1
|
Adams VR, Collins LB, Williams TI, Holmes J, Hess P, Atkins HM, Scheidemantle G, Liu X, Lodge M, Johnson AJ, Kennedy A. Myeloid cell MHC I expression drives CD8 + T cell activation in nonalcoholic steatohepatitis. Front Immunol 2024; 14:1302006. [PMID: 38274832 PMCID: PMC10808415 DOI: 10.3389/fimmu.2023.1302006] [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: 09/25/2023] [Accepted: 11/28/2023] [Indexed: 01/27/2024] Open
Abstract
Background & aims Activated CD8+ T cells are elevated in Nonalcoholic steatohepatitis (NASH) and are important for driving fibrosis and inflammation. Despite this, mechanisms of CD8+ T cell activation in NASH are largely limited. Specific CD8+ T cell subsets may become activated through metabolic signals or cytokines. However, studies in NASH have not evaluated the impact of antigen presentation or the involvement of specific antigens. Therefore, we determined if activated CD8+ T cells are dependent on MHC class I expression in NASH to regulate fibrosis and inflammation. Methods We used H2Kb and H2Db deficient (MHC I KO), Kb transgenic mice, and myeloid cell Kb deficient mice (LysM Kb KO) to investigate how MHC class I impacts CD8+ T cell function and NASH. Flow cytometry, gene expression, and histology were used to examine hepatic inflammation and fibrosis. The hepatic class I immunopeptidome was evaluated by mass spectrometry. Results In NASH, MHC class I isoform H2Kb was upregulated in myeloid cells. MHC I KO demonstrated protective effects against NASH-induced inflammation and fibrosis. Kb mice exhibited increased fibrosis in the absence of H2Db while LysM Kb KO mice showed protection against fibrosis but not inflammation. H2Kb restricted peptides identified a unique NASH peptide Ncf2 capable of CD8+ T cell activation in vitro. The Ncf2 peptide was not detected during fibrosis resolution. Conclusion These results suggest that activated hepatic CD8+ T cells are dependent on myeloid cell MHC class I expression in diet induced NASH to promote inflammation and fibrosis. Additionally, our studies suggest a role of NADPH oxidase in the production of Ncf2 peptide generation.
Collapse
Affiliation(s)
- Victoria R. Adams
- Department of Molecular and Structural Biochemistry, NC State University, Raleigh, NC, United States
| | - Leonard B. Collins
- Molecular Education, Technology and Research Innovation Center (METRIC), NC State University, Raleigh, NC, United States
| | - Taufika Islam Williams
- Molecular Education, Technology and Research Innovation Center (METRIC), NC State University, Raleigh, NC, United States
- Department of Chemistry, NC State University, Raleigh, NC, United States
| | - Jennifer Holmes
- College of Veterinary Medicine, NC State University, Raleigh, NC, United States
| | - Paul Hess
- College of Veterinary Medicine, NC State University, Raleigh, NC, United States
| | - Hannah M. Atkins
- Center for Human Health and Environment, NC State University, Raleigh, NC, United States
- Division of Comparative Medicine, UNC Chapel Hill, Chapel Hill, NC, United States
| | - Grace Scheidemantle
- Department of Molecular and Structural Biochemistry, NC State University, Raleigh, NC, United States
| | - Xiaojing Liu
- Department of Molecular and Structural Biochemistry, NC State University, Raleigh, NC, United States
| | - Mareca Lodge
- Department of Molecular and Structural Biochemistry, NC State University, Raleigh, NC, United States
| | - Aaron J. Johnson
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | - Arion Kennedy
- Department of Molecular and Structural Biochemistry, NC State University, Raleigh, NC, United States
| |
Collapse
|
2
|
Hülskötter K, Lühder F, Leitzen E, Flügel A, Baumgärtner W. CD28-signaling can be partially compensated in CD28-knockout mice but is essential for virus elimination in a murine model of multiple sclerosis. Front Immunol 2023; 14:1105432. [PMID: 37090733 PMCID: PMC10113529 DOI: 10.3389/fimmu.2023.1105432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/20/2023] [Indexed: 04/08/2023] Open
Abstract
The intracerebral infection of mice with Theiler’s murine encephalomyelitis virus (TMEV) represents a well-established animal model for multiple sclerosis (MS). Because CD28 is the main co-stimulatory molecule for the activation of T cells, we wanted to investigate its impact on the course of the virus infection as well as on a potential development of autoimmunity as seen in susceptible mouse strains for TMEV. In the present study, 5 weeks old mice on a C57BL/6 background with conventional or tamoxifen-induced, conditional CD28-knockout were infected intracerebrally with TMEV-BeAn. In the acute phase at 14 days post TMEV-infection (dpi), both CD28-knockout strains showed virus spread within the central nervous system (CNS) as an uncommon finding in C57BL/6 mice, accompanied by histopathological changes such as reduced microglial activation. In addition, the conditional, tamoxifen-induced CD28-knockout was associated with acute clinical deterioration and weight loss, which limited the observation period for this mouse strain to 14 dpi. In the chronic phase (42 and 147 dpi) of TMEV-infection, surprisingly only 33% of conventional CD28-knockout mice showed chronic TMEV-infection with loss of motor function concomitant with increased spinal cord inflammation, characterized by T- and B cell infiltration, microglial activation and astrogliosis at 33-42 dpi. Therefore, the clinical outcome largely depends on the time point of the CD28-knockout during development of the immune system. Whereas a fatal clinical outcome can already be observed in the early phase during TMEV-infection for conditional, tamoxifen-induced CD28-knockout mice, only one third of conventional CD28-knockout mice develop clinical symptoms later, accompanied by ongoing inflammation and an inability to clear the virus. However, the development of autoimmunity could not be observed in this C57BL/6 TMEV model irrespective of the time point of CD28 deletion.
Collapse
Affiliation(s)
- Kirsten Hülskötter
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Fred Lühder
- Institute for Neuroimmunology and Multiple Sclerosis Research (IMSF), University Medical Center Goettingen, Goettingen, Germany
| | - Eva Leitzen
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Alexander Flügel
- Institute for Neuroimmunology and Multiple Sclerosis Research (IMSF), University Medical Center Goettingen, Goettingen, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
- *Correspondence: Wolfgang Baumgärtner,
| |
Collapse
|
3
|
Gilli F, Ceccarelli A. Magnetic resonance imaging approaches for studying mouse models of multiple sclerosis: A mini review. J Neurosci Res 2023. [DOI: 10.1002/jnr.25193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 01/30/2023] [Accepted: 03/10/2023] [Indexed: 04/03/2023]
Affiliation(s)
- Francesca Gilli
- Department of Neurology, Dartmouth Hitchcock Medical Center Geisel School of Medicine at Dartmouth Lebanon New Hampshire USA
| | - Antonia Ceccarelli
- Department of Neurology EpiCURA Centre Hospitalier Ath Belgium
- Hearthrhythmanagement, UZB Brussels Belgium
| |
Collapse
|
4
|
Lorrey SJ, Waibl Polania J, Wachsmuth LP, Hoyt-Miggelbrink A, Tritz ZP, Edwards R, Wolf DM, Johnson AJ, Fecci PE, Ayasoufi K. Systemic immune derangements are shared across various CNS pathologies and reflect novel mechanisms of immune privilege. Neurooncol Adv 2023; 5:vdad035. [PMID: 37207119 PMCID: PMC10191195 DOI: 10.1093/noajnl/vdad035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2023] Open
Abstract
Background The nervous and immune systems interact in a reciprocal manner, both under physiologic and pathologic conditions. Literature spanning various CNS pathologies including brain tumors, stroke, traumatic brain injury and de-myelinating diseases describes a number of associated systemic immunologic changes, particularly in the T-cell compartment. These immunologic changes include severe T-cell lymphopenia, lymphoid organ contraction, and T-cell sequestration within the bone marrow. Methods We performed an in-depth systematic review of the literature and discussed pathologies that involve brain insults and systemic immune derangements. Conclusions In this review, we propose that the same immunologic changes hereafter termed 'systemic immune derangements', are present across CNS pathologies and may represent a novel, systemic mechanism of immune privilege for the CNS. We further demonstrate that systemic immune derangements are transient when associated with isolated insults such as stroke and TBI but persist in the setting of chronic CNS insults such as brain tumors. Systemic immune derangements have vast implications for informed treatment modalities and outcomes of various neurologic pathologies.
Collapse
Affiliation(s)
- Selena J Lorrey
- Department of Immunology, Duke University, Durham, NC, USA
- Brain Tumor Immunotherapy Program, Duke University, Durham, NC, USA
| | - Jessica Waibl Polania
- Brain Tumor Immunotherapy Program, Duke University, Durham, NC, USA
- Department of Pathology, Duke University, Durham, NC, USA
| | - Lucas P Wachsmuth
- Brain Tumor Immunotherapy Program, Duke University, Durham, NC, USA
- Department of Pathology, Duke University, Durham, NC, USA
- Medical Scientist Training Program, Duke University, Durham, NC, USA
| | - Alexandra Hoyt-Miggelbrink
- Brain Tumor Immunotherapy Program, Duke University, Durham, NC, USA
- Department of Pathology, Duke University, Durham, NC, USA
| | | | - Ryan Edwards
- Brain Tumor Immunotherapy Program, Duke University, Durham, NC, USA
| | - Delaney M Wolf
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | | | - Peter E Fecci
- Department of Immunology, Duke University, Durham, NC, USA
- Brain Tumor Immunotherapy Program, Duke University, Durham, NC, USA
- Department of Pathology, Duke University, Durham, NC, USA
- Department of Neurosurgery, Duke University, Durham, NC, USA
| | | |
Collapse
|
5
|
C-type lectin receptor DCIR contributes to hippocampal injury in acute neurotropic virus infection. Sci Rep 2021; 11:23819. [PMID: 34893671 PMCID: PMC8664856 DOI: 10.1038/s41598-021-03201-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 11/26/2021] [Indexed: 11/16/2022] Open
Abstract
Neurotropic viruses target the brain and contribute to neurologic diseases. C-type lectin receptors (CLRs) are pattern recognition receptors that recognize carbohydrate structures on endogenous molecules and pathogens. The myeloid CLR dendritic cell immunoreceptor (DCIR) is expressed by antigen presenting cells and mediates inhibitory intracellular signalling. To investigate the effect of DCIR on neurotropic virus infection, mice were infected experimentally with Theiler’s murine encephalomyelitis virus (TMEV). Brain tissue of TMEV-infected C57BL/6 mice and DCIR−/− mice were analysed by histology, immunohistochemistry and RT-qPCR, and spleen tissue by flow cytometry. To determine the impact of DCIR deficiency on T cell responses upon TMEV infection in vitro, antigen presentation assays were utilised. Genetic DCIR ablation in C57BL/6 mice was associated with an ameliorated hippocampal integrity together with reduced cerebral cytokine responses and reduced TMEV loads in the brain. Additionally, absence of DCIR favoured increased peripheral cytotoxic CD8+ T cell responses following TMEV infection. Co-culture experiments revealed that DCIR deficiency enhances the activation of antigen-specific CD8+ T cells by virus-exposed dendritic cells (DCs), indicated by increased release of interleukin-2 and interferon-γ. Results suggest that DCIR deficiency has a supportive influence on antiviral immune mechanisms, facilitating virus control in the brain and ameliorates neuropathology during acute neurotropic virus infection.
Collapse
|
6
|
Goddery EN, Fain CE, Lipovsky CG, Ayasoufi K, Yokanovich LT, Malo CS, Khadka RH, Tritz ZP, Jin F, Hansen MJ, Johnson AJ. Microglia and Perivascular Macrophages Act as Antigen Presenting Cells to Promote CD8 T Cell Infiltration of the Brain. Front Immunol 2021; 12:726421. [PMID: 34526998 PMCID: PMC8435747 DOI: 10.3389/fimmu.2021.726421] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/09/2021] [Indexed: 01/01/2023] Open
Abstract
CD8 T cell infiltration of the central nervous system (CNS) is necessary for host protection but contributes to neuropathology. Antigen presenting cells (APCs) situated at CNS borders are thought to mediate T cell entry into the parenchyma during neuroinflammation. The identity of the CNS-resident APC that presents antigen via major histocompatibility complex (MHC) class I to CD8 T cells is unknown. Herein, we characterize MHC class I expression in the naïve and virally infected brain and identify microglia and macrophages (CNS-myeloid cells) as APCs that upregulate H-2Kb and H-2Db upon infection. Conditional ablation of H-2Kb and H-2Db from CNS-myeloid cells allowed us to determine that antigen presentation via H-2Db, but not H-2Kb, was required for CNS immune infiltration during Theiler's murine encephalomyelitis virus (TMEV) infection and drives brain atrophy as a consequence of infection. These results demonstrate that CNS-myeloid cells are key APCs mediating CD8 T cell brain infiltration.
Collapse
Affiliation(s)
- Emma N. Goddery
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Cori E. Fain
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Chloe G. Lipovsky
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | | | - Lila T. Yokanovich
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Courtney S. Malo
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Roman H. Khadka
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Zachariah P. Tritz
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Fang Jin
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | | | - Aaron J. Johnson
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
7
|
Ayasoufi K, Pfaller CK, Evgin L, Khadka RH, Tritz ZP, Goddery EN, Fain CE, Yokanovich LT, Himes BT, Jin F, Zheng J, Schuelke MR, Hansen MJ, Tung W, Parney IF, Pease LR, Vile RG, Johnson AJ. Brain cancer induces systemic immunosuppression through release of non-steroid soluble mediators. Brain 2020; 143:3629-3652. [PMID: 33253355 PMCID: PMC7954397 DOI: 10.1093/brain/awaa343] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 08/08/2020] [Accepted: 08/11/2020] [Indexed: 01/09/2023] Open
Abstract
Immunosuppression of unknown aetiology is a hallmark feature of glioblastoma and is characterized by decreased CD4 T-cell counts and downregulation of major histocompatibility complex class II expression on peripheral blood monocytes in patients. This immunosuppression is a critical barrier to the successful development of immunotherapies for glioblastoma. We recapitulated the immunosuppression observed in glioblastoma patients in the C57BL/6 mouse and investigated the aetiology of low CD4 T-cell counts. We determined that thymic involution was a hallmark feature of immunosuppression in three distinct models of brain cancer, including mice harbouring GL261 glioma, B16 melanoma, and in a spontaneous model of diffuse intrinsic pontine glioma. In addition to thymic involution, we determined that tumour growth in the brain induced significant splenic involution, reductions in peripheral T cells, reduced MHC II expression on blood leucocytes, and a modest increase in bone marrow resident CD4 T cells. Using parabiosis we report that thymic involution, declines in peripheral T-cell counts, and reduced major histocompatibility complex class II expression levels were mediated through circulating blood-derived factors. Conversely, T-cell sequestration in the bone marrow was not governed through circulating factors. Serum isolated from glioma-bearing mice potently inhibited proliferation and functions of T cells both in vitro and in vivo. Interestingly, the factor responsible for immunosuppression in serum is non-steroidal and of high molecular weight. Through further analysis of neurological disease models, we determined that the immunosuppression was not unique to cancer itself, but rather occurs in response to brain injury. Non-cancerous acute neurological insults also induced significant thymic involution and rendered serum immunosuppressive. Both thymic involution and serum-derived immunosuppression were reversible upon clearance of brain insults. These findings demonstrate that brain cancers cause multifaceted immunosuppression and pinpoint circulating factors as a target of intervention to restore immunity.
Collapse
Affiliation(s)
| | - Christian K Pfaller
- Mayo Clinic Department of Molecular Medicine, Rochester, MN, USA
- Paul-Ehrlich-Institute, Division of Veterinary Medicine, Langen, Germany
| | - Laura Evgin
- Mayo Clinic Department of Molecular Medicine, Rochester, MN, USA
| | - Roman H Khadka
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Zachariah P Tritz
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Emma N Goddery
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Cori E Fain
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Lila T Yokanovich
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Benjamin T Himes
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Department of Neurologic Surgery, Rochester, MN, USA
| | - Fang Jin
- Mayo Clinic Department of Immunology, Rochester, MN, USA
| | - Jiaying Zheng
- Mayo Clinic Department of Molecular Medicine, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Matthew R Schuelke
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Department of Molecular Medicine, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
- Department of Immunology, Mayo Clinic Medical Scientist Training Program, Rochester, Minnesota, USA
| | | | - Wesley Tung
- Mayo Clinic Department of Immunology, Rochester, MN, USA
| | - Ian F Parney
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Department of Neurologic Surgery, Rochester, MN, USA
| | - Larry R Pease
- Mayo Clinic Department of Immunology, Rochester, MN, USA
| | - Richard G Vile
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Department of Molecular Medicine, Rochester, MN, USA
| | - Aaron J Johnson
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Department of Molecular Medicine, Rochester, MN, USA
- Mayo Clinic Department of Neurology, Rochester, MN, USA
| |
Collapse
|
8
|
Gerhauser I, Hansmann F, Ciurkiewicz M, Löscher W, Beineke A. Facets of Theiler's Murine Encephalomyelitis Virus-Induced Diseases: An Update. Int J Mol Sci 2019; 20:ijms20020448. [PMID: 30669615 PMCID: PMC6358740 DOI: 10.3390/ijms20020448] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/15/2019] [Accepted: 01/18/2019] [Indexed: 12/31/2022] Open
Abstract
Theiler’s murine encephalomyelitis virus (TMEV), a naturally occurring, enteric pathogen of mice is a Cardiovirus of the Picornaviridae family. Low neurovirulent TMEV strains such as BeAn cause a severe demyelinating disease in susceptible SJL mice following intracerebral infection. Furthermore, TMEV infections of C57BL/6 mice cause acute polioencephalitis initiating a process of epileptogenesis that results in spontaneous recurrent epileptic seizures in approximately 50% of affected mice. Moreover, C3H mice develop cardiac lesions after an intraperitoneal high-dose application of TMEV. Consequently, TMEV-induced diseases are widely used as animal models for multiple sclerosis, epilepsy, and myocarditis. The present review summarizes morphological lesions and pathogenic mechanisms triggered by TMEV with a special focus on the development of hippocampal degeneration and seizures in C57BL/6 mice as well as demyelination in the spinal cord in SJL mice. Furthermore, a detailed description of innate and adaptive immune responses is given. TMEV studies provide novel insights into the complexity of organ- and mouse strain-specific immunopathology and help to identify factors critical for virus persistence.
Collapse
Affiliation(s)
- Ingo Gerhauser
- Department of Pathology, University of Veterinary Medicine, Bünteweg 17, 30559 Hannover, Germany.
| | - Florian Hansmann
- Department of Pathology, University of Veterinary Medicine, Bünteweg 17, 30559 Hannover, Germany.
- Center for System Neuroscience, 30559 Hannover, Germany.
| | - Malgorzata Ciurkiewicz
- Department of Pathology, University of Veterinary Medicine, Bünteweg 17, 30559 Hannover, Germany.
- Center for System Neuroscience, 30559 Hannover, Germany.
| | - Wolfgang Löscher
- Center for System Neuroscience, 30559 Hannover, Germany.
- Department of Pharmacology, University of Veterinary Medicine, Bünteweg 17, 30559 Hannover, Germany.
| | - Andreas Beineke
- Department of Pathology, University of Veterinary Medicine, Bünteweg 17, 30559 Hannover, Germany.
- Center for System Neuroscience, 30559 Hannover, Germany.
| |
Collapse
|
9
|
Klein RS, Hunter CA. Protective and Pathological Immunity during Central Nervous System Infections. Immunity 2017; 46:891-909. [PMID: 28636958 PMCID: PMC5662000 DOI: 10.1016/j.immuni.2017.06.012] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/05/2017] [Accepted: 06/05/2017] [Indexed: 02/08/2023]
Abstract
The concept of immune privilege of the central nervous system (CNS) has dominated the study of inflammatory processes in the brain. However, clinically relevant models have highlighted that innate pathways limit pathogen invasion of the CNS and adaptive immunity mediates control of many neural infections. As protective responses can result in bystander damage, there are regulatory mechanisms that balance protective and pathological inflammation, but these mechanisms might also allow microbial persistence. The focus of this review is to consider the host-pathogen interactions that influence neurotropic infections and to highlight advances in our understanding of innate and adaptive mechanisms of resistance as key determinants of the outcome of CNS infection. Advances in these areas have broadened our comprehension of how the immune system functions in the brain and can readily overcome immune privilege.
Collapse
Affiliation(s)
- Robyn S Klein
- Departments of Medicine, Pathology and Immunology, Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|