1
|
Kumar S, Kahle AD, Keeler AB, Zunder ER, Deppmann CD. Characterizing Microglial Signaling Dynamics During Inflammation Using Single-Cell Mass Cytometry. Glia 2025; 73:1022-1035. [PMID: 39780484 PMCID: PMC11920681 DOI: 10.1002/glia.24670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 12/12/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025]
Abstract
Microglia play a critical role in maintaining central nervous system (CNS) homeostasis and display remarkable plasticity in their response to inflammatory stimuli. However, the specific signaling profiles that microglia adopt during such challenges remain incompletely understood. Traditional transcriptomic approaches provide valuable insights, but fail to capture dynamic post-translational changes. In this study, we utilized time-resolved single-cell mass cytometry (CyTOF) to measure distinct signaling pathways activated in microglia upon exposure to bacterial and viral mimetics-lipopolysaccharide (LPS) and polyinosinic-polycytidylic acid (Poly(I:C)), respectively. Furthermore, we evaluated the immunomodulatory role of astrocytes on microglial signaling in mixed cultures. Microglia or mixed cultures derived from neonatal mice were treated with LPS or Poly(I:C) for 48 h. Cultures were stained with a panel of 33 metal-conjugated antibodies targeting signaling and identity markers. High-dimensional clustering analysis was used to identify emergent signaling modules. We found that LPS treatment led to more robust early activation of pp38, pERK, pRSK, and pCREB compared to Poly(I:C). Despite these differences, both LPS and Poly(I:C) upregulated the classical reactivity markers CD40 and CD86 at later time points. Strikingly, the presence of astrocytes significantly blunted microglial responses to both stimuli, particularly dampening CD40 upregulation. Our studies demonstrate that single-cell mass cytometry effectively captures the dynamic signaling landscape of microglia under pro-inflammatory conditions. This approach may pave the way for targeted therapeutic investigations of various neuroinflammatory disorders. Moreover, our findings underscore the necessity of considering cellular context, such as astrocyte presence, in interpreting microglial behavior during inflammation.
Collapse
Affiliation(s)
- Sushanth Kumar
- Department of Biology, College of Arts and SciencesUniversity of VirginiaCharlottesvilleVirginiaUSA
- Neuroscience Graduate Program, School of MedicineUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - August D. Kahle
- Department of Biology, College of Arts and SciencesUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Austin B. Keeler
- Department of Biology, College of Arts and SciencesUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Eli R. Zunder
- Department of Biomedical Engineering, School of EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
- Program in Fundamental Neuroscience, College of Arts and SciencesUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Christopher D. Deppmann
- Department of Biology, College of Arts and SciencesUniversity of VirginiaCharlottesvilleVirginiaUSA
- Neuroscience Graduate Program, School of MedicineUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Biomedical Engineering, School of EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
- Program in Fundamental Neuroscience, College of Arts and SciencesUniversity of VirginiaCharlottesvilleVirginiaUSA
| |
Collapse
|
2
|
Zoladek J, Cannac M, Seite M, Davies E, Quellec J, Barthelemy J, Gorna K, Desgraupes S, Bribes I, Salinas S, Coulpier M, Arhel NJ, Palmarini M, Simonin Y, Wilson SJ, Nisole S. MITD1 is a brain-specific interferon-inducible factor that inhibits flavivirus replication. Proc Natl Acad Sci U S A 2025; 122:e2502064122. [PMID: 40112111 PMCID: PMC11962514 DOI: 10.1073/pnas.2502064122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 02/19/2025] [Indexed: 03/22/2025] Open
Abstract
West Nile virus (WNV) and Usutu virus (USUV) are closely related mosquito-borne neurotropic flaviviruses that share common transmission cycle and can infect humans. However, while human infections by WNV are widespread, infections by USUV are comparatively less frequent, less severe, and currently limited to Africa and Europe. To identify human host factors that contribute to the pathogenic signatures of these two flaviviruses, we carried out an arrayed expression screen of over 1,300 interferon-stimulated genes (ISGs). Several ISGs known to target flaviviruses, including IFI6, SHFL, and RTP4 were among the strongest hits. Interestingly, we also found MITD1, an ISG with no previously reported antiviral activity, among the strongest hits. We demonstrated that the antiviral activity of MITD1 was not limited to USUV and WNV, since it also inhibited Zika and dengue virus replication. We found MITD1 to interfere with viral RNA replication by sequestering specific endosomal sorting complexes required for transport-III (ESCRT-III) proteins involved in the formation of viral replication factories. MITD1 expression was not increased by type I interferon (IFN-I) in most human cells and mouse tissues that we examined, although WNV and USUV replication was strongly inhibited by IFN-I. Strikingly, MITD1 was induced in the brain of USUV-infected mice and importantly, in human monocyte-derived microglia. Using human microglial-like cells, we confirmed that MITD1 is an essential mediator of the anti-flavivirus activity of IFN-I in these cells. We conclude that MITD1 plays a key role in the cellular defenses against neurotropic flaviviruses.
Collapse
Affiliation(s)
- Jim Zoladek
- Viral Trafficking, Restriction and Innate Signaling, CNRS UMR 9004, Institut de Recherche en Infectiologie de Montpellier, Université de Montpellier, Montpellier34090, France
| | - Marion Cannac
- Viral Trafficking, Restriction and Innate Signaling, CNRS UMR 9004, Institut de Recherche en Infectiologie de Montpellier, Université de Montpellier, Montpellier34090, France
- Pathogenesis and Control of Chronic and Emerging Infections, INSERM, Établissement français du sang (EFS), Université de Montpellier, Montpellier34394, France
| | - Maël Seite
- Viral Trafficking, Restriction and Innate Signaling, CNRS UMR 9004, Institut de Recherche en Infectiologie de Montpellier, Université de Montpellier, Montpellier34090, France
| | - Emma Davies
- Medical Research Council-University of Glasgow Centre for Virus Research, GlasgowG61 1QH, United Kingdom
| | - Jordan Quellec
- Pathogenesis and Control of Chronic and Emerging Infections, INSERM, Établissement français du sang (EFS), Université de Montpellier, Montpellier34394, France
- Animal, Santé, Territoires, Risques et Ecosystèmes, UMR 117, Centre de coopération internationale en recherche agronomique pour le développement (CIRAD), Institut national de recherche pour l'agriculture, l'alimentation et l'environnement (INRAE), Université de Montpellier, Montpellier34398, France
| | - Jonathan Barthelemy
- Pathogenesis and Control of Chronic and Emerging Infections, INSERM, Établissement français du sang (EFS), Université de Montpellier, Montpellier34394, France
| | - Kamila Gorna
- UMR Virologie, Laboratoire de Santé Animale, Institut national de recherche pour l'agriculture, l'alimentation et l'environnement (INRAE), Anses, Ecole Nationale Vétérinaire d’Alfort, Maisons-Alfort94700, France
| | - Sophie Desgraupes
- Viral Trafficking, Restriction and Innate Signaling, CNRS UMR 9004, Institut de Recherche en Infectiologie de Montpellier, Université de Montpellier, Montpellier34090, France
| | - Ines Bribes
- Viral Trafficking, Restriction and Innate Signaling, CNRS UMR 9004, Institut de Recherche en Infectiologie de Montpellier, Université de Montpellier, Montpellier34090, France
| | - Sara Salinas
- Pathogenesis and Control of Chronic and Emerging Infections, INSERM, Établissement français du sang (EFS), Université de Montpellier, Montpellier34394, France
| | - Muriel Coulpier
- UMR Virologie, Laboratoire de Santé Animale, Institut national de recherche pour l'agriculture, l'alimentation et l'environnement (INRAE), Anses, Ecole Nationale Vétérinaire d’Alfort, Maisons-Alfort94700, France
| | - Nathalie J. Arhel
- Viral Trafficking, Restriction and Innate Signaling, CNRS UMR 9004, Institut de Recherche en Infectiologie de Montpellier, Université de Montpellier, Montpellier34090, France
| | - Massimo Palmarini
- Medical Research Council-University of Glasgow Centre for Virus Research, GlasgowG61 1QH, United Kingdom
| | - Yannick Simonin
- Pathogenesis and Control of Chronic and Emerging Infections, INSERM, Établissement français du sang (EFS), Université de Montpellier, Montpellier34394, France
| | - Sam J. Wilson
- Medical Research Council-University of Glasgow Centre for Virus Research, GlasgowG61 1QH, United Kingdom
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Department of Medicine, University of Cambridge, CambridgeCB2 0AW, United Kingdom
| | - Sébastien Nisole
- Viral Trafficking, Restriction and Innate Signaling, CNRS UMR 9004, Institut de Recherche en Infectiologie de Montpellier, Université de Montpellier, Montpellier34090, France
| |
Collapse
|
3
|
León-Rodríguez A, Grondona JM, Marín-Wong S, López-Aranda MF, López-Ávalos MD. Long-term reprogramming of primed microglia after moderate inhibition of CSF1R signaling. Glia 2025; 73:175-195. [PMID: 39448548 DOI: 10.1002/glia.24627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 10/26/2024]
Abstract
In acute neuroinflammation, microglia activate transiently, and return to a resting state later on. However, they may retain immune memory of such event, namely priming. Primed microglia are more sensitive to new stimuli and develop exacerbated responses, representing a risk factor for neurological disorders with an inflammatory component. Strategies to control the hyperactivation of microglia are, hence, of great interest. The receptor for colony stimulating factor 1 (CSF1R), expressed in myeloid cells, is essential for microglia viability, so its blockade with specific inhibitors (e.g. PLX5622) results in significant depletion of microglial population. Interestingly, upon inhibitor withdrawal, new naïve microglia repopulate the brain. Depletion-repopulation has been proposed as a strategy to reprogram microglia. However, substantial elimination of microglia is inadvisable in human therapy. To overcome such drawback, we aimed to reprogram long-term primed microglia by CSF1R partial inhibition. Microglial priming was induced in mice by acute neuroinflammation, provoked by intracerebroventricular injection of neuraminidase. After 3-weeks recovery, low-dose PLX5622 treatment was administrated for 12 days, followed by a withdrawal period of 7 weeks. Twelve hours before euthanasia, mice received a peripheral lipopolysaccharide (LPS) immune challenge, and the subsequent microglial inflammatory response was evaluated. PLX5622 provoked a 40%-50% decrease in microglial population, but basal levels were restored 7 weeks later. In the brain regions studied, hippocampus and hypothalamus, LPS induced enhanced microgliosis and inflammatory activation in neuraminidase-injected mice, while PLX5622 treatment prevented these changes. Our results suggest that PLX5622 used at low doses reverts microglial priming and, remarkably, prevents broad microglial depletion.
Collapse
Affiliation(s)
- Ana León-Rodríguez
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA Plataforma Bionand, Málaga, Spain
| | - Jesús M Grondona
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA Plataforma Bionand, Málaga, Spain
| | - Sonia Marín-Wong
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
| | - Manuel F López-Aranda
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA Plataforma Bionand, Málaga, Spain
| | - María D López-Ávalos
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA Plataforma Bionand, Málaga, Spain
| |
Collapse
|
4
|
Gern OL, Pavlou A, Mulenge F, Busker LM, Ghita L, Aringo A, Costa B, Spanier J, Waltl I, Stangel M, Kalinke U. MAVS signaling shapes microglia responses to neurotropic virus infection. J Neuroinflammation 2024; 21:264. [PMID: 39425188 PMCID: PMC11490141 DOI: 10.1186/s12974-024-03258-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024] Open
Abstract
Viral encephalitis is characterized by a series of immunological reactions that can control virus infection in the brain, but dysregulated responses may cause excessive inflammation and brain damage. Microglia are brain-resident myeloid cells that are specialized in surveilling the local CNS environment and in case of viral brain infection they contribute to the control of the infection and to restriction of viral dissemination. Here, we report that after exposure to neurotropic vesicular stomatitis virus (VSV), murine in vitro microglia cultures showed rapid upregulation of a broad range of pro-inflammatory and antiviral genes, which were stably expressed over the entire 8 h infection period. Additionally, a set of immunomodulatory genes was upregulated between 6 and 8 h post infection. In microglia cultures, the induction of several immune response pathways including cytokine responses was dependent on mitochondrial antiviral-signaling protein (MAVS). Consequently, in Mavs-deficient microglia the control of virus propagation failed as indicated by augmented virus titers and the accumulation of viral transcripts. Thus, in the analyzed in vitro system, MAVS signaling is critically required to achieve full microglia activation and to mediate profound antiviral effects. In Mavs-deficient mice, intranasal VSV instillation caused higher disease severity than in WT mice and virus dissemination was noticed beyond the olfactory bulb. Virus spread to inner regions of the olfactory bulb, i.e., the granular cell layer, correlated with the recruitment of highly inflammatory non-microglia myeloid cells into the olfactory bulb in Mavs-/- mice. Furthermore, increased cytokine levels were detected in the nasal cavity, the olfactory bulb and in other brain regions. Thus, microglial MAVS signaling is critically needed for virus sensing, full microglia activation, and for orchestration of protective immunity in the virus-infected CNS.
Collapse
Affiliation(s)
- Olivia Luise Gern
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, TWINCORE, Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Andreas Pavlou
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, TWINCORE, Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Felix Mulenge
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, TWINCORE, Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Lena Mareike Busker
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, TWINCORE, Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, 30559, Hannover, Germany
| | - Luca Ghita
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, TWINCORE, Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
- Genentech, South San Francisco, CA, USA
| | - Angela Aringo
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, TWINCORE, Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Bibiana Costa
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, TWINCORE, Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Julia Spanier
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, TWINCORE, Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Inken Waltl
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, TWINCORE, Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Martin Stangel
- Department of Neurology, Hannover Medical School, 30625, Hannover, Germany
- Translational Medicine Neuroscience, Biomedical Research, Novartis Pharma AG, Basel, 4056, Switzerland
- Center of Systems Neuroscience, Hannover, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, TWINCORE, Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany.
- Cluster of Excellence-Resolving Infection Susceptibility (RESIST, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.
| |
Collapse
|
5
|
Stillman JM, Kiniwa T, Schafer DP. Nucleic acid sensing in the central nervous system: Implications for neural circuit development, function, and degeneration. Immunol Rev 2024; 327:71-82. [PMID: 39503567 PMCID: PMC11653434 DOI: 10.1111/imr.13420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
Nucleic acids are a critical trigger for the innate immune response to infection, wherein pathogen-derived RNA and DNA are sensed by nucleic acid sensing receptors. This subsequently drives the production of type I interferon and other inflammatory cytokines to combat infection. While the system is designed such that these receptors should specifically recognize pathogen-derived nucleic acids, it is now clear that self-derived RNA and DNA can also stimulate these receptors to cause aberrant inflammation and autoimmune disease. Intriguingly, similar pathways are now emerging in the central nervous system in neurons and glial cells. As in the periphery, these signaling pathways are active in neurons and glia to present the spread of pathogens in the CNS. They further appear to be active even under steady conditions to regulate neuronal development and function, and they can become activated aberrantly during disease to propagate neuroinflammation and neurodegeneration. Here, we review the emerging new roles for nucleic acid sensing mechanisms in the CNS and raise open questions that we are poised to explore in the future.
Collapse
Affiliation(s)
- Jacob M. Stillman
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- University of Massachusetts Chan Morningside Graduate School of Biomedical Sciences, Neuroscience Program, Worcester, MA, USA
| | - Tsuyoshi Kiniwa
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Dorothy P. Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| |
Collapse
|
6
|
Pavlou A, Mulenge F, Gern OL, Busker LM, Greimel E, Waltl I, Kalinke U. Orchestration of antiviral responses within the infected central nervous system. Cell Mol Immunol 2024; 21:943-958. [PMID: 38997413 PMCID: PMC11364666 DOI: 10.1038/s41423-024-01181-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/05/2024] [Indexed: 07/14/2024] Open
Abstract
Many newly emerging and re-emerging viruses have neuroinvasive potential, underscoring viral encephalitis as a global research priority. Upon entry of the virus into the CNS, severe neurological life-threatening conditions may manifest that are associated with high morbidity and mortality. The currently available therapeutic arsenal against viral encephalitis is rather limited, emphasizing the need to better understand the conditions of local antiviral immunity within the infected CNS. In this review, we discuss new insights into the pathophysiology of viral encephalitis, with a focus on myeloid cells and CD8+ T cells, which critically contribute to protection against viral CNS infection. By illuminating the prerequisites of myeloid and T cell activation, discussing new discoveries regarding their transcriptional signatures, and dissecting the mechanisms of their recruitment to sites of viral replication within the CNS, we aim to further delineate the complexity of antiviral responses within the infected CNS. Moreover, we summarize the current knowledge in the field of virus infection and neurodegeneration and discuss the potential links of some neurotropic viruses with certain pathological hallmarks observed in neurodegeneration.
Collapse
Affiliation(s)
- Andreas Pavlou
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Felix Mulenge
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Olivia Luise Gern
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Lena Mareike Busker
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, 30559, Hannover, Germany
| | - Elisabeth Greimel
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Inken Waltl
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany.
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625, Hannover, Germany.
| |
Collapse
|
7
|
Rosendal E, Lindqvist R, Chotiwan N, Henriksson J, Överby AK. Transcriptional Response to Tick-Borne Flavivirus Infection in Neurons, Astrocytes and Microglia In Vivo and In Vitro. Viruses 2024; 16:1327. [PMID: 39205301 PMCID: PMC11359927 DOI: 10.3390/v16081327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Tick-borne encephalitis virus (TBEV) is a neurotropic member of the genus Orthoflavivirus (former Flavivirus) and is of significant health concern in Europe and Asia. TBEV pathogenesis may occur directly via virus-induced damage to neurons or through immunopathology due to excessive inflammation. While primary cells isolated from the host can be used to study the immune response to TBEV, it is still unclear how well these reflect the immune response elicited in vivo. Here, we compared the transcriptional response to TBEV and the less pathogenic tick-borne flavivirus, Langat virus (LGTV), in primary monocultures of neurons, astrocytes and microglia in vitro, with the transcriptional response in vivo captured by single-nuclei RNA sequencing (snRNA-seq) of a whole mouse cortex. We detected similar transcriptional changes induced by both LGTV and TBEV infection in vitro, with the lower response to LGTV likely resulting from slower viral kinetics. Gene set enrichment analysis showed a stronger transcriptional response in vivo than in vitro for astrocytes and microglia, with a limited overlap mainly dominated by interferon signaling. Together, this adds to our understanding of neurotropic flavivirus pathogenesis and the strengths and limitations of available model systems.
Collapse
Affiliation(s)
- Ebba Rosendal
- Department of Clinical Microbiology, Umeå University, 90185 Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, 90187 Umeå, Sweden
| | - Richard Lindqvist
- Department of Clinical Microbiology, Umeå University, 90185 Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, 90187 Umeå, Sweden
| | - Nunya Chotiwan
- Department of Clinical Microbiology, Umeå University, 90185 Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, 90187 Umeå, Sweden
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakarn 10540, Thailand
| | - Johan Henriksson
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, 90187 Umeå, Sweden
- Department of Molecular Biology, Icelab, Umeå Centre for Microbial Research (UCMR), Umeå University, 90187 Umeå, Sweden
| | - Anna K. Överby
- Department of Clinical Microbiology, Umeå University, 90185 Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, 90187 Umeå, Sweden
| |
Collapse
|
8
|
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
|
9
|
Di Pietro AA, Pasquini LA. A novel in vitro model for investigating oligodendroglial maturation and myelin deposition under demyelinating and remyelinating conditions: Impact of microglial depletion and repopulation. Mol Cell Neurosci 2024; 129:103937. [PMID: 38796120 DOI: 10.1016/j.mcn.2024.103937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/14/2024] [Accepted: 05/17/2024] [Indexed: 05/28/2024] Open
Abstract
Experimental models of multiple sclerosis (MS) have significantly contributed to our understanding of pathophysiology and the development of therapeutic interventions. Various in vivo animal models have successfully replicated key features of MS and associated pathophysiological processes, shedding light on the sequence of events leading to disease initiation, progression, and resolution. Nevertheless, these models often entail substantial costs and prolonged treatment periods. In contrast, in vitro models offer distinct advantages, including cost-effectiveness and precise control over experimental conditions, thereby facilitating more reproducible results. We have developed a novel in vitro model tailored to the study of oligodendroglial maturation and myelin deposition under demyelinating and remyelinating conditions, which encompasses all the cell types present in the central nervous system (CNS). Of note, our model enables the evaluation of microglial cell commitment through a protocol involving their depletion and subsequent repopulation. Given that the development and survival of microglia are critically reliant on colony-stimulating factor-1 receptor (CSF-1R) signaling, we have employed CSF-1R inhibition to effectively deplete microglia. This versatile model holds promise for the assessment of potential therapies aimed at promoting oligodendroglial differentiation to safeguard and repair myelin, hence mitigate neurodegenerative processes.
Collapse
Affiliation(s)
- Anabella Ayelen Di Pietro
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Cátedra de Química Biológica Patológica, Buenos Aires, Argentina; Universidad de Buenos Airess, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro C. Paladini, Facultad de Farmacia y Bioquímica, Buenos Aire, Argentina.
| | - Laura Andrea Pasquini
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Cátedra de Química Biológica Patológica, Buenos Aires, Argentina; Universidad de Buenos Airess, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro C. Paladini, Facultad de Farmacia y Bioquímica, Buenos Aire, Argentina.
| |
Collapse
|
10
|
Pavesi A, Tiecco G, Rossi L, Sforza A, Ciccarone A, Compostella F, Lovatti S, Tomasoni LR, Castelli F, Quiros-Roldan E. Inflammatory Response Associated with West Nile Neuroinvasive Disease: A Systematic Review. Viruses 2024; 16:383. [PMID: 38543749 PMCID: PMC10976239 DOI: 10.3390/v16030383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/21/2024] [Accepted: 02/27/2024] [Indexed: 04/09/2024] Open
Abstract
BACKGROUND West Nile virus (WNV) infection is a seasonal arbovirosis with the potential to cause severe neurological disease. Outcomes of the infection from WNV depend on viral factors (e.g., lineage) and host-intrinsic factors (e.g., age, sex, immunocompromising conditions). Immunity is essential to control the infection but may also prove detrimental to the host. Indeed, the persistence of high levels of pro-inflammatory cytokines and chemokines is associated with the development of blood-brain barrier (BBB) damage. Due to the importance of the inflammatory processes in the development of West Nile neuroinvasive disease (WNND), we reviewed the available literature on the subject. METHODS According to the 2020 updated PRISMA guidelines, all peer-reviewed articles regarding the inflammatory response associated with WNND were included. RESULTS One hundred and thirty-six articles were included in the data analysis and sorted into three groups (in vitro on-cell cultures, in vivo in animals, and in humans). The main cytokines found to be increased during WNND were IL-6 and TNF-α. We highlighted the generally small quantity and heterogeneity of information about the inflammatory patterns associated with WNND. CONCLUSIONS Further studies are needed to understand the pathogenesis of WNND and to investigate the extent and the way the host inflammatory response either helps in controlling the infection or in worsening the outcomes. This might prove useful both for the development of target therapies and for the development of molecular markers allowing early identification of patients displaying an inflammatory response that puts them at a higher risk of developing neuroinvasive disease and who might thus benefit from early antiviral therapies.
Collapse
Affiliation(s)
- Alessandro Pavesi
- Department of Clinical and Experimental Sciences, Unit of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (A.P.); (G.T.); (L.R.); (A.S.); (A.C.); (F.C.); (S.L.); (F.C.)
| | - Giorgio Tiecco
- Department of Clinical and Experimental Sciences, Unit of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (A.P.); (G.T.); (L.R.); (A.S.); (A.C.); (F.C.); (S.L.); (F.C.)
| | - Luca Rossi
- Department of Clinical and Experimental Sciences, Unit of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (A.P.); (G.T.); (L.R.); (A.S.); (A.C.); (F.C.); (S.L.); (F.C.)
| | - Anita Sforza
- Department of Clinical and Experimental Sciences, Unit of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (A.P.); (G.T.); (L.R.); (A.S.); (A.C.); (F.C.); (S.L.); (F.C.)
| | - Andrea Ciccarone
- Department of Clinical and Experimental Sciences, Unit of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (A.P.); (G.T.); (L.R.); (A.S.); (A.C.); (F.C.); (S.L.); (F.C.)
| | - Federico Compostella
- Department of Clinical and Experimental Sciences, Unit of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (A.P.); (G.T.); (L.R.); (A.S.); (A.C.); (F.C.); (S.L.); (F.C.)
| | - Sofia Lovatti
- Department of Clinical and Experimental Sciences, Unit of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (A.P.); (G.T.); (L.R.); (A.S.); (A.C.); (F.C.); (S.L.); (F.C.)
| | - Lina Rachele Tomasoni
- Unit of Infectious and Tropical Diseases, ASST Spedali Civili di Brescia, 25123 Brescia, Italy;
| | - Francesco Castelli
- Department of Clinical and Experimental Sciences, Unit of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (A.P.); (G.T.); (L.R.); (A.S.); (A.C.); (F.C.); (S.L.); (F.C.)
| | - Eugenia Quiros-Roldan
- Department of Clinical and Experimental Sciences, Unit of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (A.P.); (G.T.); (L.R.); (A.S.); (A.C.); (F.C.); (S.L.); (F.C.)
| |
Collapse
|
11
|
Rangel MV, Bourguet FA, Hall CI, Weilhammer DR. Evaluation of Inactivation Methods for Rift Valley Fever Virus in Mouse Microglia. Pathogens 2024; 13:159. [PMID: 38392897 PMCID: PMC10892077 DOI: 10.3390/pathogens13020159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/30/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Rift Valley fever phlebovirus (RVFV) is a highly pathogenic mosquito-borne virus with bioweapon potential due to its ability to be spread by aerosol transmission. Neurological symptoms are among the worst outcomes of infection, and understanding of pathogenesis mechanisms within the brain is limited. RVFV is classified as an overlap select agent by the CDC and USDA; therefore, experiments involving fully virulent strains of virus are tightly regulated. Here, we present two methods for inactivation of live virus within samples derived from mouse microglia cells using commercially available kits for the preparation of cells for flow cytometry and RNA extraction. Using the flow cytometry protocol, we demonstrate key differences in the response of primary murine microglia to infection with fully virulent versus attenuated RVFV.
Collapse
Affiliation(s)
- Margarita V. Rangel
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA; (M.V.R.); (F.A.B.)
| | - Feliza A. Bourguet
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA; (M.V.R.); (F.A.B.)
| | - Carolyn I. Hall
- Environment, Safety & Health, Biosafety & Biogovernance Functional Area, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA;
| | - Dina R. Weilhammer
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA; (M.V.R.); (F.A.B.)
| |
Collapse
|
12
|
Syage A, Pachow C, Cheng Y, Mangale V, Green KN, Lane TE. Microglia influence immune responses and restrict neurologic disease in response to central nervous system infection by a neurotropic murine coronavirus. Front Cell Neurosci 2023; 17:1291255. [PMID: 38099152 PMCID: PMC10719854 DOI: 10.3389/fncel.2023.1291255] [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: 09/08/2023] [Accepted: 11/09/2023] [Indexed: 12/17/2023] Open
Abstract
Intracranial (i.c.) 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 serve numerous functions including maintenance of the healthy central nervous system (CNS) and are among the first responders to injury or infection. More recently, studies have demonstrated that microglia aid in tailoring innate and adaptive immune responses following infection by neurotropic viruses including flaviviruses, herpesviruses, and picornaviruses. These findings have emphasized an important role for microglia in host defense against these viral pathogens. In addition, microglia are also critical in optimizing immune-mediated control of JHMV replication within the CNS while restricting the severity of demyelination and enhancing remyelination. This review will highlight our current understanding of the molecular and cellular mechanisms by which microglia aid in host defense, limit neurologic disease, and promote repair following CNS infection by a neurotropic murine coronavirus.
Collapse
Affiliation(s)
- Amber Syage
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
| | - Collin Pachow
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Yuting Cheng
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Vrushali Mangale
- Department of Pathology, University of Utah, Salt Lake City, UT, United States
| | - Kim N. Green
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
| | - Thomas E. Lane
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
- Center for Virus Research, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
13
|
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
|
14
|
Brito RMDM, da Silva MCM, Vieira-Santos F, de Almeida Lopes C, Souza JLN, Bastilho AL, de Barros Fernandes H, de Miranda AS, de Oliveira ACP, de Almeida Vitor RW, de Andrade-Neto VF, Bueno LL, Fujiwara RT, Magalhães LMD. Chronic infection by atypical Toxoplasma gondii strain induces disturbance in microglia population and altered behaviour in mice. Brain Behav Immun Health 2023; 30:100652. [PMID: 37396335 PMCID: PMC10308216 DOI: 10.1016/j.bbih.2023.100652] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 06/03/2023] [Accepted: 06/04/2023] [Indexed: 07/04/2023] Open
Abstract
Toxoplasma gondii chronic infection is characterized by the establishment of tissue cysts in the brain and increased levels of IFN-γ, which can lead to brain circuitry interference and consequently abnormal behaviour in mice. In this sense, the study presented here sought to investigate the impact of chronic infection by two T. gondii strains in the brain of infection-resistant mice, as a model for studying the involvement of chronic neuroinflammation with the development of behavioural alterations. For that, male BALB/c mice were divided into three groups: non-infected (Ni), infected with T. gondii ME49 clonal strain (ME49), and infected with TgCkBrRN2 atypical strain (CK2). Mice were monitored for 60 days to establish the chronic infection and then submitted to behavioural assessment. The enzyme-linked immunosorbent assay was used for measurement of specific IgG in the blood and levels of inflammatory cytokines and neurotrophic factors in the brain, and the cell's immunophenotype was determined by multiparametric flow cytometry. Mice infected with ME49 clonal strain displayed hyperlocomotor activity and memory deficit, although no signs of depressive- and/or anxiety-like behaviour were detected; on the other hand, chronic infection with CK2 atypical strain induced anxiety- and depressive-like behaviour. During chronic infection by CK2 atypical strain, mice displayed a higher number of T. gondii brain tissue cysts and inflammatory infiltrate, composed mainly of CD3+ T lymphocytes and Ly6Chi inflammatory monocytes, compared to mice infected with the ME49 clonal strain. Infected mice presented a marked decrease of microglia population compared to non-infected group. Chronic infection with CK2 strain produced elevated levels of IFN-γ and TNF-ɑ in the brain, decreased NGF levels in the prefrontal cortex and striatum, and altered levels of fractalkine (CX3CL1) in the prefrontal cortex and hippocampus. The persistent inflammation and the disturbance in the cerebral homeostasis may contribute to altered behaviour in mice, as the levels of IFN-γ were shown to be correlated with the behavioural parameters assessed here. Considering the high incidence and life-long persistence of T. gondii infection, this approach can be considered a suitable model for studying the impact of chronic infections in the brain and how it impacts in behavioural responses.
Collapse
Affiliation(s)
- Ramayana Morais de Medeiros Brito
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
- Laboratory of Malaria and Toxoplasmosis Biology, Department of Microbiology and Parasitology, Biosciences Centre, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Maria Carolina Machado da Silva
- Neuropharmacology Laboratory, Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Flaviane Vieira-Santos
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Camila de Almeida Lopes
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Jorge Lucas Nascimento Souza
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alexandre Lazoski Bastilho
- Laboratory of Malaria and Toxoplasmosis Biology, Department of Microbiology and Parasitology, Biosciences Centre, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Heliana de Barros Fernandes
- Laboratory of Neurobiology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Aline Silva de Miranda
- Laboratory of Neurobiology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Antônio Carlos Pinheiro de Oliveira
- Neuropharmacology Laboratory, Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ricardo Wagner de Almeida Vitor
- Laboratory of Toxoplasmosis, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Valter Ferreira de Andrade-Neto
- Laboratory of Malaria and Toxoplasmosis Biology, Department of Microbiology and Parasitology, Biosciences Centre, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Lilian Lacerda Bueno
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ricardo Toshio Fujiwara
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luísa Mourão Dias Magalhães
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
15
|
Frank JC, Song BH, Lee YM. Mice as an Animal Model for Japanese Encephalitis Virus Research: Mouse Susceptibility, Infection Route, and Viral Pathogenesis. Pathogens 2023; 12:pathogens12050715. [PMID: 37242385 DOI: 10.3390/pathogens12050715] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 05/09/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Japanese encephalitis virus (JEV), a zoonotic flavivirus, is principally transmitted by hematophagous mosquitoes, continually between susceptible animals and incidentally from those animals to humans. For almost a century since its discovery, JEV was geographically confined to the Asia-Pacific region with recurrent sizable outbreaks involving wildlife, livestock, and people. However, over the past decade, it has been detected for the first time in Europe (Italy) and Africa (Angola) but has yet to cause any recognizable outbreaks in humans. JEV infection leads to a broad spectrum of clinical outcomes, ranging from asymptomatic conditions to self-limiting febrile illnesses to life-threatening neurological complications, particularly Japanese encephalitis (JE). No clinically proven antiviral drugs are available to treat the development and progression of JE. There are, however, several live and killed vaccines that have been commercialized to prevent the infection and transmission of JEV, yet this virus remains the main cause of acute encephalitis syndrome with high morbidity and mortality among children in the endemic regions. Therefore, significant research efforts have been directed toward understanding the neuropathogenesis of JE to facilitate the development of effective treatments for the disease. Thus far, multiple laboratory animal models have been established for the study of JEV infection. In this review, we focus on mice, the most extensively used animal model for JEV research, and summarize the major findings on mouse susceptibility, infection route, and viral pathogenesis reported in the past and present, and discuss some unanswered key questions for future studies.
Collapse
Affiliation(s)
- Jordan C Frank
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA
| | - Byung-Hak Song
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA
| | - Young-Min Lee
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA
| |
Collapse
|
16
|
McMillan RE, Wang E, Carlin AF, Coufal NG. Human microglial models to study host-virus interactions. Exp Neurol 2023; 363:114375. [PMID: 36907350 PMCID: PMC10521930 DOI: 10.1016/j.expneurol.2023.114375] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/13/2023] [Accepted: 03/02/2023] [Indexed: 03/14/2023]
Abstract
Microglia, the resident macrophage of the central nervous system, are increasingly recognized as contributing to diverse aspects of human development, health, and disease. In recent years, numerous studies in both mouse and human models have identified microglia as a "double edged sword" in the progression of neurotropic viral infections: protecting against viral replication and cell death in some contexts, while acting as viral reservoirs and promoting excess cellular stress and cytotoxicity in others. It is imperative to understand the diversity of human microglial responses in order to therapeutically modulate them; however, modeling human microglia has been historically challenging due to significant interspecies differences in innate immunity and rapid transformation upon in vitro culture. In this review, we discuss the contribution of microglia to the neuropathogenesis of key neurotropic viral infections: human immunodeficiency virus 1 (HIV-1), Zika virus (ZIKV), Japanese encephalitis virus (JEV), West Nile virus (WNV), Herpes simplex virus (HSV), and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We pay special attention to recent work with human stem cell-derived microglia and propose strategies to leverage these powerful models to further uncover species- and disease-specific microglial responses and novel therapeutic interventions for neurotropic viral infections.
Collapse
Affiliation(s)
- Rachel E McMillan
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, United States of America; Department of Pathology and Medicine, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States of America
| | - Ellen Wang
- Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States of America; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92093, United States of America
| | - Aaron F Carlin
- Department of Pathology and Medicine, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States of America.
| | - Nicole G Coufal
- Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States of America; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92093, United States of America.
| |
Collapse
|
17
|
Spiteri AG, Wishart CL, Ni D, Viengkhou B, Macia L, Hofer MJ, King NJC. Temporal tracking of microglial and monocyte single-cell transcriptomics in lethal flavivirus infection. Acta Neuropathol Commun 2023; 11:60. [PMID: 37016414 PMCID: PMC10074823 DOI: 10.1186/s40478-023-01547-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/08/2023] [Indexed: 04/06/2023] Open
Abstract
As the resident parenchymal myeloid population in the central nervous system (CNS), microglia are strategically positioned to respond to neurotropic virus invasion and have been implicated in promoting both disease resolution and progression in the acute and post-infectious phase of virus encephalitis. In a mouse model of West Nile virus encephalitis (WNE), infection of the CNS results in recruitment of large numbers of peripheral immune cells into the brain, the majority being nitric oxide (NO)-producing Ly6Chi inflammatory monocyte-derived cells (MCs). In this model, these cells enhance immunopathology and mortality. However, the contribution of microglia to this response is currently undefined. Here we used a combination of experimental tools, including single-cell RNA sequencing (scRNA-seq), microglia and MC depletion reagents, high-dimensional spectral cytometry and computational algorithms to dissect the differential contribution of microglia and MCs to the anti-viral immune response in severe neuroinflammation seen in WNE. Intriguingly, analysis of scRNA-seq data revealed 6 unique microglia and 3 unique MC clusters that were predominantly timepoint-specific, demonstrating substantial transcriptional adaptation with disease progression over the course of WNE. While microglia and MC adopted unique gene expression profiles, gene ontology enrichment analysis, coupled with microglia and MC depletion studies, demonstrated a role for both of these cells in the trafficking of peripheral immune cells into the CNS, T cell responses and viral clearance. Over the course of infection, microglia transitioned from a homeostatic to an anti-viral and then into an immune cell-recruiting phenotype. Conversely, MC adopted antigen-presenting, immune cell-recruiting and NO-producing phenotypes, which all had anti-viral function. Overall, this study defines for the first time the single-cell transcriptomic responses of microglia and MCs over the course of WNE, demonstrating both protective and pathological roles of these cells that could potentially be targeted for differential therapeutic intervention to dampen immune-mediated pathology, while maintaining viral clearance functions.
Collapse
Affiliation(s)
- Alanna G Spiteri
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
- Sydney Cytometry, The University of Sydney and Centenary Institute, Sydney, NSW, 2006, Australia
- Ramaciotti Facility for Human Systems Biology, The University of Sydney and Centenary Institute, Sydney, NSW, 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Claire L Wishart
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
- Sydney Cytometry, The University of Sydney and Centenary Institute, Sydney, NSW, 2006, Australia
- Ramaciotti Facility for Human Systems Biology, The University of Sydney and Centenary Institute, Sydney, NSW, 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Duan Ni
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
- Chronic Diseases Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Barney Viengkhou
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Laurence Macia
- Sydney Cytometry, The University of Sydney and Centenary Institute, Sydney, NSW, 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
- Chronic Diseases Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Markus J Hofer
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
- The University of Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Nicholas J C King
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia.
- Sydney Cytometry, The University of Sydney and Centenary Institute, Sydney, NSW, 2006, Australia.
- Ramaciotti Facility for Human Systems Biology, The University of Sydney and Centenary Institute, Sydney, NSW, 2006, Australia.
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia.
- The University of Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, NSW, 2006, Australia.
- Sydney Nano, The University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
18
|
Stonedahl S, Leser JS, Clarke P, Potter H, Boyd TD, Tyler KL. Treatment with Granulocyte-Macrophage Colony-Stimulating Factor Reduces Viral Titers in the Brains of West Nile Virus-Infected Mice and Improves Survival. J Virol 2023; 97:e0180522. [PMID: 36802227 PMCID: PMC10062152 DOI: 10.1128/jvi.01805-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/29/2023] [Indexed: 02/23/2023] Open
Abstract
West Nile virus (WNV) is the leading cause of epidemic arboviral encephalitis in the United States. As there are currently no proven antiviral therapies or licensed human vaccines, understanding the neuropathogenesis of WNV is critical for rational therapeutic design. In WNV-infected mice, the depletion of microglia leads to enhanced viral replication, increased central nervous system (CNS) tissue injury, and increased mortality, suggesting that microglia play a critical role in protection against WNV neuroinvasive disease. To determine if augmenting microglial activation would provide a potential therapeutic strategy, we administered granulocyte-macrophage colony-stimulating factor (GM-CSF) to WNV-infected mice. Recombinant human GM-CSF (rHuGMCSF) (sargramostim [Leukine]) is an FDA-approved drug used to increase white blood cells following leukopenia-inducing chemotherapy or bone marrow transplantation. Daily treatment of both uninfected and WNV-infected mice with subcutaneous injections of GM-CSF resulted in microglial proliferation and activation as indicated by the enhanced expression of the microglia activation marker ionized calcium binding adaptor molecule 1 (Iba1) and several microglia-associated inflammatory cytokines, including CCL2 (C-C motif chemokine ligand 2), interleukin 6 (IL-6), and IL-10. In addition, more microglia adopted an activated morphology as demonstrated by increased sizes and more pronounced processes. GM-CSF-induced microglial activation in WNV-infected mice was associated with reduced viral titers and apoptotic activity (caspase 3) in the brains of WNV-infected mice and significantly increased survival. WNV-infected ex vivo brain slice cultures (BSCs) treated with GM-CSF also showed reduced viral titers and caspase 3 apoptotic cell death, indicating that GM-CSF specifically targets the CNS and that its actions are not dependent on peripheral immune activity. Our studies suggest that stimulation of microglial activation may be a viable therapeutic approach for the treatment of WNV neuroinvasive disease. IMPORTANCE Although rare, WNV encephalitis poses a devastating health concern, with few treatment options and frequent long-term neurological sequelae. Currently, there are no human vaccines or specific antivirals against WNV infections, so further research into potential new therapeutic agents is critical. This study presents a novel treatment option for WNV infections using GM-CSF and lays the foundation for further studies into the use of GM-CSF as a treatment for WNV encephalitis as well as a potential treatment for other viral infections.
Collapse
Affiliation(s)
- Sarah Stonedahl
- Department of Immunology, University of Colorado, Aurora, Colorado, USA
- Department of Microbiology, University of Colorado, Aurora, Colorado, USA
| | - J. Smith Leser
- Department of Neurology, University of Colorado, Aurora, Colorado, USA
| | - Penny Clarke
- Department of Neurology, University of Colorado, Aurora, Colorado, USA
| | - Huntington Potter
- Department of Neurology, University of Colorado, Aurora, Colorado, USA
- University of Colorado Alzheimer’s and Cognition Center, Aurora, Colorado, USA
- Linda Crnic Institute for Down Syndrome, Aurora, Colorado, USA
| | - Timothy D. Boyd
- University of Colorado Alzheimer’s and Cognition Center, Aurora, Colorado, USA
- Linda Crnic Institute for Down Syndrome, Aurora, Colorado, USA
| | - Kenneth L. Tyler
- Department of Neurology, University of Colorado, Aurora, Colorado, USA
- Division of Infectious Disease, Department of Medicine, University of Colorado, Aurora, Colorado, USA
- Denver VA Medical Center, Aurora, Colorado, USA
| |
Collapse
|
19
|
How viral infections cause neuronal dysfunction: a focus on the role of microglia and astrocytes. Biochem Soc Trans 2023; 51:259-274. [PMID: 36606670 DOI: 10.1042/bst20220771] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 01/07/2023]
Abstract
In recent decades, a number of infectious viruses have emerged from wildlife or reemerged that pose a serious threat to global health and economies worldwide. Although many of these viruses have a specific target tissue, neurotropic viruses have evolved mechanisms to exploit weaknesses in immune defenses that eventually allow them to reach and infect cells of the central nervous system (CNS). Once in the CNS, these viruses can cause severe neuronal damage, sometimes with long-lasting, life-threatening consequences. Remarkably, the ability to enter the CNS and cause neuronal infection does not appear to determine whether a viral strain causes neurological complications. The cellular mechanisms underlying the neurological consequences of viral infection are not fully understood, but they involve neuroimmune interactions that have so far focused mainly on microglia. As the major immune cells in the brain, reactive microglia play a central role in neuroinflammation by responding directly or indirectly to viruses. Chronic reactivity of microglia leads to functions that are distinct from their beneficial roles under physiological conditions and may result in neuronal damage that contributes to the pathogenesis of various neurological diseases. However, there is increasing evidence that reactive astrocytes also play an important role in the response to viruses. In this review article, we summarize the recent contributions of microglia and astrocytes to the neurological impairments caused by viral infections. By expanding knowledge in this area, therapeutic approaches targeting immunological pathways may reduce the incidence of neurological and neurodegenerative disorders and increase the therapeutic window for neural protection.
Collapse
|
20
|
Johnson HJ, Koshy AA. Understanding neuroinflammation through central nervous system infections. Curr Opin Neurobiol 2022; 76:102619. [PMID: 35985075 PMCID: PMC10147316 DOI: 10.1016/j.conb.2022.102619] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/09/2022] [Accepted: 07/08/2022] [Indexed: 11/28/2022]
Abstract
Neuroinflammation is now recognized to compound many central nervous system (CNS) pathologies, from stroke to dementia. As immune responses evolved to handle infections, studying CNS infections can offer unique insights into the CNS immune response and address questions such as: What defenses and strategies do CNS parenchymal cells deploy in response to a dangerous pathogen? How do CNS cells interact with each other and infiltrating immune cells to control microbes? What pathways are beneficial for the host or for the pathogen? Here, we review recent studies that use CNS-tropic infections in combination with cutting-edge techniques to delve into the complex relationships between microbes, immune cells, and cells of the CNS.
Collapse
Affiliation(s)
- Hannah J Johnson
- Neuroscience Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, USA
| | - Anita A Koshy
- Neuroscience Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, USA; Department of Neurology, University of Arizona, Tucson, AZ, USA; BIO5 Institute, University of Arizona, Tucson, AZ, USA; Department of Immunobiology, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
21
|
Aberrant Synaptic Pruning in CNS Diseases: A Critical Player in HIV-Associated Neurological Dysfunction? Cells 2022; 11:cells11121943. [PMID: 35741071 PMCID: PMC9222069 DOI: 10.3390/cells11121943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/28/2022] [Accepted: 06/14/2022] [Indexed: 02/04/2023] Open
Abstract
Even in the era of effective antiretroviral therapies, people living with Human Immunodeficiency Virus (HIV) are burdened with debilitating neurological dysfunction, such as HIV-associated neurocognitive disorders (HAND) and HIV-associated pain, for which there are no FDA approved treatments. Disruption to the neural circuits of cognition and pain in the form of synaptic degeneration is implicated in developing these dysfunctions. Glia-mediated synaptic pruning is a mechanism of structural plasticity in the healthy central nervous system (CNS), but recently, it has been discovered that dysregulated glia-mediated synaptic pruning is the cause of synaptic degeneration, leading to maladaptive plasticity and cognitive deficits in multiple diseases of the CNS. Considering the essential contribution of activated glial cells during the development of HAND and HIV-associated pain, it is possible that glia-mediated synaptic pruning is the causative mechanism of synaptic degeneration induced by HIV. This review will analyze the known examples of synaptic pruning during disease in order to better understand how this mechanism could contribute to the progression of HAND and HIV-associated pain.
Collapse
|
22
|
Stonedahl S, Leser JS, Clarke P, Tyler KL. Depletion of Microglia in an Ex Vivo Brain Slice Culture Model of West Nile Virus Infection Leads to Increased Viral Titers and Cell Death. Microbiol Spectr 2022; 10:e0068522. [PMID: 35412380 PMCID: PMC9045141 DOI: 10.1128/spectrum.00685-22] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 03/21/2022] [Indexed: 01/03/2023] Open
Abstract
West Nile virus (WNV) is a major cause of viral encephalitis in the United States. WNV infection of the brain leads to neuroinflammation characterized by activation of microglia, the resident phagocytic cells of the central nervous system (CNS). In this study, depletion of CNS microglia using the CSF1R antagonist PLX5622 increased the viral load in the brain and decreased the survival of mice infected with WNV (strain TX02). PLX5622 was also used in ex vivo brain slice cultures (BSCs) to investigate the role of intrinsic neuroinflammatory responses during WNV infection. PLX5622 effectively depleted microglia (>90% depletion) from BSCs resulting in increased viral titers (3 to 4-fold increase in PLX5622-treated samples) and enhanced virus-induced caspase 3 activity and cell death. Microglia depletion did not result in widespread alterations in cytokine and chemokine production in either uninfected or WNV infected BSCs. The results of this study demonstrated how microglia contribute to limiting viral growth and preventing cell death in WNV infected BSCs but were not required for the cytokine/chemokine response to WNV infection. This study highlighted the importance of microglia in the protection from neuroinvasive WNV infection and demonstrated that microglia responses were independent of WNV-induced peripheral immune responses. IMPORTANCE WNV infections of the CNS are rare but can have devastating long-term effects. There are currently no vaccines or specific antiviral treatments, so a better understanding of the pathogenesis and immune response to this virus is crucial. Previous studies have shown microglia to be important for protection from WNV, but more work is needed to fully comprehend the impact these cells have on neuroinvasive WNV infections. This study used PLX5622 to eliminate microglia in an ex vivo brain slice culture (BSC) model to investigate the role of microglia during a WNV infection. The use of BSCs provided a system in which immune responses innate to the CNS could be studied without interference from peripheral immunity. This study will allow for a better understanding of the complex nature of microglia during viral infections and will likely impact the development of new therapeutics that target microglia.
Collapse
Affiliation(s)
- Sarah Stonedahl
- Department of Immunology and Microbiology, University of Colorado, Aurora, Colorado, USA
| | | | - Penny Clarke
- Department of Neurology, University of Colorado, Aurora, Colorado, USA
| | - Kenneth L. Tyler
- Department of Neurology, University of Colorado, Aurora, Colorado, USA
- Division of Infectious Disease, Department of Medicine, University of Colorado, Aurora, Colorado, USA
- Denver Veteran Affairs Medical Center, Aurora, Colorado, USA
| |
Collapse
|
23
|
Spiteri AG, Ni D, Ling ZL, Macia L, Campbell IL, Hofer MJ, King NJC. PLX5622 Reduces Disease Severity in Lethal CNS Infection by Off-Target Inhibition of Peripheral Inflammatory Monocyte Production. Front Immunol 2022; 13:851556. [PMID: 35401512 PMCID: PMC8990748 DOI: 10.3389/fimmu.2022.851556] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/03/2022] [Indexed: 11/18/2022] Open
Abstract
PLX5622 is a CSF-1R inhibitor and microglia-depleting reagent, widely used to investigate the biology of this central nervous system (CNS)-resident myeloid population, but the indirect or off-target effects of this agent remain largely unexplored. In a murine model of severe neuroinflammation induced by West Nile virus encephalitis (WNE), we showed PLX5622 efficiently depleted both microglia and a sub-population of border-associated macrophages in the CNS. However, PLX5622 also significantly depleted mature Ly6Chi monocytes in the bone marrow (BM), inhibiting their proliferation and lethal recruitment into the infected brain, reducing neuroinflammation and clinical disease scores. Notably, in addition, BM dendritic cell subsets, plasmacytoid DC and classical DC, were depleted differentially in infected and uninfected mice. Confirming its protective effect in WNE, cessation of PLX5622 treatment exacerbated disease scores and was associated with robust repopulation of microglia, rebound BM monopoiesis and markedly increased inflammatory monocyte infiltration into the CNS. Monoclonal anti-CSF-1R antibody blockade late in WNE also impeded BM monocyte proliferation and recruitment to the brain, suggesting that the protective effect of PLX5622 is via the inhibition of CSF-1R, rather than other kinase targets. Importantly, BrdU incorporation in PLX5622-treated mice, suggest remaining microglia proliferate independently of CSF-1 in WNE. Our study uncovers significantly broader effects of PLX5622 on the myeloid lineage beyond microglia depletion, advising caution in the interpretation of PLX5622 data as microglia-specific. However, this work also strikingly demonstrates the unexpected therapeutic potential of this molecule in CNS viral infection, as well as other monocyte-mediated diseases.
Collapse
Affiliation(s)
- Alanna G Spiteri
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Sydney Cytometry, The University of Sydney and Centenary Institute, Sydney, NSW, Australia.,Ramaciotti Facility for Human Systems Biology, The University of Sydney and Centenary Institute, Sydney, NSW, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Duan Ni
- Sydney Cytometry, The University of Sydney and Centenary Institute, Sydney, NSW, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.,Chronic Diseases Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Zheng Lung Ling
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Sydney Cytometry, The University of Sydney and Centenary Institute, Sydney, NSW, Australia.,Ramaciotti Facility for Human Systems Biology, The University of Sydney and Centenary Institute, Sydney, NSW, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Laurence Macia
- Sydney Cytometry, The University of Sydney and Centenary Institute, Sydney, NSW, Australia.,Ramaciotti Facility for Human Systems Biology, The University of Sydney and Centenary Institute, Sydney, NSW, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.,Chronic Diseases Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Iain L Campbell
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Markus J Hofer
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, Australia.,The University of Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, NSW, Australia
| | - Nicholas J C King
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Sydney Cytometry, The University of Sydney and Centenary Institute, Sydney, NSW, Australia.,Ramaciotti Facility for Human Systems Biology, The University of Sydney and Centenary Institute, Sydney, NSW, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.,The University of Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, NSW, Australia.,The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
24
|
Cowan MN, Sethi I, Harris TH. Microglia in CNS infections: insights from Toxoplasma gondii and other pathogens. Trends Parasitol 2022; 38:217-229. [PMID: 35039238 PMCID: PMC8852251 DOI: 10.1016/j.pt.2021.12.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 12/11/2022]
Abstract
Microglia, the resident immune cells of the central nervous system (CNS), are poised to respond to neuropathology. Microglia play multiple roles in maintaining homeostasis and promoting inflammation in numerous disease states. The study of microglial innate immune programs has largely focused on exploring neurodegenerative disease states with the use of genetic targeting approaches. Our understanding of how microglia participate in immune responses against pathogens is just beginning to take shape. Here, we review existing animal models of CNS infection, with a focus on how microglial physiology and inflammatory processes control protozoan and viral infections of the brain. We further discuss how microglial participation in over-exuberant immune responses can drive immunopathology that is detrimental to CNS health and homeostasis.
Collapse
Affiliation(s)
- Maureen N. Cowan
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, VA, United States
| | - Ish Sethi
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, VA, United States
| | - Tajie H. Harris
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, VA, United States,Correspondence: (T. H. Harris)
| |
Collapse
|
25
|
O'Brien CA, Bennett FC, Bennett ML. Microglia in antiviral immunity of the brain and spinal cord. Semin Immunol 2022; 60:101650. [PMID: 36099864 PMCID: PMC9934594 DOI: 10.1016/j.smim.2022.101650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/17/2022] [Accepted: 08/30/2022] [Indexed: 01/15/2023]
Abstract
Viral infections of the central nervous system (CNS) are a significant cause of neurological impairment and mortality worldwide. As tissue resident macrophages, microglia are critical initial responders to CNS viral infection. Microglia seem to coordinate brain-wide antiviral responses of both brain resident cells and infiltrating immune cells. This review discusses how microglia may promote this antiviral response at a molecular level, from potential mechanisms of virus recognition to downstream cytokine responses and interaction with antiviral T cells. Recent advancements in genetic tools to specifically target microglia in vivo promise to further our understanding about the precise mechanistic role of microglia in CNS infection.
Collapse
Affiliation(s)
- Carleigh A O'Brien
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States.
| | - F Chris Bennett
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States; Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Mariko L Bennett
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States; Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| |
Collapse
|
26
|
Microglia Do Not Restrict SARS-CoV-2 Replication following Infection of the Central Nervous System of K18-Human ACE2 Transgenic Mice. J Virol 2022; 96:e0196921. [PMID: 34935438 PMCID: PMC8865461 DOI: 10.1128/jvi.01969-21] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Unlike SARS-CoV-1 and MERS-CoV, infection with SARS-CoV-2, the viral pathogen responsible for COVID-19, is often associated with neurologic symptoms that range from mild to severe, yet increasing evidence argues the virus does not exhibit extensive neuroinvasive properties. We demonstrate SARS-CoV-2 can infect and replicate in human iPSC-derived neurons and that infection shows limited antiviral and inflammatory responses but increased activation of EIF2 signaling following infection as determined by RNA sequencing. Intranasal infection of K18 human ACE2 transgenic mice (K18-hACE2) with SARS-CoV-2 resulted in lung pathology associated with viral replication and immune cell infiltration. In addition, ∼50% of infected mice exhibited CNS infection characterized by wide-spread viral replication in neurons accompanied by increased expression of chemokine (Cxcl9, Cxcl10, Ccl2, Ccl5 and Ccl19) and cytokine (Ifn-λ and Tnf-α) transcripts associated with microgliosis and a neuroinflammatory response consisting primarily of monocytes/macrophages. Microglia depletion via administration of colony-stimulating factor 1 receptor inhibitor, PLX5622, in SARS-CoV-2 infected mice did not affect survival or viral replication but did result in dampened expression of proinflammatory cytokine/chemokine transcripts and a reduction in monocyte/macrophage infiltration. These results argue that microglia are dispensable in terms of controlling SARS-CoV-2 replication in in the K18-hACE2 model but do contribute to an inflammatory response through expression of pro-inflammatory genes. Collectively, these findings contribute to previous work demonstrating the ability of SARS-CoV-2 to infect neurons as well as emphasizing the potential use of the K18-hACE2 model to study immunological and neuropathological aspects related to SARS-CoV-2-induced neurologic disease. 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 role of microglia in aiding in host defense following experimental infection of the central nervous system (CNS) of K18-hACE2 with SARS-CoV-2, the causative agent of COVID-19. Neurologic symptoms that range in severity are common in COVID-19 patients and understanding immune responses that contribute to restricting neurologic disease can provide important insight into better understanding consequences associated with SARS-CoV-2 infection of the CNS.
Collapse
|
27
|
Spiteri AG, Wishart CL, Pamphlett R, Locatelli G, King NJC. Microglia and monocytes in inflammatory CNS disease: integrating phenotype and function. Acta Neuropathol 2022; 143:179-224. [PMID: 34853891 PMCID: PMC8742818 DOI: 10.1007/s00401-021-02384-2] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/11/2021] [Accepted: 11/11/2021] [Indexed: 02/08/2023]
Abstract
In neurological diseases, the actions of microglia, the resident myeloid cells of the CNS parenchyma, may diverge from, or intersect with, those of recruited monocytes to drive immune-mediated pathology. However, defining the precise roles of each cell type has historically been impeded by the lack of discriminating markers and experimental systems capable of accurately identifying them. Our ability to distinguish microglia from monocytes in neuroinflammation has advanced with single-cell technologies, new markers and drugs that identify and deplete them, respectively. Nevertheless, the focus of individual studies on particular cell types, diseases or experimental approaches has limited our ability to connect phenotype and function more widely and across diverse CNS pathologies. Here, we critically review, tabulate and integrate the disease-specific functions and immune profiles of microglia and monocytes to provide a comprehensive atlas of myeloid responses in viral encephalitis, demyelination, neurodegeneration and ischemic injury. In emphasizing the differential roles of microglia and monocytes in the severe neuroinflammatory disease of viral encephalitis, we connect inflammatory pathways common to equally incapacitating diseases with less severe inflammation. We examine these findings in the context of human studies and highlight the benefits and inherent limitations of animal models that may impede or facilitate clinical translation. This enables us to highlight common and contrasting, non-redundant and often opposing roles of microglia and monocytes in disease that could be targeted therapeutically.
Collapse
|
28
|
Kendall LV, Boyd TD, Sillau SH, Bosco-Lauth A, Markham N, Fong D, Clarke P, Tyler KL, Potter H. GM-CSF Promotes Immune Response and Survival in a Mouse Model of COVID-19. RESEARCH SQUARE 2022:rs.3.rs-1213395. [PMID: 35118463 PMCID: PMC8811947 DOI: 10.21203/rs.3.rs-1213395/v1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
COVID-19 results in increased expression of inflammatory cytokines, but inflammation-targeting clinical trials have yielded poor to mixed results. Our studies of other disorders with an inflammatory component, including Alzheimer's disease, chemobrain, Down syndrome, normal aging, and West Nile Virus infection, showed that treatment with the 'pro-inflammatory' cytokine granulocyte-macrophage colony stimulating factor (GM-CSF) in humans or mouse models alleviated clinical, behavioral, and pathological features. We proposed that human recombinant GM-CSF (sargramostim) be repurposed to promote both the innate and adaptive immune responses in COVID-19 to reduce viral load and mortality1. Here, we report the results of a placebo-controlled study of GM-CSF in human ACE2 transgenic mice inoculated intranasally with SARS-CoV2 virus, a model of COVID-19. Infection resulted in high viral titers in lungs and brains and over 85% mortality. GM-CSF treatment beginning one day after infection increased anti-viral antibody titers, lowered mean lung viral titers proportionately (p=0.0020) and increased the odds of long-term survival by up to 5.8-fold (p=0.0358), compared to placebo. These findings suggest that, as an activator of both the innate and adaptive immune systems, GM-CSF/sargramostim may be an effective COVID-19 therapy with the potential to protect from re-infection more effectively than treatment with antiviral drugs or monoclonal antibodies.
Collapse
Affiliation(s)
- L V Kendall
- Colorado State University, Department of Microbiology, Immunology and Pathology, Fort Collins, CO
| | - T D Boyd
- University of Colorado Alzheimer's and Cognition Center, Aurora, CO
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, CO
| | - S H Sillau
- University of Colorado Alzheimer's and Cognition Center, Aurora, CO
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO
| | - A Bosco-Lauth
- Colorado State University, Department of Biomedical Sciences, Fort Collins, CO
| | - N Markham
- University of Colorado Alzheimer's and Cognition Center, Aurora, CO
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, CO
| | - D Fong
- Department of Pathology, University of Colorado Anschutz School of Medicine, Aurora, CO
| | - P Clarke
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO
| | - K L Tyler
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO
- Denver VA Medical Center, Denver CO
- Departments of Immunology and Microbiology, and Medicine, University of Colorado School of Medicine, Aurora, CO
| | - H Potter
- University of Colorado Alzheimer's and Cognition Center, Aurora, CO
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, CO
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO
| |
Collapse
|
29
|
Microglia activate early anti-viral responses upon HSV-1 entry into the brain to counteract development of encephalitis-like disease in mice. J Virol 2022; 96:e0131121. [PMID: 35045263 DOI: 10.1128/jvi.01311-21] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Spread of herpes simplex virus 1 (HSV1) from the periphery to the central nervous system (CNS) can lead to extensive infection and pathological inflammation in the brain, causing herpes simplex encephalitis (HSE). It has been shown that microglia, the CNS-resident macrophages, are involved in early sensing of HSV1 and an induction of antiviral responses. In addition, infiltration of peripheral immune cells may contribute to control of viral infection. In this study, we tested the effect of microglia depletion in a mouse model of HSE. Increased viral titers and increased disease severity were observed in microglia-depleted mice. The effect of microglia depletion was more pronounced in wild-type than in cGas-/- mice, revealing that this immune sensor contributes to the antiviral activity of microglia. Importantly, microglia depletion led to reduced production of type I interferon (IFN), pro-inflammatory cytokines and chemokines at early time points after viral entry into the CNS. In line with this, in vitro experiments on murine primary CNS cells demonstrated microglial presence to be essential for IFN RNA induction, and control of HSV1 replication. However, the effect of microglia depletion on expression of IFNs, and inflammatory cytokines was restricted to early time point of HSV1 entry into the CNS. There was no major alteration of infiltration of CD45-positive cells in microglia-depleted mice. Collectively, our data demonstrate a key role for microglia in controlling HSV1 replication early after viral entry into the CNS and highlight the importance of a prompt antiviral innate response to reduce the risk of HSE development. Importance One of the most devastating and acute neurological conditions is encephalitis, i.e. inflammation of brain tissue. Herpes simplex virus 1 (HSV1) is a highly prevalent pathogen in humans, and the most frequent cause of viral sporadic encephalitis, called herpes simplex encephalitis (HSE). HSV1 has the ability to infect peripheral neurons and reach the central nervous system (CNS) of humans, where it can be detected by brain resident cells and infiltrating immune cells, leading to protective and damaging immune responses. In this study, we investigated the effects of a depletion of microglia, the main brain-resident immune cell type. For this purpose, we used a mouse model of HSE. We found that viral levels increased and disease symptoms worsened in microglia-depleted mice. In addition, mice lacking a major sensor of viral DNA, cGAS, manifested more pronounced disease than wild-type mice, highlighting the importance of this immune sensor in the activity of microglia. Evidently, microglia depletion led to a reduced production of many known antiviral factors, most notably type I interferon (IFN). The importance of microglia in the early control of HSV1 spread and the generation of antiviral responses is further demonstrated by experiments on murine mixed glial cell cultures. Interestingly, mice with microglia depletion exhibited an unaltered activation of antiviral responses and recruitment of immune cells from the periphery at later time points of infection, but this did not prevent the development of the disease. Overall, the data highlight the importance of a rapid activation of the host defense, with microglia playing a critical role in controlling HSV1 infection, which eventually prevents damage to neurons and brain tissue.
Collapse
|
30
|
Soto-Diaz K, Vailati-Riboni M, Louie AY, McKim DB, Gaskins HR, Johnson RW, Steelman AJ. Treatment With the CSF1R Antagonist GW2580, Sensitizes Microglia to Reactive Oxygen Species. Front Immunol 2021; 12:734349. [PMID: 34899694 PMCID: PMC8664563 DOI: 10.3389/fimmu.2021.734349] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 11/01/2021] [Indexed: 01/29/2023] Open
Abstract
Microglia activation and proliferation are hallmarks of many neurodegenerative disorders and may contribute to disease pathogenesis. Neurons actively regulate microglia survival and function, in part by secreting the microglia mitogen interleukin (IL)-34. Both IL-34 and colony stimulating factor (CSF)-1 bind colony stimulating factor receptor (CSFR)1 expressed on microglia. Systemic treatment with central nervous system (CNS) penetrant, CSFR1 antagonists, results in microglia death in a dose dependent matter, while others, such as GW2580, suppress activation during disease states without altering viability. However, it is not known how treatment with non-penetrant CSF1R antagonists, such as GW2580, affect the normal physiology of microglia. To determine how GW2580 affects microglia function, C57BL/6J mice were orally gavaged with vehicle or GW2580 (80mg/kg/d) for 8 days. Body weights and burrowing behavior were measured throughout the experiment. The effects of GW2580 on circulating leukocyte populations, brain microglia morphology, and the transcriptome of magnetically isolated adult brain microglia were determined. Body weights, burrowing behavior, and circulating leukocytes were not affected by treatment. Analysis of Iba-1 stained brain microglia indicated that GW2580 treatment altered morphology, but not cell number. Analysis of RNA-sequencing data indicated that genes related to reactive oxygen species (ROS) regulation and survival were suppressed by treatment. Treatment of primary microglia cultures with GW2580 resulted in a dose-dependent reduction in viability only when the cells were concurrently treated with LPS, an inducer of ROS. Pre-treatment with the ROS inhibitor, YCG063, blocked treatment induced reductions in viability. Finally, GW2580 sensitized microglia to hydrogen peroxide induced cell death. Together, these data suggest that partial CSF1R antagonism may render microglia more susceptible to reactive oxygen and nitrogen species.
Collapse
Affiliation(s)
- Katiria Soto-Diaz
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Mario Vailati-Riboni
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Allison Y Louie
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Daniel B McKim
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - H Rex Gaskins
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Department of Biomedical and Translational Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Rodney W Johnson
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Andrew J Steelman
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| |
Collapse
|
31
|
Beneficial and detrimental functions of microglia during viral encephalitis. Trends Neurosci 2021; 45:158-170. [PMID: 34906391 DOI: 10.1016/j.tins.2021.11.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 10/28/2021] [Accepted: 11/16/2021] [Indexed: 12/11/2022]
Abstract
Microglia are resident immune cells of the central nervous system (CNS) with multiple functions in health and disease. Their response during encephalitis depends on whether inflammation is triggered in a sterile or infectious manner, and in the latter case on the type of the infecting pathogen. Even though recent technological innovations advanced the understanding of the broad spectrum of microglia responses during viral encephalitis (VE), it is not entirely clear which microglia gene expression profiles are associated with antiviral and detrimental activities. Here, we review novel approaches to study microglia and the latest concepts of their function in VE. Improved understanding of microglial functions will be essential for the development of new therapeutic interventions for VE.
Collapse
|
32
|
Tsai MS, Wang LC, Tsai HY, Lin YJ, Wu HL, Tzeng SF, Hsu SM, Chen SH. Microglia Reduce Herpes Simplex Virus 1 Lethality of Mice with Decreased T Cell and Interferon Responses in Brains. Int J Mol Sci 2021; 22:ijms222212457. [PMID: 34830340 PMCID: PMC8624831 DOI: 10.3390/ijms222212457] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/09/2021] [Accepted: 11/16/2021] [Indexed: 12/11/2022] Open
Abstract
Herpes simplex virus 1 (HSV-1) infects the majority of the human population and can induce encephalitis, which is the most common cause of sporadic, fatal encephalitis. An increase of microglia is detected in the brains of encephalitis patients. The issues regarding whether and how microglia protect the host and neurons from HSV-1 infection remain elusive. Using a murine infection model, we showed that HSV-1 infection on corneas increased the number of microglia to outnumber those of infiltrating leukocytes (macrophages, neutrophils, and T cells) and enhanced microglia activation in brains. HSV-1 antigens were detected in brain neurons, which were surrounded by microglia. Microglia depletion increased HSV-1 lethality of mice with elevated brain levels of viral loads, infected neurons, neuron loss, CD4 T cells, CD8 T cells, neutrophils, interferon (IFN)-β, and IFN-γ. In vitro studies demonstrated that microglia from infected mice reduced virus infectivity. Moreover, microglia induced IFN-β and the signaling pathway of signal transducer and activator of transcription (STAT) 1 to inhibit viral replication and damage of neurons. Our study reveals how microglia protect the host and neurons from HSV-1 infection.
Collapse
Affiliation(s)
- Meng-Shan Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; (M.-S.T.); (H.-L.W.)
| | - Li-Chiu Wang
- School of Medicine, I-Shou University, Kaohsiung 824, Taiwan;
| | - Hsien-Yang Tsai
- Department of Ophthalmology, Tzu Chi Hospital, Taichung 427, Taiwan;
| | - Yu-Jheng Lin
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan;
| | - Hua-Lin Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; (M.-S.T.); (H.-L.W.)
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Shun-Fen Tzeng
- Department of Life Sciences, College of Biological Science and Biotechnology, National Cheng Kung University, Tainan 701, Taiwan;
| | - Sheng-Min Hsu
- Department of Ophthalmology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- Correspondence: (S.-M.H.); (S.-H.C.)
| | - Shun-Hua Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; (M.-S.T.); (H.-L.W.)
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan;
- Correspondence: (S.-M.H.); (S.-H.C.)
| |
Collapse
|
33
|
Olivarria GM, Cheng Y, Furman S, Pachow C, Hohsfield LA, Smith-geater C, Miramontes R, Wu J, Burns MS, Tsourmas KI, Stocksdale J, Manlapaz C, Yong WH, Teijaro J, Edwards R, Green KN, Thompson LM, Lane TE. Microglia do not restrict SARS-CoV-2 replication following infection of the central nervous system of K18-hACE2 transgenic mice.. [PMID: 34816260 PMCID: PMC8609895 DOI: 10.1101/2021.11.15.468761] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
AbstractUnlike SARS-CoV-1 and MERS-CoV, infection with SARS-CoV-2, the viral pathogen responsible for COVID-19, is often associated with neurologic symptoms that range from mild to severe, yet increasing evidence argues the virus does not exhibit extensive neuroinvasive properties. We demonstrate SARS-CoV-2 can infect and replicate in human iPSC-derived neurons and that infection shows limited anti-viral and inflammatory responses but increased activation of EIF2 signaling following infection as determined by RNA sequencing. Intranasal infection of K18 human ACE2 transgenic mice (K18-hACE2) with SARS-CoV-2 resulted in lung pathology associated with viral replication and immune cell infiltration. In addition, ∼50% of infected mice exhibited CNS infection characterized by wide-spread viral replication in neurons accompanied by increased expression of chemokine (Cxcl9, Cxcl10, Ccl2, Ccl5 and Ccl19) and cytokine (Ifn-λ and Tnf-α) transcripts associated with microgliosis and a neuroinflammatory response consisting primarily of monocytes/macrophages. Microglia depletion via administration of colony-stimulating factor 1 receptor inhibitor, PLX5622, in SARS-CoV-2 infected mice did not affect survival or viral replication but did result in dampened expression of proinflammatory cytokine/chemokine transcripts and a reduction in monocyte/macrophage infiltration. These results argue that microglia are dispensable in terms of controlling SARS-CoV-2 replication in in the K18-hACE2 model but do contribute to an inflammatory response through expression of pro-inflammatory genes. Collectively, these findings contribute to previous work demonstrating the ability of SARS-CoV-2 to infect neurons as well as emphasizing the potential use of the K18-hACE2 model to study immunological and neuropathological aspects related to SARS-CoV-2-induced neurologic disease.ImportanceUnderstanding 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 role of microglia in aiding in host defense following experimental infection of the central nervous system (CNS) of K18-hACE2 with SARS-CoV-2, the causative agent of COVID-19. Neurologic symptoms that range in severity are common in COVID-19 patients and understanding immune responses that contribute to restricting neurologic disease can provide important insight into better understanding consequences associated with SARS-CoV-2 infection of the CNS.
Collapse
|
34
|
Jhan MK, Chen CL, Shen TJ, Tseng PC, Wang YT, Satria RD, Yu CY, Lin CF. Polarization of Type 1 Macrophages Is Associated with the Severity of Viral Encephalitis Caused by Japanese Encephalitis Virus and Dengue Virus. Cells 2021; 10:3181. [PMID: 34831405 PMCID: PMC8621422 DOI: 10.3390/cells10113181] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 12/20/2022] Open
Abstract
Infection with flaviviruses causes mild to severe diseases, including viral hemorrhagic fever, vascular shock syndrome, and viral encephalitis. Several animal models explore the pathogenesis of viral encephalitis, as shown by neuron destruction due to neurotoxicity after viral infection. While neuronal cells are injuries caused by inflammatory cytokine production following microglial/macrophage activation, the blockade of inflammatory cytokines can reduce neurotoxicity to improve the survival rate. This study investigated the involvement of macrophage phenotypes in facilitating CNS inflammation and neurotoxicity during flavivirus infection, including the Japanese encephalitis virus, dengue virus (DENV), and Zika virus. Mice infected with different flaviviruses presented encephalitis-like symptoms, including limbic seizure and paralysis. Histology indicated that brain lesions were identified in the hippocampus and surrounded by mononuclear cells. In those regions, both the infiltrated macrophages and resident microglia were significantly increased. RNA-seq analysis showed the gene profile shifting toward type 1 macrophage (M1) polarization, while M1 markers validated this phenomenon. Pharmacologically blocking C-C chemokine receptor 2 and tumor necrosis factor-α partly retarded DENV-induced M1 polarization. In summary, flavivirus infection, such as JEV and DENV, promoted type 1 macrophage polarization in the brain associated with encephalitic severity.
Collapse
MESH Headings
- Animals
- Animals, Suckling
- Cell Line
- Cell Polarity
- Dengue Virus/physiology
- Disease Models, Animal
- Encephalitis Virus, Japanese/physiology
- Encephalitis, Japanese/immunology
- Encephalitis, Japanese/pathology
- Encephalitis, Japanese/virology
- Encephalitis, Viral/immunology
- Encephalitis, Viral/pathology
- Encephalitis, Viral/virology
- Hippocampus/pathology
- Inflammation/pathology
- Macrophages/pathology
- Mice, Inbred ICR
- Neurotoxins/toxicity
- Receptors, CCR2/metabolism
- Severity of Illness Index
- Tumor Necrosis Factor-alpha/metabolism
- Mice
Collapse
Affiliation(s)
- Ming-Kai Jhan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (M.-K.J.); (T.-J.S.)
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (P.-C.T.); (Y.-T.W.); (R.D.S.)
| | - Chia-Ling Chen
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Ting-Jing Shen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (M.-K.J.); (T.-J.S.)
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (P.-C.T.); (Y.-T.W.); (R.D.S.)
| | - Po-Chun Tseng
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (P.-C.T.); (Y.-T.W.); (R.D.S.)
- Core Laboratory of Immune Monitoring, Office of Research & Development, Taipei Medical University, Taipei 110, Taiwan
| | - Yung-Ting Wang
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (P.-C.T.); (Y.-T.W.); (R.D.S.)
| | - Rahmat Dani Satria
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (P.-C.T.); (Y.-T.W.); (R.D.S.)
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Laboratory Medicine, Department of Clinical Pathology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
- Clinical Laboratory Installation, Dr. Sardjito Central General Hospital, Yogyakarta 55281, Indonesia
| | - Chia-Yi Yu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350, Taiwan;
| | - Chiou-Feng Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (M.-K.J.); (T.-J.S.)
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (P.-C.T.); (Y.-T.W.); (R.D.S.)
- Core Laboratory of Immune Monitoring, Office of Research & Development, Taipei Medical University, Taipei 110, Taiwan
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
35
|
Differential Effects of Prostaglandin D 2 Signaling on Macrophages and Microglia in Murine Coronavirus Encephalomyelitis. mBio 2021; 12:e0196921. [PMID: 34488442 PMCID: PMC8546556 DOI: 10.1128/mbio.01969-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Microglia and macrophages initiate and orchestrate the innate immune response to central nervous system (CNS) virus infections. Microglia initiate neurotropic coronavirus clearance from the CNS, but the role of infiltrating macrophages is not well understood. Here, using mice lacking cell-specific expression of DP1, the receptor for prostaglandin D2 (PGD2), we delineate the relative roles of PGD2 signaling in microglia and macrophages in murine coronavirus-infected mice. We show that the absence of PGD2/DP1 signaling on microglia recapitulated the suboptimal immune response observed in global DP1−/− mice. Unexpectedly, the absence of the DP1 receptor on macrophages had an opposite effect, resulting in enhanced activation and more rapid virus clearance. However, microglia are still required for disease resolution, even when macrophages are highly activated, in part because they are required for macrophage recruitment to sites of infection. Together, these results identify key differences in the effects of PGD2/DP1 signaling on microglia and macrophages and illustrate the complex relationship between the two types of myeloid cells.
Collapse
|
36
|
Gern OL, Mulenge F, Pavlou A, Ghita L, Steffen I, Stangel M, Kalinke U. Toll-like Receptors in Viral Encephalitis. Viruses 2021; 13:v13102065. [PMID: 34696494 PMCID: PMC8540543 DOI: 10.3390/v13102065] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 12/23/2022] Open
Abstract
Viral encephalitis is a rare but serious syndrome. In addition to DNA-encoded herpes viruses, such as herpes simplex virus and varicella zoster virus, RNA-encoded viruses from the families of Flaviviridae, Rhabdoviridae and Paramyxoviridae are important neurotropic viruses. Whereas in the periphery, the role of Toll-like receptors (TLR) during immune stimulation is well understood, TLR functions within the CNS are less clear. On one hand, TLRs can affect the physiology of neurons during neuronal progenitor cell differentiation and neurite outgrowth, whereas under conditions of infection, the complex interplay between TLR stimulated neurons, astrocytes and microglia is just on the verge of being understood. In this review, we summarize the current knowledge about which TLRs are expressed by cell subsets of the CNS. Furthermore, we specifically highlight functional implications of TLR stimulation in neurons, astrocytes and microglia. After briefly illuminating some examples of viral evasion strategies from TLR signaling, we report on the current knowledge of primary immunodeficiencies in TLR signaling and their consequences for viral encephalitis. Finally, we provide an outlook with examples of TLR agonist mediated intervention strategies and potentiation of vaccine responses against neurotropic virus infections.
Collapse
Affiliation(s)
- Olivia Luise Gern
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625 Hannover, Germany; (F.M.); (A.P.); (L.G.); (U.K.)
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany
- Correspondence:
| | - Felix Mulenge
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625 Hannover, Germany; (F.M.); (A.P.); (L.G.); (U.K.)
| | - Andreas Pavlou
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625 Hannover, Germany; (F.M.); (A.P.); (L.G.); (U.K.)
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
- Center for Systems Neuroscience, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Luca Ghita
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625 Hannover, Germany; (F.M.); (A.P.); (L.G.); (U.K.)
- Division of Infectious Diseases and Geographic Medicine, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Imke Steffen
- Department of Biochemistry and Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany;
| | - Martin Stangel
- Translational Medicine, Novartis Institute for Biomedical Research (NIBR), 4056 Basel, Switzerland;
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625 Hannover, Germany; (F.M.); (A.P.); (L.G.); (U.K.)
- Cluster of Excellence—Resolving Infection Susceptibility (RESIST, EXC 2155), Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| |
Collapse
|
37
|
Krzyzowska M, Kowalczyk A, Skulska K, Thörn K, Eriksson K. Fas/FasL Contributes to HSV-1 Brain Infection and Neuroinflammation. Front Immunol 2021; 12:714821. [PMID: 34526992 PMCID: PMC8437342 DOI: 10.3389/fimmu.2021.714821] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/16/2021] [Indexed: 12/14/2022] Open
Abstract
The Fas/FasL pathway plays a key role in immune homeostasis and immune surveillance. In the central nervous system (CNS) Fas/FasL is involved in axonal outgrowth and adult neurogenesis. However, little is known about the role of the Fas/FasL pathway in herpes encephalitis. In this study, we used a neuropathogenic clinical strain of herpes simplex virus type 1 (HSV-1) to explore infection-induced inflammation and immune responses in the mouse brain and the role of Fas/FasL in antiviral CNS immunity. HSV-1 CNS infection induced the infiltration of Fas- FasL-bearing monocytes and T cells in the brain and also to an up-regulation of Fas and FasL expression on resident astrocytes and microglia within infected sites. Upon infection, Fas- and FasL-deficient mice (lpr and gld) were partially protected from encephalitis with a decreased morbidity and mortality compared to WT mice. Fas/FasL deficiency promoted cell-mediated immunity within the CNS. Fas receptor stimulation abrogated HSV-1 induced activation and inflammatory reactions in microglia from WT mice, while lack of Fas or FasL led to a more pronounced activation of monocytes and microglia and also to an enhanced differentiation of these cells into a pro-inflammatory M1 phenotype. Furthermore, the specific immune system was more efficient in Fas- and FasL-deficient mice with significantly higher numbers of infiltrating HSV-1-specific cytotoxic T cells in the brain. Our data indicate that the Fas/FasL pathway leads to excessive neuroinflammation during HSV-1 infection, which is associated with a diminished anti-viral response and an excessive neuroinflammation.
Collapse
Affiliation(s)
- Malgorzata Krzyzowska
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Virology and Cell Biology, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland.,Laboratory of Nanobiology and Biomaterials, Military Institute of Hygiene and Epidemiology, Warsaw, Poland
| | - Andrzej Kowalczyk
- Department of Virology and Cell Biology, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| | - Katarzyna Skulska
- Department of Virology and Cell Biology, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| | - Karolina Thörn
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kristina Eriksson
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
38
|
Sanchez JMS, DePaula-Silva AB, Doty DJ, Hanak TJ, Truong A, Libbey JE, Fujinami RS. The CSF1R-Microglia Axis Has Protective Host-Specific Roles During Neurotropic Picornavirus Infection. Front Immunol 2021; 12:621090. [PMID: 34566948 PMCID: PMC8458822 DOI: 10.3389/fimmu.2021.621090] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 08/16/2021] [Indexed: 01/19/2023] Open
Abstract
Viral encephalitis is a major cause of morbidity and mortality, but the manifestation of disease varies greatly between individuals even in response to the same virus. Microglia are professional antigen presenting cells that reside in the central nervous system (CNS) parenchyma that are poised to respond to viral insults. However, the role of microglia in initiating and coordinating the antiviral response is not completely understood. Utilizing Theiler's murine encephalomyelitis virus (TMEV), a neurotropic picornavirus, and PLX5622, a small molecule inhibitor of colony-stimulating factor 1 receptor (CSF1R) signaling that can deplete microglia in the CNS; we investigated the role of the CSF1R-microglia axis in neurotropic picornavirus infection of C57BL/6J and SJL/J mice. These mouse strains differ in their ability to clear TMEV and exhibit different neurological disease in response to TMEV infection. CSF1R antagonism in C57BL/6J mice, which normally clear TMEV in the CNS, led to acute fatal encephalitis. In contrast, CSF1R antagonism in SJL/J mice, which normally develop a chronic CNS TMEV infection, did not result in acute encephalitis, but exacerbated TMEV-induced demyelination. Immunologically, inhibition of CSF1R in C57BL/6J mice reduced major histocompatibility complex II expression in microglia, decreased the proportion of regulatory T cells in the CNS, and upregulated proinflammatory pathways in CNS T cells. Acute CSF1R inhibition in SJL/J mice had no effect on microglial MHC-II expression and upregulated anti-inflammatory pathways in CNS T cells, however chronic CSF1R inhibition resulted in broad immunosuppression. Our results demonstrate strain-specific effects of the CSF1R-microglia axis in the context of neurotropic viral infection as well as inherent differences in microglial antigen presentation and subsequent T cell crosstalk that contribute to susceptibility to neurotropic picornavirus infection.
Collapse
Affiliation(s)
- John Michael S. Sanchez
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, United States
| | | | - Daniel J. Doty
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Tyler J. Hanak
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Amanda Truong
- Department of Oncological Sciences, Huntsman Cancer Institute, Salt Lake City, UT, United States
| | - Jane E. Libbey
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Robert S. Fujinami
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, United States
| |
Collapse
|
39
|
Reinert LS, Rashidi AS, Tran DN, Katzilieris-Petras G, Hvidt AK, Gohr M, Fruhwürth S, Bodda C, Thomsen MK, Vendelbo MH, Khan AR, Hansen B, Bergström P, Agholme L, Mogensen TH, Christensen MH, Nyengaard JR, Sen GC, Zetterberg H, Verjans GM, Paludan SR. Brain immune cells undergo cGAS/STING-dependent apoptosis during herpes simplex virus type 1 infection to limit type I IFN production. J Clin Invest 2021; 131:136824. [PMID: 32990676 DOI: 10.1172/jci136824] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 09/23/2020] [Indexed: 12/15/2022] Open
Abstract
Protection of the brain from viral infections involves the type I IFN (IFN-I) system, defects in which render humans susceptible to herpes simplex encephalitis (HSE). However, excessive cerebral IFN-I levels lead to pathologies, suggesting the need for tight regulation of responses. Based on data from mouse models, human HSE cases, and primary cell culture systems, we showed that microglia and other immune cells undergo apoptosis in the HSV-1-infected brain through a mechanism dependent on the cyclic GMP-AMP synthase/stimulator of interferon genes (cGAS/STING) pathway, but independent of IFN-I. HSV-1 infection of microglia induced cGAS-dependent apoptosis at high viral doses, whereas lower viral doses led to IFN-I responses. Importantly, inhibition of caspase activity prevented microglial cell death and augmented IFN-I responses. Accordingly, HSV-1-infected organotypic brain slices or mice treated with a caspase inhibitor exhibited lower viral load and an improved infection outcome. Collectively, we identify an activation-induced apoptosis program in brain immune cells that downmodulates local immune responses.
Collapse
Affiliation(s)
- Line S Reinert
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Ahmad S Rashidi
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Viroscience, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Diana N Tran
- Department of Viroscience, Erasmus Medical Centre, Rotterdam, Netherlands
| | | | - Astrid K Hvidt
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Mette Gohr
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Stefanie Fruhwürth
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Sweden
| | | | | | - Mikkel H Vendelbo
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Denmark
| | - Ahmad R Khan
- Center of Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark.,Centre of Biomedical Research, SGPGI Campus, Lucknow, India
| | - Brian Hansen
- Center of Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark
| | - Petra Bergström
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Germany
| | - Lotta Agholme
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Germany
| | | | | | - Jens R Nyengaard
- Department of Clinical Medicine, University of Aarhus, Aarhus, Denmark
| | - Ganes C Sen
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom.,UK Dementia Research Institute at UCL, London, United Kingdom
| | | | - Søren R Paludan
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Sweden
| |
Collapse
|
40
|
Intrinsic Innate Immune Responses Control Viral Growth and Protect against Neuronal Death in an Ex Vivo Model of West Nile Virus-Induced Central Nervous System Disease. J Virol 2021; 95:e0083521. [PMID: 34190599 DOI: 10.1128/jvi.00835-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Recruitment of immune cells from the periphery is critical for controlling West Nile virus (WNV) growth in the central nervous system (CNS) and preventing subsequent WNV-induced CNS disease. Neuroinflammatory responses, including the release of proinflammatory cytokines and chemokines by CNS cells, influence the entry and function of peripheral immune cells that infiltrate the CNS. However, these same cytokines and chemokines contribute to tissue damage in other models of CNS injury. Rosiglitazone is a peroxisome proliferator-activated receptor gamma (PPARγ) agonist that inhibits neuroinflammation. We used rosiglitazone in WNV-infected ex vivo brain slice cultures (BSC) to investigate the role of neuroinflammation within the CNS in the absence of peripheral immune cells. Rosiglitazone treatment inhibited WNV-induced expression of proinflammatory chemokines and cytokines, interferon beta (IFN-β), and IFN-stimulated genes (ISG) and also decreased WNV-induced activation of microglia. These decreased neuroinflammatory responses were associated with activation of astrocytes, robust viral growth, increased activation of caspase 3, and increased neuronal loss. Rosiglitazone had a similar effect on in vivo WNV infection, causing increased viral growth, tissue damage, and disease severity in infected mice, even though the number of infiltrating peripheral immune cells was higher in rosiglitazone-treated, WNV-infected mice than in untreated, infected controls. These results indicate that local neuroinflammatory responses are capable of controlling viral growth within the CNS and limiting neuronal loss and may function to keep the virus in check prior to the infiltration of peripheral immune cells, limiting both virus- and immune-mediated neuronal damage. IMPORTANCE West Nile virus is the most common cause of epidemic encephalitis in the United States and can result in debilitating CNS disease. There are no effective vaccines or treatments for WNV-induced CNS disease in humans. The peripheral immune response is critical for protection against WNV CNS infections. We now demonstrate that intrinsic immune responses also control viral growth and limit neuronal loss. These findings have important implications for developing new therapies for WNV-induced CNS disease.
Collapse
|
41
|
Enlow W, Bordeleau M, Piret J, Ibáñez FG, Uyar O, Venable MC, Goyette N, Carbonneau J, Tremblay ME, Boivin G. Microglia are involved in phagocytosis and extracellular digestion during Zika virus encephalitis in young adult immunodeficient mice. J Neuroinflammation 2021; 18:178. [PMID: 34399779 PMCID: PMC8369691 DOI: 10.1186/s12974-021-02221-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/16/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Zika virus (ZIKV) has been associated with several neurological complications in adult patients. METHODS We used a mouse model deficient in TRIF and IPS-1 adaptor proteins, which are involved in type I interferon production, to study the role of microglia during brain infection by ZIKV. Young adult mice were infected intravenously with the contemporary ZIKV strain PRVABC59 (1 × 105 PFUs/100 µL). RESULTS Infected mice did not present overt clinical signs of the disease nor body weight loss compared with noninfected animals. However, mice exhibited a viremia and a brain viral load that were maximal (1.3 × 105 genome copies/mL and 9.8 × 107 genome copies/g of brain) on days 3 and 7 post-infection (p.i.), respectively. Immunohistochemistry analysis showed that ZIKV antigens were distributed in several regions of the brain, especially the dorsal hippocampus. The number of Iba1+/TMEM119+ microglia remained similar in infected versus noninfected mice, but their cell body and arborization areas significantly increased in the stratum radiatum and stratum lacunosum-moleculare layers of the dorsal hippocampus cornu ammoni (CA)1, indicating a reactive state. Ultrastructural analyses also revealed that microglia displayed increased phagocytic activities and extracellular digestion of degraded elements during infection. Mice pharmacologically depleted in microglia with PLX5622 presented a higher brain viral load compared to untreated group (2.8 × 1010 versus 8.5 × 108 genome copies/g of brain on day 10 p.i.) as well as an increased number of ZIKV antigens labeled with immunogold in the cytoplasm and endoplasmic reticulum of neurons and astrocytes indicating an enhanced viral replication. Furthermore, endosomes of astrocytes contained nanogold particles together with digested materials, suggesting a compensatory phagocytic activity upon microglial depletion. CONCLUSIONS These results indicate that microglia are involved in the control of ZIKV replication and/or its elimination in the brain. After depletion of microglia, the removal of ZIKV-infected cells by phagocytosis could be partly compensated by astrocytes.
Collapse
Affiliation(s)
- William Enlow
- Centre de Recherche en Infectiologie, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Maude Bordeleau
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada.,Neurosciences Axis, Centre de recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Jocelyne Piret
- Centre de Recherche en Infectiologie, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Fernando González Ibáñez
- Neurosciences Axis, Centre de recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada.,Department of Molecular Medicine, Université Laval, Quebec City, QC, Canada.,Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Olus Uyar
- Centre de Recherche en Infectiologie, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Marie-Christine Venable
- Centre de Recherche en Infectiologie, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Nathalie Goyette
- Centre de Recherche en Infectiologie, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Julie Carbonneau
- Centre de Recherche en Infectiologie, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Marie-Eve Tremblay
- Neurosciences Axis, Centre de recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada. .,Department of Molecular Medicine, Université Laval, Quebec City, QC, Canada. .,Division of Medical Sciences, University of Victoria, Victoria, BC, Canada. .,Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada. .,Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada.
| | - Guy Boivin
- Centre de Recherche en Infectiologie, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada.
| |
Collapse
|
42
|
Spiteri AG, Terry RL, Wishart CL, Ashhurst TM, Campbell IL, Hofer MJ, King NJC. High-parameter cytometry unmasks microglial cell spatio-temporal response kinetics in severe neuroinflammatory disease. J Neuroinflammation 2021; 18:166. [PMID: 34311763 PMCID: PMC8314570 DOI: 10.1186/s12974-021-02214-y] [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: 07/07/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Differentiating infiltrating myeloid cells from resident microglia in neuroinflammatory disease is challenging, because bone marrow-derived inflammatory monocytes infiltrating the inflamed brain adopt a 'microglia-like' phenotype. This precludes the accurate identification of either cell type without genetic manipulation, which is important to understand their temporal contribution to disease and inform effective intervention in its pathogenesis. During West Nile virus (WNV) encephalitis, widespread neuronal infection drives substantial CNS infiltration of inflammatory monocytes, causing severe immunopathology and/or death, but the role of microglia in this remains unclear. METHODS Using high-parameter cytometry and dimensionality-reduction, we devised a simple, novel gating strategy to identify microglia and infiltrating myeloid cells during WNV-infection. Validating our strategy, we (1) blocked the entry of infiltrating myeloid populations from peripheral blood using monoclonal blocking antibodies, (2) adoptively transferred BM-derived monocytes and tracked their phenotypic changes after infiltration and (3) labelled peripheral leukocytes that infiltrate into the brain with an intravenous dye. We demonstrated that myeloid immigrants populated only the identified macrophage gates, while PLX5622 depletion reduced all 4 subsets defined by the microglial gates. RESULTS Using this gating approach, we identified four consistent microglia subsets in the homeostatic and WNV-infected brain. These were P2RY12hi CD86-, P2RY12hi CD86+ and P2RY12lo CD86- P2RY12lo CD86+. During infection, 2 further populations were identified as 'inflammatory' and 'microglia-like' macrophages, recruited from the bone marrow. Detailed kinetic analysis showed significant increases in the proportions of both P2RY12lo microglia subsets in all anatomical areas, largely at the expense of the P2RY12hi CD86- subset, with the latter undergoing compensatory proliferation, suggesting replenishment of, and differentiation from this subset in response to infection. Microglia altered their morphology early in infection, with all cells adopting temporal and regional disease-specific phenotypes. Late in disease, microglia produced IL-12, downregulated CX3CR1, F4/80 and TMEM119 and underwent apoptosis. Infiltrating macrophages expressed both TMEM119 and P2RY12 de novo, with the microglia-like subset notably exhibiting the highest proportional myeloid population death. CONCLUSIONS Our approach enables detailed kinetic analysis of resident vs infiltrating myeloid cells in a wide range of neuroinflammatory models without non-physiological manipulation. This will more clearly inform potential therapeutic approaches that specifically modulate these cells.
Collapse
Affiliation(s)
- Alanna G Spiteri
- Discipline of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Rachel L Terry
- Discipline of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
- Current Address: Children's Cancer Institute, Randwick, New South Wales, Australia
- Current Affiliation: Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Claire L Wishart
- Discipline of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Thomas M Ashhurst
- Discipline of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
- Sydney Cytometry Facility, The University of Sydney and Centenary Institute, Sydney, Australia
- Ramaciotti Facility for Human Systems Biology, The University of Sydney and Centenary Institute, Sydney, Australia
- Marie Bashir Institute for Infectious Diseases and Biosecurity (MBI), Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Iain L Campbell
- Marie Bashir Institute for Infectious Diseases and Biosecurity (MBI), Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, Australia
- School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia
| | - Markus J Hofer
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
- Marie Bashir Institute for Infectious Diseases and Biosecurity (MBI), Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, Australia
- School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia
| | - Nicholas J C King
- Discipline of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, Australia.
- Charles Perkins Centre, The University of Sydney, Sydney, Australia.
- Sydney Cytometry Facility, The University of Sydney and Centenary Institute, Sydney, Australia.
- Ramaciotti Facility for Human Systems Biology, The University of Sydney and Centenary Institute, Sydney, Australia.
- Marie Bashir Institute for Infectious Diseases and Biosecurity (MBI), Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, Australia.
- Nano Institute, The University of Sydney, Sydney, Australia.
| |
Collapse
|
43
|
Awogbindin IO, Ben-Azu B, Olusola BA, Akinluyi ET, Adeniyi PA, Di Paolo T, Tremblay MÈ. Microglial Implications in SARS-CoV-2 Infection and COVID-19: Lessons From Viral RNA Neurotropism and Possible Relevance to Parkinson's Disease. Front Cell Neurosci 2021; 15:670298. [PMID: 34211370 PMCID: PMC8240959 DOI: 10.3389/fncel.2021.670298] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 05/05/2021] [Indexed: 12/24/2022] Open
Abstract
Since December 2019, humankind has been experiencing a ravaging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) outbreak, the second coronavirus pandemic in a decade after the Middle East respiratory syndrome coronavirus (MERS-CoV) disease in 2012. Infection with SARS-CoV-2 results in Coronavirus disease 2019 (COVID-19), which is responsible for over 3.1 million deaths worldwide. With the emergence of a second and a third wave of infection across the globe, and the rising record of multiple reinfections and relapses, SARS-CoV-2 infection shows no sign of abating. In addition, it is now evident that SARS-CoV-2 infection presents with neurological symptoms that include early hyposmia, ischemic stroke, meningitis, delirium and falls, even after viral clearance. This may suggest chronic or permanent changes to the neurons, glial cells, and/or brain vasculature in response to SARS-CoV-2 infection or COVID-19. Within the central nervous system (CNS), microglia act as the central housekeepers against altered homeostatic states, including during viral neurotropic infections. In this review, we highlight microglial responses to viral neuroinfections, especially those with a similar genetic composition and route of entry as SARS-CoV-2. As the primary sensor of viral infection in the CNS, we describe the pathogenic and neuroinvasive mechanisms of RNA viruses and SARS-CoV-2 vis-à-vis the microglial means of viral recognition. Responses of microglia which may culminate in viral clearance or immunopathology are also covered. Lastly, we further discuss the implication of SARS-CoV-2 CNS invasion on microglial plasticity and associated long-term neurodegeneration. As such, this review provides insight into some of the mechanisms by which microglia could contribute to the pathophysiology of post-COVID-19 neurological sequelae and disorders, including Parkinson's disease, which could be pervasive in the coming years given the growing numbers of infected and re-infected individuals globally.
Collapse
Affiliation(s)
- Ifeoluwa O. Awogbindin
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Neuroimmunology Group, Molecular Drug Metabolism and Toxicology Laboratory, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Benneth Ben-Azu
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Nigeria
| | - Babatunde A. Olusola
- Department of Virology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Elizabeth T. Akinluyi
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Philip A. Adeniyi
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Therese Di Paolo
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Neurology and Neurosurgery Department, McGill University, Montréal, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec, QC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
44
|
Matejuk A, Vandenbark AA, Offner H. Cross-Talk of the CNS With Immune Cells and Functions in Health and Disease. Front Neurol 2021; 12:672455. [PMID: 34135852 PMCID: PMC8200536 DOI: 10.3389/fneur.2021.672455] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/19/2021] [Indexed: 12/16/2022] Open
Abstract
The immune system's role is much more than merely recognizing self vs. non-self and involves maintaining homeostasis and integrity of the organism starting from early development to ensure proper organ function later in life. Unlike other systems, the central nervous system (CNS) is separated from the peripheral immune machinery that, for decades, has been envisioned almost entirely as detrimental to the nervous system. New research changes this view and shows that blood-borne immune cells (both adaptive and innate) can provide homeostatic support to the CNS via neuroimmune communication. Neurodegeneration is mostly viewed through the lens of the resident brain immune populations with little attention to peripheral circulation. For example, cognition declines with impairment of peripheral adaptive immunity but not with the removal of microglia. Therapeutic failures of agents targeting the neuroinflammation framework (inhibiting immune response), especially in neurodegenerative disorders, call for a reconsideration of immune response contributions. It is crucial to understand cross-talk between the CNS and the immune system in health and disease to decipher neurodestructive and neuroprotective immune mechanisms for more efficient therapeutic strategies.
Collapse
Affiliation(s)
- Agata Matejuk
- Department of Immunology, Collegium Medicum, University of Zielona Góra, Zielona Góra, Poland
| | - Arthur A Vandenbark
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, United States.,Department of Neurology, Oregon Health and Science University, Portland, OR, United States.,Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, United States
| | - Halina Offner
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, United States.,Department of Neurology, Oregon Health and Science University, Portland, OR, United States.,Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
45
|
Neuroimmune cleanup crews in brain injury. Trends Immunol 2021; 42:480-494. [PMID: 33941486 DOI: 10.1016/j.it.2021.04.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 12/21/2022]
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability. Mounting evidence indicates that the immune system is critically involved in TBI pathogenesis, where it is deployed to dispose of neurotoxic material generated from head trauma and to instruct the wound healing process. However, the immune response to brain damage must be carefully held in check as aberrant regulation of immune signaling can lead to deleterious neuroinflammation, brain pathology, and neurological dysfunction. Efficient clearance of neurotoxic material by microglia (the brain's resident phagocytes) and the glymphatic-meningeal lymphatic drainage system are paramount to keeping the immune system in balance following head trauma. In this review, we highlight emerging evidence that defines pivotal roles for microglia and the recently discovered glymphatic-meningeal lymphatic system in TBI pathogenesis.
Collapse
|
46
|
Chang CY, Wu CC, Wang JD, Li JR, Wang YY, Lin SY, Chen WY, Liao SL, Chen CJ. DHA attenuated Japanese Encephalitis virus infection-induced neuroinflammation and neuronal cell death in cultured rat Neuron/glia. Brain Behav Immun 2021; 93:194-205. [PMID: 33486004 DOI: 10.1016/j.bbi.2021.01.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 11/20/2020] [Accepted: 01/11/2021] [Indexed: 12/26/2022] Open
Abstract
Japanese Encephalitis Virus (JEV) is a neurotropic virus and its Central Nervous System (CNS) infection causes fatal encephalitis with high mortality and morbidity. Microglial activation and consequences of bystander damage appear to be the dominant mechanisms for Japanese Encephalitis and complications. Docosahexaenoic acid (DHA), an essential fatty acid and a major component of brain cell membranes, possesses additional biological activities, including anti-apoptosis, anti-inflammation, and neuroprotection. Through this study, we have provided experimental evidence showing the anti-inflammatory, neuroprotective, and anti-viral effects of DHA against JEV infection in rat Neuron/glia cultures. By Neuron/glia and Neuron cultures, DHA protected against neuronal cell death upon JEV infection and reduced JEV amplification. In Neuron/glia and Microglia cultures, the effects of DHA were accompanied by the downregulation of pro-inflammatory M1 microglia, upregulation of anti-inflammatory M2 microglia, and reduction of neurotoxic cytokine expression, which could be attributed to its interference in the Toll-Like Receptor (TLR), Mitogen-Activated Protein Kinase (MAPK), and Interferon/Janus Kinase/Signal Transducers and Activators of Transcription (Stat), along with the NF-κB, AP-1, and c-AMP Response Element Binding Protein (CREB) controlled transcriptional programs. Parallel anti-inflammatory effects against JEV infection were duplicated by G Protein-Coupled Receptor (GPR120) and GPR40 agonists and a reversal of DHA-mediated anti-inflammation was seen in the presence of GPR120 antagonist, while the GPR40 was less effectiveness. Since increasing evidence indicates its neuroprotection against neurodegenerative diseases, DHA is a proposed anti-inflammatory and neuroprotective candidate for the treatment of neuroinflammation-accompanied viral pathogenesis such as Japanese Encephalitis.
Collapse
Affiliation(s)
- Cheng-Yi Chang
- Department of Surgery, Feng Yuan Hospital, Taichung City, Taiwan
| | - Chih-Cheng Wu
- Department of Anesthesiology, Taichung Veterans General Hospital, Taichung City, Taiwan; Department of Financial Engineering, Providence University, Taichung City, Taiwan; Department of Data Science and Big Data Analytics, Providence University, Taichung City, Taiwan
| | - Jiaan-Der Wang
- Children's Medical Center, Taichung Veterans General Hospital, Taichung City, Taiwan; Department of Industrial Engineering and Enterprise Information, Tunghai University, Taichung City, Taiwan
| | - Jian-Ri Li
- Division of Urology, Taichung Veterans General Hospital, Taichung City, Taiwan; Department of Nursing, HungKuang University, Taichung City, Taiwan
| | - Ya-Yu Wang
- Department of Family Medicine, Taichung Veterans General Hospital, Taichung City, Taiwan; Institute of Clinical Medicine, National Yang Ming University, Taipei City, Taiwan
| | - Shih-Yi Lin
- Institute of Clinical Medicine, National Yang Ming University, Taipei City, Taiwan; Center for Geriatrics and Gerontology, Taichung Veterans General Hospital, Taichung City, Taiwan
| | - Wen-Ying Chen
- Department of Veterinary Medicine, College of Life Sciences, National Chung-Hsing University, Taichung City, Taiwan
| | - Su-Lan Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City, Taiwan
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City, Taiwan; Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung City, Taiwan; Ph.D. Program in Translational Medicine, College of Life Sciences, National Chung-Hsing University, Taichung City, Taiwan.
| |
Collapse
|
47
|
Chhatbar C, Prinz M. The roles of microglia in viral encephalitis: from sensome to therapeutic targeting. Cell Mol Immunol 2021; 18:250-258. [PMID: 33437050 PMCID: PMC7802409 DOI: 10.1038/s41423-020-00620-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 12/08/2020] [Indexed: 01/31/2023] Open
Abstract
Viral encephalitis is a devastating disease with high mortality, and survivors often suffer from severe neurological complications. Microglia are innate immune cells of the central nervous system (CNS) parenchyma whose turnover is reliant on local proliferation. Microglia express a diverse range of proteins, which allows them to continuously sense the environment and quickly react to changes. Under inflammatory conditions such as CNS viral infection, microglia promote innate and adaptive immune responses to protect the host. However, during viral infection, a dysregulated microglia-T-cell interplay may result in altered phagocytosis of neuronal synapses by microglia that causes neurocognitive impairment. In this review, we summarize the current knowledge on the role of microglia in viral encephalitis, propose questions to be answered in the future and suggest possible therapeutic targets.
Collapse
Affiliation(s)
- Chintan Chhatbar
- grid.5963.9Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- grid.5963.9Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany ,grid.5963.9Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany ,grid.5963.9Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| |
Collapse
|
48
|
Limbic Encephalitis Brain Damage Induced by Cocal Virus in Adult Mice Is Reduced by Environmental Enrichment: Neuropathological and Behavioral Studies. Viruses 2020; 13:v13010048. [PMID: 33396704 PMCID: PMC7824630 DOI: 10.3390/v13010048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 12/14/2020] [Indexed: 12/26/2022] Open
Abstract
We previously demonstrated, using the Piry virus model, that environmental enrichment promotes higher T-cell infiltration, fewer microglial changes, and faster central nervous system (CNS) virus clearance in adult mice. However, little is known about disease progression, behavioral changes, CNS cytokine concentration, and neuropathology in limbic encephalitis in experimental models. Using Cocal virus, we infected C57Bl6 adult mice and studied the neuroanatomical distribution of viral antigens in correlation with the microglial morphological response, measured the CNS cytokine concentration, and assessed behavioral changes. C57Bl6 adult mice were maintained in an impoverished environment (IE) or enriched environment (EE) for four months and then subjected to the open field test. Afterwards, an equal volume of normal or virus-infected brain homogenate was nasally instilled. The brains were processed to detect viral antigens and microglial morphological changes using selective immunolabeling. We demonstrated earlier significant weight loss and higher mortality in IE mice. Additionally, behavioral analysis revealed a significant influence of the environment on locomotor and exploratory activity that was associated with less neuroinvasion and a reduced microglial response. Thus, environmental enrichment was associated with a more effective immune response in a mouse model of limbic encephalitis, allowing faster viral clearance/decreased viral dissemination, reduced disease progression, and less CNS damage.
Collapse
|
49
|
An Early Microglial Response Is Needed To Efficiently Control Herpes Simplex Virus Encephalitis. J Virol 2020; 94:JVI.01428-20. [PMID: 32938766 DOI: 10.1128/jvi.01428-20] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/09/2020] [Indexed: 12/25/2022] Open
Abstract
The role of a signaling pathway through macrophage colony-stimulating factor (MCSF) and its receptor, macrophage colony-stimulating factor 1 receptor (CSF1R), during experimental herpes simplex virus 1 (HSV-1) encephalitis (HSE) was studied by two different approaches. First, we evaluated the effect of stimulation of the MCSF/CSF1R axis before infection. Exogenous MCSF (40 μg/kg of body weight intraperitoneally [i.p.]) was administered once daily to BALB/c mice on days 4 and 2 before intranasal infection with 2,500 PFU of HSV-1. MCSF treatment significantly increased mouse survival compared to saline (50% versus 10%; P = 0.0169). On day 6 postinfection (p.i.), brain viral titers were significantly decreased, whereas beta interferon (IFN-β) was significantly increased in mice treated with MCSF compared to mice treated with saline. The number of CD68+ (a phagocytosis marker) microglial cells was significantly increased in MCSF-treated mice compared to the saline-treated group. Secondly, we conditionally depleted CSF1R on microglial cells of CSF1R-loxP-CX3CR1-cre/ERT2 mice (in a C57BL/6 background) through induction with tamoxifen. The mice were then infected intranasally with 600,000 PFU of HSV-1. The survival rate of mice depleted of CSF1R (knockout [KO] mice) was significantly lower than that of wild-type (WT) mice (0% versus 67%). Brain viral titers and cytokine/chemokine levels were significantly higher in KO than in WT animals on day 6 p.i. Furthermore, increased infiltration of monocytes into the brains of WT mice was seen on day 6 p.i., but not in KO mice. Our results suggest that microglial cells are essential to control HSE at early stages of the disease and that the MCSF/CSF1R axis could be a therapeutic target to regulate their response to infection.IMPORTANCE Microglia appear to be one of the principal regulators of neuroinflammation in the central nervous system (CNS). An increasing number of studies have demonstrated that the activation of microglia could result in either beneficial or detrimental effects in different CNS disorders. Hence, the role of microglia during herpes simplex virus encephalitis (HSE) has not been fully characterized. Using experimental mouse models, we showed that an early activation of the MCSF/CSF1R axis improved the outcome of the disease, possibly by inducing a proliferation of microglia. In contrast, depletion of microglia before HSV-1 infection worsened the prognosis of HSE. Thus, an early microglial response followed by sustained infiltration of monocytes and T cells into the brain seem to be key components for a better clinical outcome. These data suggest that microglia could be a potential target for immunomodulatory strategies combined with antiviral therapy to better control the outcome of this devastating disease.
Collapse
|
50
|
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
|