract
Highly active antiretroviral therapy (HAART) has led to major declines in morbidity and mortality of HIV-1-infected individuals, but the increasing prevalence of drug-resistant viral isolates, combined with the toxicity and other limitations of current treatments, make the development of new therapies a high priority. As knowledge of viral entry has expanded, this step of the viral life cycle has become a target for novel therapeutic strategies. An emerging group of antiretrovirals, known collectively as entry inhibitors, targets several distinct steps in viral entry including CD4 binding, chemokine receptor engagement and the structural changes in the viral envelope required for fusion between viral and cellular membranes. Many entry inhibitors are in various stages of clinical development, with one already licensed for use. This review will provide an overview of the mechanisms involved in the entry process, highlight promising entry blockers under development and discuss several considerations related to treatment that are unique to this class of antiretroviral drugs.
Collapse
Affiliation(s)
- Brian Tomkowicz
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia 19104, USA
| | | |
Collapse
|
45
|
Klein RS. Regulation of neuroinflammation: the role of CXCL10 in lymphocyte infiltration during autoimmune encephalomyelitis. J Cell Biochem 2005; 92:213-22. [PMID: 15108349 DOI: 10.1002/jcb.20052] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The movement of lymphocytes from the microvasculature into the central nervous system (CNS) parenchyma is an essential step in the pathogenesis of a variety of infectious and autoimmune neuroinflammatory diseases. The lymphocyte chemoattractant CXCL10 and its receptor, CXCR3, are expressed by the CNS and by CNS infiltrating lymphocytes, respectively, only in patients with ongoing CNS inflammation, suggesting an important role for these molecules in the pathogenic process. Numerous studies utilizing animal models and transgenic approaches have indeed supported a role for CXCL10 in the intraparenchymal trafficking of lymphocytes during acute CNS inflammation; however, other studies suggest that its expression is not required for the development of autoimmune forms of CNS inflammation and, in fact, that interference with CXCL10 signaling could lead to increased neuroinflammation. This review will consider the data from these studies and attempt to reconcile them through comparisons of both the neuroinflammatory models and the effects of CXCL10 in the CNS versus lymphoid tissues. Finally, it will define directions for future analyses of CXCL10 and CXCR3 in CNS inflammation so that their potential therapeutic utility can be more completely determined.
Collapse
Affiliation(s)
- Robyn S Klein
- Department Internal Medicine, Washington University School of Medicine, St. Louis, Missouri 63119, USA.
| |
Collapse
|
46
|
Trifilo MJ, Liu MT, Glass WG, Lane TE. Chemokines in Coronavirus-Induced Demyelination. EXPERIMENTAL MODELS OF MULTIPLE SCLEROSIS 2005. [PMCID: PMC7122598 DOI: 10.1007/0-387-25518-4_45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Inflammation within the central nervous system (CNS) is critical in the development of the neuropathology associated with the human demyelinating disease multiple sclerosis (MS). Recent studies have identified a family of soluble proinflammatory molecules called chemokines that are able to direct leukocyte infiltration into the CNS in response to infection or injury. Identification of chemokines within and around demyelinating lesions in MS patients indicate a potential role for these molecules in contributing to the pathogenesis of MS. To address this issue, we have used mouse hepatitis virus (MHV) infection of the CNS to understand the dynamic interaction of chemokine expression as it relates to inflammation and neuropathology. Our results indicate that chemokine expression within the CNS results in persistent recruitment of both T lymphocytes and macrophages and results in subsequent myelin destruction. Herein, we demonstrate the complexity of the chemokine response to MHV infection of the CNS and the delicate balance that exists between host defense and development of disease.
Collapse
|
47
|
Dave RS, Pomerantz RJ. Antiviral effects of human immunodeficiency virus type 1-specific small interfering RNAs against targets conserved in select neurotropic viral strains. J Virol 2004; 78:13687-96. [PMID: 15564478 PMCID: PMC533941 DOI: 10.1128/jvi.78.24.13687-13696.2004] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
RNA interference, a natural biological phenomenon mediated by small interfering RNAs (siRNAs), has been demonstrated in recent studies to be an effective strategy against human immunodeficiency virus type 1 (HIV-1). In the present study, we used 21-bp chemically synthesized siRNA duplexes whose sequences were derived from the gp41 gene, nef, tat, and rev regions of viral RNA. These sequences are conserved in select neurotropic strains of HIV-1 (JR-FL, JR-CSF, and YU-2). The designed siRNAs exerted a potent antiviral effect on these HIV-1 strains. The antiviral effect was mediated at the RNA level (as observed by the down-regulation of the HIV-1-specific spliced transcript generating a 1.2-kbp reverse transcription [RT]-PCR product) as well as viral assembly on the cell membrane. Spliced transcripts (apart from the most abundant transcript generating a 1.2-kbp RT-PCR product) arising from an unspliced precursor likely contributed, albeit to a lesser extent, to the antiviral effect. The resultant progeny viruses had infectivities similar to that of input virus. We therefore conclude that these siRNAs interfere with the processing of the unspliced transcripts for the gp41 gene, tat, rev, and nef, eventually affecting viral assembly and leading to the overall inhibition of viral production. Apart from using the gp41 gene as a target, the conservation of each of these targets in the above-mentioned viral strains, as well as several primary isolates, would enable these siRNAs to be used as potent antiviral tools for investigations with cells derived from the central nervous system in order to evaluate their therapeutic potential and assess their utility in inhibiting HIV-1 neuropathogenesis and neuroinvasion.
Collapse
MESH Headings
- Antiviral Agents/pharmacology
- Base Sequence
- Brain/virology
- Cells, Cultured
- Cerebrospinal Fluid/virology
- Gene Products, nef/chemistry
- Gene Products, nef/genetics
- Gene Products, nef/metabolism
- Gene Products, rev/chemistry
- Gene Products, rev/genetics
- Gene Products, rev/metabolism
- Gene Products, tat/chemistry
- Gene Products, tat/genetics
- Gene Products, tat/metabolism
- HIV Envelope Protein gp41/chemistry
- HIV Envelope Protein gp41/genetics
- HIV Envelope Protein gp41/metabolism
- HIV-1/chemistry
- HIV-1/genetics
- HIV-1/metabolism
- HeLa Cells
- Humans
- Macrophages/virology
- Monocytes/virology
- RNA Interference
- RNA, Small Interfering/chemistry
- RNA, Small Interfering/genetics
- RNA, Small Interfering/pharmacology
- nef Gene Products, Human Immunodeficiency Virus
- rev Gene Products, Human Immunodeficiency Virus
- tat Gene Products, Human Immunodeficiency Virus
Collapse
Affiliation(s)
- Rajnish S Dave
- Dorrance H. Hamilton Laboratories, Center for Human Virology and Biodefense, Division of Infectious Diseases and Environmental Medicine, Department of Medicine, Thomas Jefferson University, 1020 Locust St., Suite 329, Philadelphia, PA 19107, USA
| | | |
Collapse
|
48
|
Held KS, Chen BP, Kuziel WA, Rollins BJ, Lane TE. Differential roles of CCL2 and CCR2 in host defense to coronavirus infection. Virology 2004; 329:251-60. [PMID: 15518805 PMCID: PMC7111831 DOI: 10.1016/j.virol.2004.09.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2004] [Revised: 05/21/2004] [Accepted: 09/08/2004] [Indexed: 11/17/2022]
Abstract
The CC chemokine ligand 2 (CCL2, monocyte chemoattractant protein-1) is important in coordinating the immune response following microbial infection by regulating T cell polarization as well as leukocyte migration and accumulation within infected tissues. The present study examines the consequences of mouse hepatitis virus (MHV) infection in mice lacking CCL2 (CCL2(-/-)) in order to determine if signaling by this chemokine is relevant in host defense. Intracerebral infection of CCL2(-/-) mice with MHV did not result in increased morbidity or mortality as compared to either wild type or CCR2(-/-) mice and CCL2(-/-) mice cleared replicating virus from the brain. In contrast, CCR2(-/-) mice displayed an impaired ability to clear virus from the brain that was accompanied by a reduction in the numbers of antigen-specific T cells as compared to both CCL2(-/-) and wild-type mice. The paucity in T cell accumulation within the central nervous system (CNS) of MHV-infected CCR2(-/-) mice was not the result of either a deficiency in antigen-presenting cell (APC) accumulation within draining cervical lymph nodes (CLN) or the generation of virus-specific T cells within this compartment. A similar reduction in macrophage infiltration into the CNS was observed in both CCL2(-/-) and CCR2(-/-) mice when compared to wild-type mice, indicating that both CCL2 and CC chemokine receptor 2 (CCR2) contribute to macrophage migration and accumulation within the CNS following MHV infection. Together, these data demonstrate that CCR2, but not CCL2, is important in host defense following viral infection of the CNS, and CCR2 ligand(s), other than CCL2, participates in generating a protective response.
Collapse
Affiliation(s)
- Katherine S. Held
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, United States
| | - Benjamin P. Chen
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, United States
| | - William A. Kuziel
- Department of Molecular Genetics and Microbiology, Institute for Cellular and Molecular Biology, University of Texas, Austin, TX 78712, United States
| | - Barrett J. Rollins
- Department of Medicine, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, United States
| | - Thomas E. Lane
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, United States
- Center for Immunology, University of California, Irvine, CA 92697, United States
| |
Collapse
|
49
|
Glass WG, Rosenberg HF, Murphy PM. Chemokine regulation of inflammation during acute viral infection. Curr Opin Allergy Clin Immunol 2004; 3:467-73. [PMID: 14612671 DOI: 10.1097/00130832-200312000-00008] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Chemokines are important inflammatory mediators, and regulate disease due to viral infection. This article will discuss scientific papers published primarily since June 2002 that have introduced new concepts in how chemokines regulate the inflammatory response to specific viruses. RECENT FINDINGS Acute respiratory viruses commonly induce inflammatory chemokines such as CCL3 (also known as macrophage inflammatory protein-1alpha) and CCL5 (RANTES), which can amplify inflammatory responses leading to immunopathology. Where single agent therapy fails, combination antiviral and anti-CCL3 treatment is synergistic and able to prevent mortality in mice infected with the highly lethal pneumonia virus of mice. Human herpesvirus-6 also induces production of CCL3 and CCL5, which are able to block HIV-1 replication in coinfected human lymphoid tissue. On this basis, Margolis has proposed a new and general approach to the treatment and prevention of infection by viral pathogens. SUMMARY Inflammatory chemokines play both beneficial and harmful roles in infectious diseases caused by viruses. Blocking them or using them as immunomodulators, depending on the virus, may be rational approaches to treatment or prevention of disease. With regard to blockade, combination antiviral/antichemokine therapy is a new strategy worth considering as a general therapeutic approach to viral infections, including severe acute respiratory syndrome (SARS). With regard to immunomodulation, use of weak or attenuated viruses to skew the local cytokine network to a configuration able to inhibit a pathogen is a new and interesting concept, but is fraught with important safety issues. Identifying master chemokines to target or exploit in human viral infection is a major opportunity and challenge for clinical immunologists.
Collapse
Affiliation(s)
- William G Glass
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | |
Collapse
|
50
|
Glass WG, Hickey MJ, Hardison JL, Liu MT, Manning JE, Lane TE. Antibody targeting of the CC chemokine ligand 5 results in diminished leukocyte infiltration into the central nervous system and reduced neurologic disease in a viral model of multiple sclerosis. THE JOURNAL OF IMMUNOLOGY 2004; 172:4018-25. [PMID: 15034013 DOI: 10.4049/jimmunol.172.7.4018] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Intracerebral infection of mice with mouse hepatitis virus, a member of the Coronaviridae family, reproducibly results in an acute encephalomyelitis that progresses to a chronic demyelinating disease. The ensuing neuropathology during the chronic stage of disease is primarily immune mediated and similar to that of the human demyelinating disease multiple sclerosis. Secretion of chemokines within the CNS signals the infiltration of leukocytes, which results in destruction of white matter and neurological impairment. The CC chemokine ligand (CCL)5 is localized in white matter tracts undergoing demyelination, suggesting that this chemokine participates in the pathogenesis of disease by attracting inflammatory cells into the CNS. In this study, we administer a mAb directed against CCL5 to mice with established mouse hepatitis virus-induced demyelination and impaired motor skills. Anti-CCL5 treatment decreased T cell accumulation within the CNS based, in part, on viral Ag specificity, indicating the ability to differentially target select populations of T cells. In addition, administration of anti-CCL5 improved neurological function and significantly (p < or = 0.005) reduced the severity of demyelination and macrophage accumulation within the CNS. These results demonstrate that the severity of CNS disease can be reduced through the use of a neutralizing mAb directed against CCL5 in a viral model of demyelination.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/therapeutic use
- Cell Migration Inhibition
- Central Nervous System/immunology
- Central Nervous System/pathology
- Central Nervous System/virology
- Chemokine CCL5/antagonists & inhibitors
- Chemokine CCL5/biosynthesis
- Chemokine CCL5/genetics
- Chemokine CCL5/immunology
- Chemokines, CC/immunology
- Chemokines, CC/metabolism
- Chemotaxis, Leukocyte/immunology
- Coronavirus Infections/immunology
- Coronavirus Infections/pathology
- Coronavirus Infections/therapy
- Demyelinating Diseases/immunology
- Demyelinating Diseases/pathology
- Demyelinating Diseases/prevention & control
- Disease Models, Animal
- Encephalomyelitis/immunology
- Encephalomyelitis/pathology
- Encephalomyelitis/therapy
- Injections, Intraperitoneal
- Ligands
- Macrophage Activation/immunology
- Macrophages/immunology
- Macrophages/pathology
- Mice
- Mice, Inbred C57BL
- Multiple Sclerosis/immunology
- Multiple Sclerosis/pathology
- Multiple Sclerosis/prevention & control
- Murine hepatitis virus/immunology
- RNA, Messenger/antagonists & inhibitors
- RNA, Messenger/biosynthesis
- Severity of Illness Index
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/pathology
- T-Lymphocyte Subsets/virology
Collapse
Affiliation(s)
- William G Glass
- Center for Immunology and Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, USA
| | | | | | | | | | | |
Collapse
|