1
|
Moseley CE, Virupakshaiah A, Forsthuber TG, Steinman L, Waubant E, Zamvil SS. MOG CNS Autoimmunity and MOGAD. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200275. [PMID: 38996203 PMCID: PMC11256982 DOI: 10.1212/nxi.0000000000200275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 04/30/2024] [Indexed: 07/14/2024]
Abstract
At one time considered a possible form of neuromyelitis optica (NMO) spectrum disorder (NMOSD), it is now accepted that myelin oligodendrocyte glycoprotein (MOG) antibody (Ab)-associated disorder (MOGAD) is a distinct entity from either NMO or multiple sclerosis (MS) and represents a broad spectrum of clinical phenotypes. Whereas Abs targeting aquaporin-4 (AQP4) in NMO are pathogenic, the extent that anti-MOG Abs contribute to CNS damage in MOGAD is unclear. Both AQP4-specific Abs in NMO and MOG-specific Abs in MOGAD are predominantly IgG1, a T cell-dependent immunoglobulin (Ig) subclass. Key insights in neuroimmunology and MOGAD pathogenesis have been learned from MOG experimental autoimmune encephalomyelitis (EAE), described 2 decades before the term MOGAD was introduced. MOG-specific T cells are required in MOG EAE, and while anti-MOG Abs can exacerbate EAE and CNS demyelination, those Abs are neither necessary nor sufficient to cause EAE. Knowledge regarding the spectrum of MOGAD clinical and radiologic presentations is advancing rapidly, yet our grasp of MOGAD pathogenesis is incomplete. Understanding both the humoral and cellular immunology of MOGAD has implications for diagnosis, treatment, and prognosis.
Collapse
Affiliation(s)
- Carson E Moseley
- From the Department of Neurology (C.E.M., A.V., E.W., S.S.Z.), Weill Institute for Neurosciences, University of California, San Francisco; Department of Molecular Microbiology and Immunology (T.G.F.), University of Texas at San Antonio; Department of Neurology and Neurological Science (L.S.), Stanford University; and Program in Immunology (S.S.Z.), University of California, San Francisco, CA
| | - Akash Virupakshaiah
- From the Department of Neurology (C.E.M., A.V., E.W., S.S.Z.), Weill Institute for Neurosciences, University of California, San Francisco; Department of Molecular Microbiology and Immunology (T.G.F.), University of Texas at San Antonio; Department of Neurology and Neurological Science (L.S.), Stanford University; and Program in Immunology (S.S.Z.), University of California, San Francisco, CA
| | - Thomas G Forsthuber
- From the Department of Neurology (C.E.M., A.V., E.W., S.S.Z.), Weill Institute for Neurosciences, University of California, San Francisco; Department of Molecular Microbiology and Immunology (T.G.F.), University of Texas at San Antonio; Department of Neurology and Neurological Science (L.S.), Stanford University; and Program in Immunology (S.S.Z.), University of California, San Francisco, CA
| | - Lawrence Steinman
- From the Department of Neurology (C.E.M., A.V., E.W., S.S.Z.), Weill Institute for Neurosciences, University of California, San Francisco; Department of Molecular Microbiology and Immunology (T.G.F.), University of Texas at San Antonio; Department of Neurology and Neurological Science (L.S.), Stanford University; and Program in Immunology (S.S.Z.), University of California, San Francisco, CA
| | - Emmanuelle Waubant
- From the Department of Neurology (C.E.M., A.V., E.W., S.S.Z.), Weill Institute for Neurosciences, University of California, San Francisco; Department of Molecular Microbiology and Immunology (T.G.F.), University of Texas at San Antonio; Department of Neurology and Neurological Science (L.S.), Stanford University; and Program in Immunology (S.S.Z.), University of California, San Francisco, CA
| | - Scott S Zamvil
- From the Department of Neurology (C.E.M., A.V., E.W., S.S.Z.), Weill Institute for Neurosciences, University of California, San Francisco; Department of Molecular Microbiology and Immunology (T.G.F.), University of Texas at San Antonio; Department of Neurology and Neurological Science (L.S.), Stanford University; and Program in Immunology (S.S.Z.), University of California, San Francisco, CA
| |
Collapse
|
2
|
Huang N, Winans T, Wyman B, Oaks Z, Faludi T, Choudhary G, Lai ZW, Lewis J, Beckford M, Duarte M, Krakko D, Patel A, Park J, Caza T, Sadeghzadeh M, Morel L, Haas M, Middleton F, Banki K, Perl A. Rab4A-directed endosome traffic shapes pro-inflammatory mitochondrial metabolism in T cells via mitophagy, CD98 expression, and kynurenine-sensitive mTOR activation. Nat Commun 2024; 15:2598. [PMID: 38519468 PMCID: PMC10960037 DOI: 10.1038/s41467-024-46441-2] [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: 02/23/2023] [Accepted: 02/28/2024] [Indexed: 03/25/2024] Open
Abstract
Activation of the mechanistic target of rapamycin (mTOR) is a key metabolic checkpoint of pro-inflammatory T-cell development that contributes to the pathogenesis of autoimmune diseases, such as systemic lupus erythematosus (SLE), however, the underlying mechanisms remain poorly understood. Here, we identify a functional role for Rab4A-directed endosome traffic in CD98 receptor recycling, mTOR activation, and accumulation of mitochondria that connect metabolic pathways with immune cell lineage development and lupus pathogenesis. Based on integrated analyses of gene expression, receptor traffic, and stable isotope tracing of metabolic pathways, constitutively active Rab4AQ72L exerts cell type-specific control over metabolic networks, dominantly impacting CD98-dependent kynurenine production, mTOR activation, mitochondrial electron transport and flux through the tricarboxylic acid cycle and thus expands CD4+ and CD3+CD4-CD8- double-negative T cells over CD8+ T cells, enhancing B cell activation, plasma cell development, antinuclear and antiphospholipid autoantibody production, and glomerulonephritis in lupus-prone mice. Rab4A deletion in T cells and pharmacological mTOR blockade restrain CD98 expression, mitochondrial metabolism and lineage skewing and attenuate glomerulonephritis. This study identifies Rab4A-directed endosome traffic as a multilevel regulator of T cell lineage specification during lupus pathogenesis.
Collapse
Affiliation(s)
- Nick Huang
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
- Department of Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Thomas Winans
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
- Department of Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Brandon Wyman
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
- Department of Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Zachary Oaks
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
- Department of Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Tamas Faludi
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Gourav Choudhary
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
- Department of Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Zhi-Wei Lai
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Joshua Lewis
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Miguel Beckford
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Manuel Duarte
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Daniel Krakko
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Akshay Patel
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
- Department of Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Joy Park
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
- Department of Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Tiffany Caza
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Mahsa Sadeghzadeh
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
- Department of Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Laurence Morel
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Mark Haas
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Frank Middleton
- Department of Neuroscience and Physiology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Katalin Banki
- Department of Pathology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Andras Perl
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA.
- Department of Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA.
- Department of Microbiology and Immunology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA.
| |
Collapse
|
3
|
Orian JM, Maxwell DL, Lim VJT. Active Induction of a Multiple Sclerosis-Like Disease in Common Laboratory Mouse Strains. Methods Mol Biol 2024; 2746:179-200. [PMID: 38070090 DOI: 10.1007/978-1-0716-3585-8_15] [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] [Indexed: 12/18/2023]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a neuroinflammatory disease with facets in common with multiple sclerosis (MS). It is induced in susceptible mammalian species, with rodents as the preferred hosts, and has been used for decades as a model to investigate the immunopathogenesis of MS as well as for preclinical evaluation of candidate MS therapeutics. Most commonly, EAE is generated by active immunization with central nervous system (CNS) antigens, such as whole CNS homogenate, myelin proteins, or peptides derived from these proteins. However, EAE actually represents a spectrum of diseases in which specific combinations of host/CNS antigen exhibit defined clinical profiles, each associated with unique immunological and pathological features. Similar to MS, EAE is a complex disease where development and progression are also modulated by environmental factors; therefore, the establishment of any given EAE variant can be challenging and requires careful optimization. Here, we describe protocols for three EAE variants, successfully generated in our laboratory, and provide additional information as to how to maintain their unique features and reproducibility.
Collapse
Affiliation(s)
- Jacqueline M Orian
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia.
| | - Dain L Maxwell
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia
| | - Vernise J T Lim
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| |
Collapse
|
4
|
Dedoni S, Scherma M, Camoglio C, Siddi C, Dazzi L, Puliga R, Frau J, Cocco E, Fadda P. An overall view of the most common experimental models for multiple sclerosis. Neurobiol Dis 2023:106230. [PMID: 37453561 DOI: 10.1016/j.nbd.2023.106230] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 07/01/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023] Open
Abstract
Multiple sclerosis (MS) is a complex chronic disease with an unknown etiology. It is considered an inflammatory demyelinating and neurodegenerative disorder of the central nervous system (CNS) characterized, in most cases, by an unpredictable onset of relapse and remission phases. The disease generally starts in subjects under 40; it has a higher incidence in women and is described as a multifactorial disorder due to the interaction between genetic and environmental risk factors. Unfortunately, there is currently no definitive cure for MS. Still, therapies can modify the disease's natural history, reducing the relapse rate and slowing the progression of the disease or managing symptoms. The limited access to human CNS tissue slows down. It limits the progression of research on MS. This limit has been partially overcome over the years by developing various experimental models to study this disease. Animal models of autoimmune demyelination, such as experimental autoimmune encephalomyelitis (EAE) and viral and toxin or transgenic MS models, represent the most significant part of MS research approaches. These models have now been complemented by ex vivo studies, using organotypic brain slice cultures and in vitro, through induced Pluripotent Stem cells (iPSCs). We will discuss which clinical features of the disorders might be reproduced and investigated in vivo, ex vivo, and in vitro in models commonly used in MS research to understand the processes behind the neuropathological events occurring in the CNS of MS patients. The primary purpose of this review is to give the reader a global view of the main paradigms used in MS research, spacing from the classical animal models to transgenic mice and 2D and 3D cultures.
Collapse
Affiliation(s)
- S Dedoni
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - M Scherma
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - C Camoglio
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - C Siddi
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy
| | - L Dazzi
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato (Cagliari), Italy.
| | - R Puliga
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato (Cagliari), Italy.
| | - J Frau
- Regional Multiple Sclerosis Center, ASSL Cagliari, ATS Sardegna, Italy
| | - E Cocco
- Regional Multiple Sclerosis Center, ASSL Cagliari, ATS Sardegna, Italy; Department Medical Science and Public Health, University of Cagliari, Italy.
| | - P Fadda
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy; Neuroscience Institute, Section of Cagliari, National Research Council of Italy (CNR), Cagliari, Italy.
| |
Collapse
|
5
|
Abdolmaleki A, Kondori BJ, Raei M, Ghaleh HEG. Cell therapy procedure using anti-inflammatory macrophage M2 can potentially reduce Clinical Score in animals with Experimental Autoimmune Encephalomyelitis: A preclinical systematic review and meta-analysis study. Fundam Clin Pharmacol 2023; 37:215-225. [PMID: 36300567 DOI: 10.1111/fcp.12844] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/02/2022] [Accepted: 10/17/2022] [Indexed: 11/07/2022]
Abstract
Macrophage M2 (MP2)-based cell therapy is a novel medicinal treatment for animals with Experimental Autoimmune Encephalomyelitis (EAE) as an experimental model of multiple sclerosis (MS). This systematic review and meta-analysis study was designed to assess the overall therapeutic effects of MP2 cell therapy on Clinical Score and motor impairment in EAE-induced animals. All experiments on MP2 cell therapy in animals with EAE were gathered (by October 2, 2022) from English (PubMed, Scopus, WoS, Science Direct, and ISC) and Persian (MagIran and SID) databases. The searching strategy was designed using "Experimental Autoimmune Encephalomyelitis," "Multiple Sclerosis," and "Macrophage M2" keywords. Following primary and secondary screenings, eligible papers were selected based on the PRISMA 2020 guideline, and the study quality was assessed using the Animal Research: Reporting of In Vivo Experiments (ARRIVE) checklist. The difference in means of Clinical Score (score 0-5) as the effect size (ES) was analyzed based on the random effect model (CMA software, v.2). Subgrouping (EAE phases of Onset, Peak, and Recovery) was applied, and I2 index was used to assess the heterogeneity index. Publication bias and sensitivity indices were also evaluated. P < 0.05 was considered significant, and the confidence interval (CI) was determined 95%. Among 22 gathered papers, medium to high quality studies were selected for meta-analysis. Difference in means, P value, and I2 for Onset, Peak, and Recovery phases were 0.082 (CI95%: -0.323-0.159, P value: 0.504, I2 : 67.961%), -0.606 (CI95%: -1.518 to -0.305, P value: 0.192, I2 : 96.070%), and -1.103 (CI95%: -1.390 to -0.816, P value: 0.000, I2 : 30.880%), respectively and Overall Effect was found -0.509 (CI95%: -0.689 to -0.328, P value < 0.001). Also, P value (two-tailed) indices for publication bias were 0.366 and 0.583 for Egger's regression intercept and Begg rank correlation, respectively. The P value for sensitivity was detected 0.003. Cell therapy procedure using MP2 can potentially alleviate the Clinical Scores Index and correct the motor defects in Recovery phase of EAE animals. In healthy mice, the brain and myelin surrounding neurons are in a healthy and physiological state (1). To evaluate MS in humans, it is necessary to model this type of disease in animals using EAE procedure through subcutaneous injection of CFA, MOG35-55 , MT, and Pert. Thus, inflammation and autoimmunity occur, which finally lead to myelin destruction and motor symptoms (2). By aspiration of progenitor cells available in bone marrow, the MP2 can be isolated and cultured. By activation of these types of cells, a rich collection of MP2 can be prepared for the cell-therapy process (3). After injection through the tail vein or intra-peritoneal procedure, these cells can be located in CNS through crossing from the BBB. They begin their anti-inflammatory activities and help repair the damaged myelin (4). Eventually, the clinical symptoms can be modified considerably, and the animal motor function improves (5). CFA, complete Freund's adjuvant; MOG35-55 , myelin oligodendrocyte glycoprotein; MT, Mycobacterium tuberculosis; Pert, pertussis; EAE, Experimental Autoimmune Encephalomyelitis; BM, bone marrow; MP2, macrophage M2; and BBB, blood brain barrier.
Collapse
Affiliation(s)
- Amir Abdolmaleki
- Department of Anatomical Sciences, Medical School, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Bahman Jalali Kondori
- Department of Anatomical Sciences, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran.,Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mehdi Raei
- Health Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
6
|
Amatruda M, Harris K, Matis A, Davies AL, McElroy D, Clark M, Linington C, Desai R, Smith KJ. Oxygen treatment reduces neurological deficits and demyelination in two animal models of multiple sclerosis. Neuropathol Appl Neurobiol 2023; 49:e12868. [PMID: 36520661 PMCID: PMC10107096 DOI: 10.1111/nan.12868] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 11/07/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022]
Abstract
AIMS The objective of the study is to explore the importance of tissue hypoxia in causing neurological deficits and demyelination in the inflamed CNS, and the value of inspiratory oxygen treatment, using both active and passive experimental autoimmune encephalomyelitis (EAE). METHODS Normobaric oxygen treatment was administered to Dark Agouti rats with either active or passive EAE, compared with room air-treated, and naïve, controls. RESULTS Severe neurological deficits in active EAE were significantly improved after just 1 h of breathing approximately 95% oxygen. The improvement was greater and more persistent when oxygen was applied either prophylactically (from immunisation for 23 days), or therapeutically from the onset of neurological deficits for 24, 48, or 72 h. Therapeutic oxygen for 72 h significantly reduced demyelination and the integrated stress response in oligodendrocytes at the peak of disease, and protected from oligodendrocyte loss, without evidence of increased oxidative damage. T-cell infiltration and cytokine expression in the spinal cord remained similar to that in untreated animals. The severe neurological deficit of animals with passive EAE occurred in conjunction with spinal hypoxia and was significantly reduced by oxygen treatment initiated before their onset. CONCLUSIONS Severe neurological deficits in both active and passive EAE can be caused by hypoxia and reduced by oxygen treatment. Oxygen treatment also reduces demyelination in active EAE, despite the autoimmune origin of the disease.
Collapse
Affiliation(s)
- Mario Amatruda
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, UK.,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kate Harris
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, UK
| | - Alina Matis
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, UK
| | - Andrew L Davies
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, UK
| | - Daniel McElroy
- Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, Glasgow Biomedical Research Centre, Glasgow, UK
| | - Michael Clark
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, UK
| | - Christopher Linington
- Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, Glasgow Biomedical Research Centre, Glasgow, UK
| | - Roshni Desai
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, UK
| | - Kenneth J Smith
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
7
|
Mey GM, Mahajan KR, DeSilva TM. Neurodegeneration in multiple sclerosis. WIREs Mech Dis 2023; 15:e1583. [PMID: 35948371 PMCID: PMC9839517 DOI: 10.1002/wsbm.1583] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/28/2022] [Accepted: 07/11/2022] [Indexed: 01/31/2023]
Abstract
Axonal loss in multiple sclerosis (MS) is a key component of disease progression and permanent neurologic disability. MS is a heterogeneous demyelinating and neurodegenerative disease of the central nervous system (CNS) with varying presentation, disease courses, and prognosis. Immunomodulatory therapies reduce the frequency and severity of inflammatory demyelinating events that are a hallmark of MS, but there is minimal therapy to treat progressive disease and there is no cure. Data from patients with MS, post-mortem histological analysis, and animal models of demyelinating disease have elucidated patterns of MS pathogenesis and underlying mechanisms of neurodegeneration. MRI and molecular biomarkers have been proposed to identify predictors of neurodegeneration and risk factors for disease progression. Early signs of axonal dysfunction have come to light including impaired mitochondrial trafficking, structural axonal changes, and synaptic alterations. With sustained inflammation as well as impaired remyelination, axons succumb to degeneration contributing to CNS atrophy and worsening of disease. These studies highlight the role of chronic demyelination in the CNS in perpetuating axonal loss, and the difficulty in promoting remyelination and repair amidst persistent inflammatory insult. Regenerative and neuroprotective strategies are essential to overcome this barrier, with early intervention being critical to rescue axonal integrity and function. The clinical and basic research studies discussed in this review have set the stage for identifying key propagators of neurodegeneration in MS, leading the way for neuroprotective therapeutic development. This article is categorized under: Immune System Diseases > Molecular and Cellular Physiology Neurological Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Gabrielle M. Mey
- Department of NeurosciencesLerner Research Institute, Cleveland Clinic Foundation, and Case Western Reserve UniversityClevelandOhioUSA
| | - Kedar R. Mahajan
- Department of NeurosciencesLerner Research Institute, Cleveland Clinic Foundation, and Case Western Reserve UniversityClevelandOhioUSA
- Mellen Center for MS Treatment and ResearchNeurological Institute, Cleveland Clinic FoundationClevelandOhioUSA
| | - Tara M. DeSilva
- Department of NeurosciencesLerner Research Institute, Cleveland Clinic Foundation, and Case Western Reserve UniversityClevelandOhioUSA
| |
Collapse
|
8
|
Saez-Calveras N, Brewster AL, Stuve O. The validity of animal models to explore the pathogenic role of the complement system in multiple sclerosis: A review. Front Mol Neurosci 2022; 15:1017484. [PMID: 36311030 PMCID: PMC9606595 DOI: 10.3389/fnmol.2022.1017484] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/26/2022] [Indexed: 11/26/2022] Open
Abstract
Animal models of multiple sclerosis (MS) have been extensively used to characterize the disease mechanisms in MS, as well as to identify potential pharmacologic targets for this condition. In recent years, the immune complement system has gained increased attention as an important effector in the pathogenesis of MS. Evidence from histological, serum, and CSF studies of patients supports an involvement of complement in both relapsing-remitting and progressive MS. In this review, we discuss the history and advances made on the use of MS animal models to profile the effects of the complement system in this condition. The first studies that explored the complement system in the context of MS used cobra venom factor (CVF) as a complement depleting agent in experimental autoimmune encephalomyelitis (EAE) Lewis rats. Since then, multiple mice and rat models of MS have revealed a role of C3 and the alternative complement cascade in the opsonization and phagocytosis of myelin by microglia and myeloid cells. Studies using viral vectors, genetic knockouts and pharmacologic complement inhibitors have also shown an effect of complement in synaptic loss. Antibody-mediated EAE models have revealed an involvement of the C1 complex and the classical complement as an effector of the humoral response in this disease. C1q itself may also be involved in modulating microglia activation and oligodendrocyte differentiation in these animals. In addition, animal and in vitro models have revealed that multiple complement factors may act as modulators of both the innate and adaptive immune responses. Finally, evidence gathered from mice models suggests that the membrane attack complex (MAC) may even exert protective roles in the chronic stages of EAE. Overall, this review summarizes the importance of MS animal models to better characterize the role of the complement system and guide future therapeutic approaches in this condition.
Collapse
Affiliation(s)
- Nil Saez-Calveras
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Neurology Section, Parkland Hospital, Dallas, TX, United States
| | - Amy L. Brewster
- Department of Biological Sciences, Southern Methodist University, Dallas, TX, United States
| | - Olaf Stuve
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Neurology Section, VA North Texas Health Care System, Dallas, TX, United States
- Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States
- *Correspondence: Olaf Stuve,
| |
Collapse
|
9
|
Fang M, Tang T, Qiu M, Xu X. Hedgehog Signaling in CNS Remyelination. Cells 2022; 11:cells11142260. [PMID: 35883703 PMCID: PMC9320235 DOI: 10.3390/cells11142260] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/14/2022] [Accepted: 07/20/2022] [Indexed: 12/14/2022] Open
Abstract
Remyelination is a fundamental repair process in the central nervous system (CNS) that is triggered by demyelinating events. In demyelinating diseases, oligodendrocytes (OLs) are targeted, leading to myelin loss, axonal damage, and severe functional impairment. While spontaneous remyelination often fails in the progression of demyelinating diseases, increased understanding of the mechanisms and identification of targets that regulate myelin regeneration becomes crucial. To date, several signaling pathways have been implicated in the remyelination process, including the Hedgehog (Hh) signaling pathway. This review summarizes the current data concerning the complicated roles of the Hh signaling pathway in the context of remyelination. We will highlight the open issues that have to be clarified prior to bringing molecules targeting the Hh signaling to demyelinating therapy.
Collapse
Affiliation(s)
- Minxi Fang
- Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China;
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tao Tang
- Department of Anatomy, Cell Biology & Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, USA;
| | - Mengsheng Qiu
- Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China;
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
- School of Basic Medicial Sciences, Hangzhou Normal University, Hangzhou 311121, China
- Correspondence: (M.Q.); (X.X.)
| | - Xiaofeng Xu
- Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China;
- Correspondence: (M.Q.); (X.X.)
| |
Collapse
|
10
|
Song YC, Liu CT, Lee HJ, Yen HR. Cordycepin prevents and ameliorates experimental autoimmune encephalomyelitis by inhibiting leukocyte infiltration and reducing neuroinflammation. Biochem Pharmacol 2022; 197:114918. [DOI: 10.1016/j.bcp.2022.114918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 11/30/2022]
|
11
|
Au KM, Tisch R, Wang AZ. Immune Checkpoint Ligand Bioengineered Schwann Cells as Antigen-Specific Therapy for Experimental Autoimmune Encephalomyelitis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107392. [PMID: 34775659 PMCID: PMC8813901 DOI: 10.1002/adma.202107392] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/08/2021] [Indexed: 05/05/2023]
Abstract
Failure to establish immune tolerance leads to the development of autoimmune disease. The ability to regulate autoreactive T cells without inducing systemic immunosuppression represents a major challenge in the development of new strategies to treat autoimmune disease. Here, a translational method for bioengineering programmed death-ligand 1 (PD-L1)- and cluster of differentiation 86 (CD86)-functionalized mouse Schwann cells (SCs) to prevent and ameliorate multiple sclerosis (MS) in established mouse models of chronic and relapsing-remitting experimental autoimmune encephalomyelitis (EAE) is described. It is shown that the intravenous (i.v.) administration of immune checkpoint ligand functionalized mouse SCs modifies the course of disease and ameliorates EAE. Further, it is found that such bioengineered mouse SCs inhibit the differentiation of myelin-specific helper T cells into pathogenic T helper type-1 (Th 1) and type-17 (Th 17) cells, promote the development of tolerogenic myelin-specific regulatory T (Treg ) cells, and resolve inflammatory central nervous system microenvironments without inducing systemic immunosuppression.
Collapse
Affiliation(s)
- Kin Man Au
- Laboratory of Nano- and Translational Medicine, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, 75230, USA
| | - Roland Tisch
- Department of Microbiology and Immunology School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Andrew Z Wang
- Laboratory of Nano- and Translational Medicine, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, 75230, USA
| |
Collapse
|
12
|
Nissen JC, Tsirka SE. Preclinical model of multiple sclerosis: Methods in autoimmune demyelination. Methods Cell Biol 2022; 168:67-86. [DOI: 10.1016/bs.mcb.2021.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
13
|
Laaker C, Hsu M, Fabry Z, Miller SD, Karpus WJ. Experimental Autoimmune Encephalomyelitis in the Mouse. Curr Protoc 2021; 1:e300. [PMID: 34870897 DOI: 10.1002/cpz1.300] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
This article details the materials and methods required for both active induction and adoptive transfer of experimental autoimmune encephalomyelitis (EAE) in the SJL mouse strain using intact proteins or peptides from the two major myelin proteins: proteolipid protein (PLP) and myelin basic protein (MBP). Additionally, active induction of EAE in the C57BL/6 strain using myelin oligodendrocyte glycoprotein (MOG) peptide is also discussed. Detailed materials and methods required for the purification of both PLP and MBP are described, and a protocol for isolating CNS-infiltrating lymphocytes in EAE mice is included. Modifications of the specified protocols may be necessary for efficient induction of active or adoptive EAE in other mouse strains. © 2021 Wiley Periodicals LLC. Basic Protocol: Active induction of EAE with PLP, MBP, and MOG protein or peptide Alternate Protocol: Adoptive induction of EAE with PLP-, MBP-, or MOG-specific lymphocytes Support Protocol 1: Purification of proteolipid protein Support Protocol 2: Purification of myelin basic protein Support Protocol 3: Isolation of CNS-infiltrating lymphocytes.
Collapse
Affiliation(s)
- Collin Laaker
- Department of Pathology and Lab Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Martin Hsu
- Department of Pathology and Lab Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Zsuzsanna Fabry
- Department of Pathology and Lab Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Stephen D Miller
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - William J Karpus
- Department of Pathology and Lab Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
14
|
Abstract
T helper (Th)17 cells are considered to contribute to inflammatory mechanisms in diseases such as multiple sclerosis (MS). However, the discussion persists regarding their true role in patients. Here, we visualized central nervous system (CNS) inflammatory processes in models of MS live in vivo and in MS brains and discovered that CNS-infiltrating Th17 cells form prolonged stable contact with oligodendrocytes. Strikingly, compared to Th2 cells, direct contact with Th17 worsened experimental demyelination, caused damage to human oligodendrocyte processes, and increased cell death. Importantly, we found that in comparison to Th2 cells, both human and murine Th17 cells express higher levels of the integrin CD29, which is linked to glutamate release pathways. Of note, contact of human Th17 cells with oligodendrocytes triggered release of glutamate, which induced cell stress and changes in biosynthesis of cholesterol and lipids, as revealed by single-cell RNA-sequencing analysis. Finally, exposure to glutamate decreased myelination, whereas blockade of CD29 preserved oligodendrocyte processes from Th17-mediated injury. Our data provide evidence for the direct and deleterious attack of Th17 cells on the myelin compartment and show the potential for therapeutic opportunities in MS.
Collapse
|
15
|
Dagkonaki A, Avloniti M, Evangelidou M, Papazian I, Kanistras I, Tseveleki V, Lampros F, Tselios T, Jensen LT, Möbius W, Ruhwedel T, Androutsou ME, Matsoukas J, Anagnostouli M, Lassmann H, Probert L. Mannan-MOG35-55 Reverses Experimental Autoimmune Encephalomyelitis, Inducing a Peripheral Type 2 Myeloid Response, Reducing CNS Inflammation, and Preserving Axons in Spinal Cord Lesions. Front Immunol 2020; 11:575451. [PMID: 33329540 PMCID: PMC7711156 DOI: 10.3389/fimmu.2020.575451] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/12/2020] [Indexed: 12/30/2022] Open
Abstract
CNS autoantigens conjugated to oxidized mannan (OM) induce antigen-specific T cell tolerance and protect mice against autoimmune encephalomyelitis (EAE). To investigate whether OM-peptides treat EAE initiated by human MHC class II molecules, we administered OM-conjugated murine myelin oligodendrocyte glycoprotein peptide 35-55 (OM-MOG) to humanized HLA-DR2b transgenic mice (DR2b.Ab°), which are susceptible to MOG-EAE. OM-MOG protected DR2b.Ab° mice against MOG-EAE by both prophylactic and therapeutic applications. OM-MOG reversed clinical symptoms, reduced spinal cord inflammation, demyelination, and neuronal damage in DR2b.Ab° mice, while preserving axons within lesions and inducing the expression of genes associated with myelin (Mbp) and neuron (Snap25) recovery in B6 mice. OM-MOG-induced tolerance was peptide-specific, not affecting PLP178-191-induced EAE or polyclonal T cell proliferation responses. OM-MOG-induced immune tolerance involved rapid induction of PD-L1- and IL-10-producing myeloid cells, increased expression of Chi3l3 (Ym1) in secondary lymphoid organs and characteristics of anergy in MOG-specific CD4+ T cells. The results show that OM-MOG treats MOG-EAE in a peptide-specific manner, across mouse/human MHC class II barriers, through induction of a peripheral type 2 myeloid cell response and T cell anergy, and suggest that OM-peptides might be useful for suppressing antigen-specific CD4+ T cell responses in the context of human autoimmune CNS demyelination.
Collapse
Affiliation(s)
- Anastasia Dagkonaki
- Laboratory of Molecular Genetics, Department of Immunology, Hellenic Pasteur Institute, Athens, Greece
| | - Maria Avloniti
- Laboratory of Molecular Genetics, Department of Immunology, Hellenic Pasteur Institute, Athens, Greece
| | - Maria Evangelidou
- Laboratory of Molecular Genetics, Department of Immunology, Hellenic Pasteur Institute, Athens, Greece
| | - Irini Papazian
- Laboratory of Molecular Genetics, Department of Immunology, Hellenic Pasteur Institute, Athens, Greece
| | - Ioannis Kanistras
- Laboratory of Molecular Genetics, Department of Immunology, Hellenic Pasteur Institute, Athens, Greece
| | - Vivian Tseveleki
- Laboratory of Molecular Genetics, Department of Immunology, Hellenic Pasteur Institute, Athens, Greece
| | - Fotis Lampros
- Laboratory of Molecular Genetics, Department of Immunology, Hellenic Pasteur Institute, Athens, Greece
| | | | - Lise Torp Jensen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Wiebke Möbius
- Electron Microscopy Core Unit, Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Torben Ruhwedel
- Electron Microscopy Core Unit, Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | | | - John Matsoukas
- Department of Chemistry, University of Patras, Patras, Greece
| | - Maria Anagnostouli
- Immunogenetics Laboratory, First Department of Neurology, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Hans Lassmann
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Lesley Probert
- Laboratory of Molecular Genetics, Department of Immunology, Hellenic Pasteur Institute, Athens, Greece
| |
Collapse
|
16
|
Maramai S, Brindisi M. Targeting Endocannabinoid Metabolism: an Arrow with Multiple Tips Against Multiple Sclerosis. ChemMedChem 2020; 15:1985-2003. [PMID: 32762071 DOI: 10.1002/cmdc.202000310] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 07/24/2020] [Indexed: 12/19/2022]
Abstract
Multiple sclerosis (MS) is a chronic, immune-mediated disease of the central nervous system. At present, there is no definitive cure, and the few available disease-modifying options display either poor efficacy or life-threatening side effects. There is clear evidence that relapsing-remitting clinical attacks in MS are driven by inflammatory demyelination and that the subsequent disease steps, being irresponsive to immunotherapy, result from neurodegeneration. The endocannabinoid system (ECS) stands halfway between three key pathomechanisms underlying MS, namely inflammation, neurodegeneration and oxidative stress, thus representing a kingpin for the identification of novel therapeutic targets in MS. This review summarizes the current state of the art in the field of endocannabinoid metabolism modulators and their in vivo effects on relevant animal models. We also highlight key molecular underpinnings of their therapeutic efficacy as well as the potential to turn them into promising clinical candidates.
Collapse
Affiliation(s)
- Samuele Maramai
- Department of Excellence of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro, 2, 53100, Siena, Italy
| | - Margherita Brindisi
- Department of Excellence of Pharmacy, University of Naples Federico II, Via D. Montesano, 49, 80131, Naples, Italy
| |
Collapse
|
17
|
Novel B cell-dependent multiple sclerosis model using extracellular domains of myelin proteolipid protein. Sci Rep 2020; 10:5011. [PMID: 32193439 PMCID: PMC7081236 DOI: 10.1038/s41598-020-61928-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 03/04/2020] [Indexed: 01/01/2023] Open
Abstract
Therapeutic success of B cell-targeting approaches in multiple sclerosis (MS) has intensified research into the pathogenic and regulatory roles these cells play in demyelinating disease. Dissecting the function of B cells in the MS mouse model experimental autoimmune encephalomyelitis (EAE) is largely confined to induction with either the myelin oligodendrocyte glycoprotein epitope MOG35-55 or the full-length recombinant human MOG protein, the latter representing the most-used B cell-dependent EAE model. There is a clear need to investigate B cell function in additional myelin antigen contexts. Unlike MOG35-55, where lack of B cells yields more severe disease, we show here that the immunodominant myelin proteolipid protein epitope (PLP178-191) elicited identical EAE in WT and μMT mice, suggesting an absence of B cell engagement by this peptide. We hypothesized that a longer PLP antigen may better engage B cells and designed a peptide encompassing the extracellular domains (ECD) of PLP. We demonstrate here that PLPECD-immunized B cell-deficient mice failed to exhibit EAE. In contrast, PLPECD induced EAE not only in WT mice, but in B cell-sufficient mice incapable of secreting antibodies, suggesting a predominant antigen presentation role. These results establish a novel, efficient B cell-dependent EAE model.
Collapse
|
18
|
Assessing the anterior visual pathway in optic neuritis: recent experimental and clinical aspects. Curr Opin Neurol 2020; 32:346-357. [PMID: 30694926 DOI: 10.1097/wco.0000000000000675] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Multiple sclerosis (MS) and related autoimmune disorders of the central nervous system such as neuromyelitis optica spectrum disorders (NMOSD) are characterized by chronic disability resulting from autoimmune neuroinflammation, with demyelination, astrocyte damage, impaired axonal transmission and neuroaxonal loss. Novel therapeutics stopping or reversing the progression of disability are still urgently warranted. This review addresses research on optic neuritis in preclinical experimental models and their translation to clinical trials. RECENT FINDINGS Optic neuritis can be used as paradigm for an MS relapse which can serve to evaluate the efficacy of novel therapeutics in clinical trials with a reasonable duration and cohort size. The advantage is the linear structure of the visual pathway allowing the assessment of visual function and retinal structure as highly sensitive outcome parameters. Experimental autoimmune encephalomyelitis is an inducible, inflammatory and demyelinating central nervous system disease extensively used as animal model of MS. Optic neuritis is part of the clinicopathological manifestations in a number of experimental autoimmune encephalomyelitis models. These have gained increasing interest for studies evaluating neuroprotective and/or remyelinating substances as longitudinal, visual and retinal readouts have become available. SUMMARY Translation of preclinical experiments, evaluating neuroprotective or remyelinating therapeutics to clinical studies is challenging. In-vivo readouts like optical coherence tomography, offers the possibility to transfer experimental study designs to clinical optic neuritis trials.
Collapse
|
19
|
Raju RM, Tsai LH. The complexity of neuroinflammation at single-cell resolution. Nat Rev Neurol 2020; 15:249-250. [PMID: 30858531 DOI: 10.1038/s41582-019-0165-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Ravikiran M Raju
- Division of Newborn Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Li-Huei Tsai
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
20
|
Ingested ACTH blocks Th17 production by inhibiting GALT IL-6. J Neurol Sci 2020; 409:116602. [PMID: 31812846 PMCID: PMC10394755 DOI: 10.1016/j.jns.2019.116602] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/19/2019] [Accepted: 11/26/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND EAE is an inflammatory autoimmune process of the CNS that resembles multiple sclerosis (MS) and provides a useful animal model for the evaluation of mechanisms of action for potential immunomodulatory therapies. Oral ACTH (adrenocorticotropic hormone) can decrease clinical disease, IL-17 and Th1-like encephalitogenic IFN-γ secretion and increase Treg frequency. The mechanism by which oral ACTH decreases inflammatory proteins and increases Treg cell frequencies is unknown. OBJECTIVE IL-6 is a pivotal cytokine in the gut that determines the relative frequencies of Th17 vs Treg cells. We examined whether oral ACTH inhibited IL-6 in the gut associated lymphoid tissue (GALT) in EAE. DESIGN/METHODS B6 mice were immunized with MOG peptide 35-55 and gavaged with scrambled ACTH (scrambled melanocyte stimulating hormone [scrambled α-MSH]) or ACTH 1-39 during ongoing disease. RESULTS Ingested (oral) ACTH inhibited ongoing clinical disease. In the lamina propria (LP) immune compartment, there were significantly less CD11b + IL-6 and IL-17 producing lymphocytes from ACTH fed mice compared to s-MSH fed mice. There was also a decrease in the frequency of IL-17 and IFN-γ producing spleen cells and an increase in CD4 + FoxP3+ Treg cell frequency in ACTH fed mice compared to s-MSH fed control spleens. There were less IFN-γ producing CNS lymphocytes in ACTH fed mice compared to s-MSH fed control CNS. CONCLUSIONS Ingested ACTH inhibits EAE clinical disease by inhibiting IL-6 in the GALT.
Collapse
|
21
|
Kalinin S, Meares GP, Lin SX, Pietruczyk EA, Saher G, Spieth L, Nave KA, Boullerne AI, Lutz SE, Benveniste EN, Feinstein DL. Liver kinase B1 depletion from astrocytes worsens disease in a mouse model of multiple sclerosis. Glia 2019; 68:600-616. [PMID: 31664743 PMCID: PMC7337013 DOI: 10.1002/glia.23742] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 09/19/2019] [Accepted: 10/05/2019] [Indexed: 12/15/2022]
Abstract
Liver kinase B1 (LKB1) is a ubiquitously expressed kinase involved in the regulation of cell metabolism, growth, and inflammatory activation. We previously reported that a single nucleotide polymorphism in the gene encoding LKB1 is a risk factor for multiple sclerosis (MS). Since astrocyte activation and metabolic function have important roles in regulating neuroinflammation and neuropathology, we examined the serine/threonine kinase LKB1 in astrocytes in a chronic experimental autoimmune encephalomyelitis mouse model of MS. To reduce LKB1, a heterozygous astrocyte-selective conditional knockout (het-cKO) model was used. While disease incidence was similar, disease severity was worsened in het-cKO mice. RNAseq analysis identified Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways enriched in het-cKO mice relating to mitochondrial function, confirmed by alterations in mitochondrial complex proteins and reductions in mRNAs related to astrocyte metabolism. Enriched pathways included major histocompatibility class II genes, confirmed by increases in MHCII protein in spinal cord and cerebellum of het-cKO mice. We observed increased numbers of CD4+ Th17 cells and increased neuronal damage in spinal cords of het-cKO mice, associated with reduced expression of choline acetyltransferase, accumulation of immunoglobulin-γ, and reduced expression of factors involved in motor neuron survival. In vitro, LKB1-deficient astrocytes showed reduced metabolic function and increased inflammatory activation. These data suggest that metabolic dysfunction in astrocytes, in this case due to LKB1 deficiency, can exacerbate demyelinating disease by loss of metabolic support and increase in the inflammatory environment.
Collapse
Affiliation(s)
- Sergey Kalinin
- Department of Anesthesiology, University of Illinois, Chicago, Illinois
| | - Gordon P Meares
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia
| | - Shao Xia Lin
- Department of Anesthesiology, University of Illinois, Chicago, Illinois
| | | | - Gesine Saher
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Gottingen, Germany
| | - Lena Spieth
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Gottingen, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Gottingen, Germany
| | - Anne I Boullerne
- Department of Anesthesiology, University of Illinois, Chicago, Illinois
| | - Sarah E Lutz
- Department of Anatomy and Cell Biology, University of Illinois, Chicago, Illinois
| | - Etty N Benveniste
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Douglas L Feinstein
- Department of Anesthesiology, University of Illinois, Chicago, Illinois.,Department of Veterans Affairs, Jesse Brown VA Medical Center, Chicago, Illinois
| |
Collapse
|
22
|
Mundt S, Greter M, Flügel A, Becher B. The CNS Immune Landscape from the Viewpoint of a T Cell. Trends Neurosci 2019; 42:667-679. [DOI: 10.1016/j.tins.2019.07.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 02/07/2023]
|
23
|
Jordão MJC, Sankowski R, Brendecke SM, Sagar, Locatelli G, Tai YH, Tay TL, Schramm E, Armbruster S, Hagemeyer N, Groß O, Mai D, Çiçek Ö, Falk T, Kerschensteiner M, Grün D, Prinz M. Single-cell profiling identifies myeloid cell subsets with distinct fates during neuroinflammation. Science 2019; 363:363/6425/eaat7554. [DOI: 10.1126/science.aat7554] [Citation(s) in RCA: 385] [Impact Index Per Article: 64.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 12/14/2018] [Indexed: 12/15/2022]
Abstract
The innate immune cell compartment is highly diverse in the healthy central nervous system (CNS), including parenchymal and non-parenchymal macrophages. However, this complexity is increased in inflammatory settings by the recruitment of circulating myeloid cells. It is unclear which disease-specific myeloid subsets exist and what their transcriptional profiles and dynamics during CNS pathology are. Combining deep single-cell transcriptome analysis, fate mapping, in vivo imaging, clonal analysis, and transgenic mouse lines, we comprehensively characterized unappreciated myeloid subsets in several CNS compartments during neuroinflammation. During inflammation, CNS macrophage subsets undergo self-renewal, and random proliferation shifts toward clonal expansion. Last, functional studies demonstrated that endogenous CNS tissue macrophages are redundant for antigen presentation. Our results highlight myeloid cell diversity and provide insights into the brain’s innate immune system.
Collapse
|
24
|
Antigen-presenting cell diversity for T cell reactivation in central nervous system autoimmunity. J Mol Med (Berl) 2018; 96:1279-1292. [PMID: 30386908 DOI: 10.1007/s00109-018-1709-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/19/2018] [Accepted: 10/24/2018] [Indexed: 12/13/2022]
Abstract
Autoreactive T cells are considered the major culprits in the pathogenesis of many autoimmune diseases like multiple sclerosis (MS). Upon activation in the lymphoid organs, autoreactive T cells migrate towards the central nervous system (CNS) and target the myelin sheath-forming oligodendrocytes, resulting in detrimental neurological symptoms. Despite the availability of extensively studied systems like the experimental autoimmune encephalomyelitis (EAE) model, our understanding of this disease and the underlying pathogenesis is still elusive. One vividly discussed subject represents the T cell reactivation in the CNS. In order to exert their effector functions in the CNS, autoreactive T cells must encounter antigen-presenting cells (APCs). This interaction provides an antigen-restricted stimulus in the context of major histocompatibility complex class II (MHC-II) and other co-stimulatory molecules. Peripherally derived dendritic cells (DCs), B cells, border-associated macrophages (BAM), CNS-resident microglia, and astrocytes have the capacity to express molecules required for antigen presentation under inflammatory conditions. Also, endothelial cells can fulfill these prerequisites in certain situations. Which of these cells in fact act as APCs for T cell reactivation and to which extent they can exert this function has been studied intensively, but unfortunately with no firm conclusion. In this review, we will summarize the findings that support or question the antigen presenting capacities of the mentioned cell types of CNS-localized T cell reactivation.
Collapse
|
25
|
Giles DA, Duncker PC, Wilkinson NM, Washnock-Schmid JM, Segal BM. CNS-resident classical DCs play a critical role in CNS autoimmune disease. J Clin Invest 2018; 128:5322-5334. [PMID: 30226829 DOI: 10.1172/jci123708] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/11/2018] [Indexed: 12/19/2022] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is an inflammatory demyelinating disease of the central nervous system (CNS), induced by the adoptive transfer of myelin-reactive CD4+ T cells into naive syngeneic mice. It is widely used as a rodent model of multiple sclerosis (MS). The development of EAE lesions is initiated when transferred CD4+ T cells access the CNS and are reactivated by local antigen-presenting cells (APCs) bearing endogenous myelin peptide/MHC class II complexes. The identity of the CNS-resident, lesion-initiating APCs is widely debated. Here we demonstrate that classical dendritic cells (cDCs) normally reside in the meninges, brain, and spinal cord in the steady state. These cells are unique among candidate CNS APCs in their ability to stimulate naive, as well as effector, myelin-specific T cells to proliferate and produce proinflammatory cytokines directly ex vivo. cDCs expanded in the meninges and CNS parenchyma in association with disease progression. Selective depletion of cDCs led to a decrease in the number of myelin-primed donor T cells in the CNS and reduced the incidence of clinical EAE by half. Based on our findings, we propose that cDCs, and the factors that regulate them, be further investigated as potential therapeutic targets in MS.
Collapse
Affiliation(s)
- David A Giles
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology.,Graduate Program in Immunology, and.,Medical Scientist Training Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Patrick C Duncker
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology.,Graduate Program in Immunology, and
| | | | | | - Benjamin M Segal
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology.,Graduate Program in Immunology, and.,Neurology Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| |
Collapse
|
26
|
Boyden AW, Brate AA, Karandikar NJ. Early IFNγ-Mediated and Late Perforin-Mediated Suppression of Pathogenic CD4 T Cell Responses Are Both Required for Inhibition of Demyelinating Disease by CNS-Specific Autoregulatory CD8 T Cells. Front Immunol 2018; 9:2336. [PMID: 30356717 PMCID: PMC6189364 DOI: 10.3389/fimmu.2018.02336] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 09/19/2018] [Indexed: 12/26/2022] Open
Abstract
Pathogenesis of immune-mediated demyelinating diseases like multiple sclerosis (MS) is thought to be governed by a complex cellular interplay between immunopathogenic and immunoregulatory responses. We have previously shown that central nervous system (CNS)-specific CD8 T cells have an unexpected protective role in the mouse model of MS, experimental autoimmune encephalomyelitis (EAE). In this study, we interrogated the suppressive potential of PLP178-191-specific CD8 T cells (PLP-CD8). Here, we show that PLP-CD8, when administered post-disease onset, rapidly ameliorated EAE progression, and suppressed PLP178-191-specific CD4 T cell responses as measured by delayed-type hypersensitivity (DTH). To accomplish DTH suppression, PLP-CD8 required differential production of perforin and IFNγ. Perforin was not required for the rapid suppressive action of these cells, but was critical for maintenance of optimal longer term DTH suppression. Conversely, IFNγ production by PLP-CD8 was necessary for swift DTH suppression, but was less significant for maintenance of longer term suppression. These data indicate that CNS-specific CD8 T cells employ an ordered regulatory mechanism program over a number of days in vivo during demyelinating disease and have mechanistic implications for this immunotherapeutic approach.
Collapse
Affiliation(s)
- Alexander W. Boyden
- Department of Pathology, University of Iowa, Iowa City, IA, United States
- Iowa City Veterans Affairs Medical Center, Iowa City, IA, United States
| | - Ashley A. Brate
- Department of Pathology, University of Iowa, Iowa City, IA, United States
- Iowa City Veterans Affairs Medical Center, Iowa City, IA, United States
- Interdisciplinary Graduate Program in Immunology, University of Iowa Health Care, Iowa City, IA, United States
| | - Nitin J. Karandikar
- Department of Pathology, University of Iowa, Iowa City, IA, United States
- Iowa City Veterans Affairs Medical Center, Iowa City, IA, United States
- Interdisciplinary Graduate Program in Immunology, University of Iowa Health Care, Iowa City, IA, United States
| |
Collapse
|
27
|
Sun L, Telles E, Karl M, Cheng F, Luetteke N, Sotomayor EM, Miller RH, Seto E. Loss of HDAC11 ameliorates clinical symptoms in a multiple sclerosis mouse model. Life Sci Alliance 2018; 1:e201800039. [PMID: 30456376 PMCID: PMC6238389 DOI: 10.26508/lsa.201800039] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 09/16/2018] [Accepted: 09/17/2018] [Indexed: 01/12/2023] Open
Abstract
In an animal model for multiple sclerosis, the absence of HDAC11 reduces clinical severity, spinal cord demyelination, and immune cell infiltration, suggesting that HDAC11 is a promising target for MS treatment. Multiple sclerosis (MS) is a chronic, immune-mediated, demyelinating disease of the central nervous system (CNS). There is no known cure for MS, and currently available drugs for managing this disease are only effective early on and have many adverse side effects. Results from recent studies suggest that histone deacetylase (HDAC) inhibitors may be useful for the treatment of autoimmune and inflammatory diseases such as MS. However, the underlying mechanisms by which HDACs influence immune-mediated diseases such as MS are unclear. More importantly, the question of which specific HDAC(s) are suitable drug targets for the potential treatment of MS remains unanswered. Here, we investigate the functional role of HDAC11 in experimental autoimmune encephalomyelitis, a mouse model for MS. Our results indicate that the loss of HDAC11 in KO mice significantly reduces clinical severity and demyelination of the spinal cord in the post-acute phase of experimental autoimmune encephalomyelitis. The absence of HDAC11 leads to reduced immune cell infiltration into the CNS and decreased monocytes and myeloid DCs in the chronic progressive phase of the disease. Mechanistically, HDAC11 controls the expression of the pro-inflammatory chemokine C–C motif ligand 2 (CCL2) gene by enabling the binding of PU.1 transcription factor to the CCL2 promoter. Our results reveal a novel pathophysiological function for HDAC11 in CNS demyelinating diseases, and warrant further investigations into the potential use of HDAC11-specific inhibitors for the treatment of chronic progressive MS.
Collapse
Affiliation(s)
- Lei Sun
- George Washington University Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, USA.,Department of Biochemistry and Molecular Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Elphine Telles
- George Washington University Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Molly Karl
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Fengdong Cheng
- George Washington University Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Noreen Luetteke
- George Washington University Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Eduardo M Sotomayor
- George Washington University Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Robert H Miller
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Edward Seto
- George Washington University Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, USA.,Department of Biochemistry and Molecular Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
28
|
Tiwari S, Lapierre J, Ojha CR, Martins K, Parira T, Dutta RK, Caobi A, Garbinski L, Ceyhan Y, Esteban-Lopez M, El-Hage N. Signaling pathways and therapeutic perspectives related to environmental factors associated with multiple sclerosis. J Neurosci Res 2018; 96:1831-1846. [PMID: 30204260 PMCID: PMC7167107 DOI: 10.1002/jnr.24322] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/09/2018] [Accepted: 08/13/2018] [Indexed: 12/16/2022]
Abstract
Multiple sclerosis (MS) is an immune-mediated demyelinating disorder of unknown etiology. Both genetic-susceptibility and environment exposures, including vitamin D deficiency, Epstein-Barr viral and Herpesvirus (HHV-6) infections are strongly implicated in the activation of T cells and MS-pathogenesis. Despite precise knowledge of how these factors could be operating alone or in combination to facilitate and aggravate the disease progression, it is clear that prolonged induction of inflammatory molecules and recruitment of other immune cells by the activated T cells results in demyelination and axonal damage. It is imperative to understand the risk factors associated with MS progression and how these factors contribute to disease pathology. Understanding of the underlying mechanisms of what factors triggers activation of T cells to attack myelin antigen are important to strategize therapeutics and therapies against MS. Current review provides a detailed literature to understand the role of both pathogenic and non-pathogenic factors on the impact of MS.
Collapse
Affiliation(s)
- Sneham Tiwari
- Departments of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Jessica Lapierre
- Departments of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Chet Raj Ojha
- Departments of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Kyle Martins
- Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Tiyash Parira
- Departments of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Rajib Kumar Dutta
- Departments of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Allen Caobi
- Departments of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Luis Garbinski
- Cell Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Yasemin Ceyhan
- Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Maria Esteban-Lopez
- Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Nazira El-Hage
- Departments of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| |
Collapse
|
29
|
Kuo PC, Brown DA, Scofield BA, Paraiso HC, Wang PY, Yu IC, Yen JH. Dithiolethione ACDT suppresses neuroinflammation and ameliorates disease severity in experimental autoimmune encephalomyelitis. Brain Behav Immun 2018; 70:76-87. [PMID: 29604346 DOI: 10.1016/j.bbi.2018.03.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/09/2018] [Accepted: 03/11/2018] [Indexed: 01/09/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disorder characterized by the central nervous system (CNS) infiltration of myelin-specific pathogenic T cells followed by brain inflammation in association with demyelination. Similarly, experimental autoimmune encephalomyelitis (EAE), the animal model of MS, also exhibits increased CNS infiltration of pathogenic T cells, including Th1 and Th17, leading to detrimental effects of neuroinflammation and demyelination. We previously reported that 3H-1,2-dithiole-3-thione (D3T), the structurally-simplest of the sulfur-containing dithiolethiones, exerted a promising therapeutic effect in EAE. In the current study we report that 5-Amino-3-thioxo-3H-(1,2)dithiole-4-carboxylic acid ethyl ester (ACDT), a substituted derivative of D3T, exhibits anti-inflammatory properties in EAE. ACDT, administered post immunization, delayed disease onset and reduced disease severity in chronic C57BL/6 EAE, and ACDT, administered during disease remission, suppressed disease relapse in relapsing-remitting SJL/J EAE. Further analysis of the cellular and molecular mechanisms underlying the protective effects of ACDT in EAE revealed that ACDT inhibited pathogenic T cell infiltration, suppressed microglia activation, repressed neurotoxic A1 astrocyte generation, lessened blood-brain barrier disruption, and diminished MMP3/9 production in the CNS of EAE. In summary, we demonstrate that ACDT suppresses neuroinflammation and ameliorates disease severity in EAE through multiple cellular mechanisms. Our findings suggest the potential of developing ACDT as a novel therapeutic agent for the treatment of MS/EAE.
Collapse
Affiliation(s)
- Ping-Chang Kuo
- Department of Microbiology and Immunology, Indiana University School of Medicine, Fort Wayne, IN, United States
| | - Dennis A Brown
- Department of Pharmaceutical Sciences, Manchester University College of Pharmacy, Natural and Health Sciences, Fort Wayne, IN, United States
| | - Barbara A Scofield
- Department of Microbiology and Immunology, Indiana University School of Medicine, Fort Wayne, IN, United States
| | - Hallel C Paraiso
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Fort Wayne, IN, United States
| | - Pei-Yu Wang
- Graduate Institute of Brain and Mind Science, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - I-Chen Yu
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Fort Wayne, IN, United States
| | - Jui-Hung Yen
- Department of Microbiology and Immunology, Indiana University School of Medicine, Fort Wayne, IN, United States.
| |
Collapse
|
30
|
O'Loughlin E, Madore C, Lassmann H, Butovsky O. Microglial Phenotypes and Functions in Multiple Sclerosis. Cold Spring Harb Perspect Med 2018; 8:8/2/a028993. [PMID: 29419406 DOI: 10.1101/cshperspect.a028993] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Microglia are the resident immune cells that constantly survey the central nervous system. They can adapt to their environment and respond to injury or insult by altering their morphology, phenotype, and functions. It has long been debated whether microglial activation is detrimental or beneficial in multiple sclerosis (MS). Recently, the two opposing yet connected roles of microglial activation have been described with the aid of novel microglial markers, RNA profiling, and in vivo models. In this review, microglial phenotypes and functions in the context of MS will be discussed with evidence from both human pathological studies, in vitro and in vivo models. Microglial functional diversity-phagocytosis, antigen presentation, immunomodulation, support, and repair-will also be examined in detail. In addition, this review discusses the emerging evidence for microglia-related targets as biomarkers and therapeutic targets for MS.
Collapse
Affiliation(s)
- Elaine O'Loughlin
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Charlotte Madore
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, A-1090 Vienna, Austria
| | - Oleg Butovsky
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115.,Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
31
|
ATG-dependent phagocytosis in dendritic cells drives myelin-specific CD4 + T cell pathogenicity during CNS inflammation. Proc Natl Acad Sci U S A 2017; 114:E11228-E11237. [PMID: 29233943 DOI: 10.1073/pnas.1713664114] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Although reactivation and accumulation of autoreactive CD4+ T cells within the CNS are considered to play a key role in the pathogenesis of multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE), the mechanisms of how these cells recognize their target organ and induce sustained inflammation are incompletely understood. Here, we report that mice with conditional deletion of the essential autophagy protein ATG5 in classical dendritic cells (DCs), which are present at low frequencies in the nondiseased CNS, are completely resistant to EAE development following adoptive transfer of myelin-specific T cells and show substantially reduced in situ CD4+ T cell accumulation during the effector phase of the disease. Endogenous myelin peptide presentation to CD4+ T cells following phagocytosis of injured, phosphatidylserine-exposing oligodendroglial cells is abrogated in the absence of ATG5. Pharmacological inhibition of ATG-dependent phagocytosis by the cardiac glycoside neriifolin, an inhibitor of the Na+, K+-ATPase, delays the onset and reduces the clinical severity of EAE induced by myelin-specific CD4+ T cells. These findings link phagocytosis of injured oligodendrocytes, a pathological hallmark of MS lesions and during EAE, with myelin antigen processing and T cell pathogenicity, and identify ATG-dependent phagocytosis in DCs as a key regulator in driving autoimmune CD4+ T cell-mediated CNS damage.
Collapse
|
32
|
Wang L, Winnewisser J, Federle C, Jessberger G, Nave KA, Werner HB, Kyewski B, Klein L, Hinterberger M. Epitope-Specific Tolerance Modes Differentially Specify Susceptibility to Proteolipid Protein-Induced Experimental Autoimmune Encephalomyelitis. Front Immunol 2017; 8:1511. [PMID: 29170668 PMCID: PMC5684123 DOI: 10.3389/fimmu.2017.01511] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 10/25/2017] [Indexed: 12/21/2022] Open
Abstract
Immunization with myelin components can elicit experimental autoimmune encephalomyelitis (EAE). EAE susceptibility varies between mouse strains, depending on the antigen employed. BL/6 mice are largely resistant to EAE induction with proteolipid protein (PLP), probably a reflection of antigen-specific tolerance. However, the extent and mechanism(s) of tolerance to PLP remain unclear. Here, we identified three PLP epitopes in PLP-deficient BL/6 mice. PLP-sufficient mice did not respond against two of these, whereas tolerance was “leaky” for an epitope with weak predicted MHCII binding, and only this epitope was encephalitogenic. In TCR transgenic mice, the “EAE-susceptibility-associated” epitope was “ignored” by specific CD4 T cells, whereas the “resistance-associated” epitope induced clonal deletion and Treg induction in the thymus. Central tolerance was autoimmune regulator dependent and required expression and presentation of PLP by thymic epithelial cells (TECs). TEC-specific ablation of PLP revealed that peripheral tolerance, mediated by dendritic cells through recessive tolerance mechanisms (deletion and anergy), could largely compensate for a lack of central tolerance. However, adoptive EAE was exacerbated in mice lacking PLP in TECs, pointing toward a non-redundant role of the thymus in dominant tolerance to PLP. Our findings reveal multiple layers of tolerance to a central nervous system autoantigen that vary among epitopes and thereby specify disease susceptibility. Understanding how different modalities of tolerance apply to distinct T cell epitopes of a target in autoimmunity has implications for antigen-specific strategies to therapeutically interfere with unwanted immune reactions against self.
Collapse
Affiliation(s)
- Lei Wang
- Institute for Immunology, Biomedical Center (BMC) Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Julia Winnewisser
- Institute for Immunology, Biomedical Center (BMC) Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Christine Federle
- Institute for Immunology, Biomedical Center (BMC) Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Gregor Jessberger
- Institute for Immunology, Biomedical Center (BMC) Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Goettingen, Germany
| | - Hauke B Werner
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Goettingen, Germany
| | - Bruno Kyewski
- Division of Developmental Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ludger Klein
- Institute for Immunology, Biomedical Center (BMC) Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Maria Hinterberger
- Institute for Immunology, Biomedical Center (BMC) Munich, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
33
|
Serre L, Girard M, Ramadan A, Menut P, Rouquié N, Lucca LE, Mahiddine K, Leobon B, Mars LT, Guerder S. Thymic-Specific Serine Protease Limits Central Tolerance and Exacerbates Experimental Autoimmune Encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2017; 199:3748-3756. [PMID: 29061767 DOI: 10.4049/jimmunol.1700667] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 09/25/2017] [Indexed: 12/20/2022]
Abstract
The genetic predisposition to multiple sclerosis (MS) is most strongly conveyed by MHC class II haplotypes, possibly by shaping the autoimmune CD4 T cell repertoire. Whether Ag-processing enzymes contribute to MS susceptibility by editing the peptide repertoire presented by these MHC haplotypes is unclear. Thymus-specific serine protease (TSSP) is expressed by thymic epithelial cells and thymic dendritic cells (DCs) and, in these two stromal compartments, TSSP edits the peptide repertoire presented by class II molecules. We show in this article that TSSP increases experimental autoimmune encephalomyelitis severity by limiting central tolerance to myelin oligodendrocyte glycoprotein. The effect on experimental autoimmune encephalomyelitis severity was MHC class II allele dependent, because the lack of TSSP expression conferred protection in NOD mice but not in C57BL/6 mice. Importantly, although human thymic DCs express TSSP, individuals segregate into two groups having a high or 10-fold lower level of expression. Therefore, the level of TSSP expression by thymic DCs may modify the risk factors for MS conferred by some MHC class II haplotypes.
Collapse
Affiliation(s)
- Laurent Serre
- INSERM, U1043, Toulouse F-31300, France.,CNRS, UMR5282, Toulouse F-31300, France.,Centre de Physiopathologie de Toulouse Purpan, Université Toulouse III Paul-Sabatier, Toulouse F-31300, France
| | - Maeva Girard
- INSERM, U1043, Toulouse F-31300, France.,CNRS, UMR5282, Toulouse F-31300, France.,Centre de Physiopathologie de Toulouse Purpan, Université Toulouse III Paul-Sabatier, Toulouse F-31300, France
| | - Abdoulraouf Ramadan
- INSERM, U1043, Toulouse F-31300, France.,CNRS, UMR5282, Toulouse F-31300, France.,Centre de Physiopathologie de Toulouse Purpan, Université Toulouse III Paul-Sabatier, Toulouse F-31300, France
| | - Paul Menut
- INSERM, U1043, Toulouse F-31300, France.,CNRS, UMR5282, Toulouse F-31300, France.,Centre de Physiopathologie de Toulouse Purpan, Université Toulouse III Paul-Sabatier, Toulouse F-31300, France
| | - Nelly Rouquié
- INSERM, U1043, Toulouse F-31300, France.,CNRS, UMR5282, Toulouse F-31300, France.,Centre de Physiopathologie de Toulouse Purpan, Université Toulouse III Paul-Sabatier, Toulouse F-31300, France
| | - Liliana E Lucca
- INSERM, U1043, Toulouse F-31300, France.,CNRS, UMR5282, Toulouse F-31300, France.,Centre de Physiopathologie de Toulouse Purpan, Université Toulouse III Paul-Sabatier, Toulouse F-31300, France
| | - Karim Mahiddine
- INSERM, U1043, Toulouse F-31300, France.,CNRS, UMR5282, Toulouse F-31300, France.,Centre de Physiopathologie de Toulouse Purpan, Université Toulouse III Paul-Sabatier, Toulouse F-31300, France
| | - Bertrand Leobon
- Department of Pediatric Cardiology and Cardiovascular Surgery, Children's Hospital of Toulouse, Toulouse F-31300, France
| | - Lennart T Mars
- INSERM, U1043, Toulouse F-31300, France.,CNRS, UMR5282, Toulouse F-31300, France.,Centre de Physiopathologie de Toulouse Purpan, Université Toulouse III Paul-Sabatier, Toulouse F-31300, France.,INSERM UMR995, Lille Inflammation Research International Center, F-59000 Lille, France; and.,Centre d'Excellence LICEND and FHU IMMINeNT, Université Lille, F-59000 Lille, France
| | - Sylvie Guerder
- INSERM, U1043, Toulouse F-31300, France; .,CNRS, UMR5282, Toulouse F-31300, France.,Centre de Physiopathologie de Toulouse Purpan, Université Toulouse III Paul-Sabatier, Toulouse F-31300, France
| |
Collapse
|
34
|
Chidlow G, Ebneter A, Wood JPM, Casson RJ. Evidence Supporting an Association Between Expression of Major Histocompatibility Complex II by Microglia and Optic Nerve Degeneration During Experimental Glaucoma. J Glaucoma 2017; 25:681-91. [PMID: 27253969 DOI: 10.1097/ijg.0000000000000447] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
AIM We acquired age-matched and sex-matched Sprague-Dawley rats from 2 independent breeding establishments. Serendipitously, we observed that constitutive, and bacterial toxin-induced, expression of major histocompatibility complex (MHC) class II RT1B chain in the uveal tract was much lower in one of the cohorts. Activated microglia are known to upregulate MHC II RT1B expression during optic nerve (ON) degeneration induced by raised intraocular pressure (IOP). We investigated whether, in a model of experimental glaucoma, microglial upregulation of MHC II RT1B was less efficacious and ON degeneration correspondingly less severe in the cohort of rats with low MHC II RT1B expression. METHODS Experimental glaucoma was induced by lasering the trabecular meshwork using a standard protocol. After 2 weeks of elevated IOP, retinal ganglion cells (RGC) survival, ON degeneration, and microglial responses were determined in both cohorts of rats. RESULTS Raised IOP-induced expression of MHC II RT1B by microglia was muted in the "Low" cohort compared with the "High" cohort. Axonal degeneration, RGC loss, and microgliosis were all significantly lower in the cohort of rats with low basal and induced expression of MHC II RT1B, despite both cohorts displaying IOP responses that were indistinguishable in terms of peak IOP and IOP exposure. CONCLUSIONS Expression of MHC II RT1B by activated microglia in the ON during experimental glaucoma was associated with more severe RGC degeneration. Further studies are needed to elucidate the role of MHC II during experimental glaucoma.
Collapse
Affiliation(s)
- Glyn Chidlow
- *Ophthalmic Research Laboratories, South Australian Institute of Ophthalmology, Hanson Institute Centre for Neurological Diseases †Department of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, SA, Australia ‡Department of Ophthalmology, Bern University Hospital and University of Bern, Inselspital, Bern, Switzerland
| | | | | | | |
Collapse
|
35
|
Alissafi T, Banos A, Boon L, Sparwasser T, Ghigo A, Wing K, Vassilopoulos D, Boumpas D, Chavakis T, Cadwell K, Verginis P. Tregs restrain dendritic cell autophagy to ameliorate autoimmunity. J Clin Invest 2017; 127:2789-2804. [PMID: 28581446 DOI: 10.1172/jci92079] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 04/07/2017] [Indexed: 12/26/2022] Open
Abstract
Design of efficacious Treg-based therapies and establishment of clinical tolerance in autoimmune diseases have proven to be challenging. The clinical implementation of Treg immunotherapy has been hampered by various impediments related to the stability and isolation procedures of Tregs as well as the specific in vivo targets of Treg modalities. Herein, we have demonstrated that Foxp3+ Tregs potently suppress autoimmune responses in vivo through inhibition of the autophagic machinery in DCs in a cytotoxic T-lymphocyte-associated protein 4-dependent (CTLA4-dependent) manner. Autophagy-deficient DCs exhibited reduced immunogenic potential and failed to prime autoantigen-specific CD4+ T cells to mediate autoimmunity. Mechanistically, CTLA4 binding promoted activation of the PI3K/Akt/mTOR axis and FoxO1 nuclear exclusion in DCs, leading to decreased transcription of the autophagy component microtubule-associated protein 1 light chain 3β (Lc3b). Human DCs treated with CTLA4-Ig, a fusion protein composed of the Fc region of IgG1 and the extracellular domain of CTLA4 (also known as abatacept, marketed as Orencia), demonstrated reduced levels of autophagosome formation, while DCs from CTLA4-Ig-treated rheumatoid arthritis patients displayed diminished LC3B transcripts. Collectively, our data identify the canonical autophagy pathway in DCs as a molecular target of Foxp3+ Treg-mediated suppression that leads to amelioration of autoimmune responses. These findings may pave the way for the development of therapeutic protocols that exploit Tregs for the treatment of autoimmunity as well as diseases in which disturbed tolerance is a common denominator.
Collapse
Affiliation(s)
- Themis Alissafi
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Aggelos Banos
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | | | - Tim Sparwasser
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Alessandra Ghigo
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Kajsa Wing
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Dimitrios Vassilopoulos
- Joint Rheumatology Program, Clinical Immunology-Rheumatology Unit, National and Kapodistrian University of Athens Medical School, Hippokration General Hospital, Athens, Greece
| | - Dimitrios Boumpas
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,Joint Rheumatology Program, 4th Department of Medicine, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine and Department of Internal Medicine, University of Dresden, Dresden, Germany
| | - Ken Cadwell
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York, New York, USA.,Departments of Microbiology and Medicine, New York University School of Medicine, New York, New York, USA
| | | |
Collapse
|
36
|
CCR8 +FOXp3 + T reg cells as master drivers of immune regulation. Proc Natl Acad Sci U S A 2017; 114:6086-6091. [PMID: 28533380 DOI: 10.1073/pnas.1621280114] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The current study identifies CCR8+ regulatory T cells (Treg cells) as drivers of immunosuppression. We show that in human peripheral blood cells, more than 30% of Treg up-regulate CCR8 following activation in the presence of CCL1. This interaction induces STAT3-dependent up-regulation of FOXp3, CD39, IL-10, and granzyme B, resulting in enhanced suppressive activity of these cells. Of the four human CCR8 ligands, CCL1 is unique in potentiating Treg cells. The relevance of these observations has been extended using an experimental model of multiple sclerosis [experimental autoimmune encephalomyelitis, (EAE)] and a stabilized version of mouse CCL1 (CCL1-Ig). First, we identified a self-feeding mechanism by which CCL1 produced by Treg cells at an autoimmune site up-regulates the expression of its own receptor, CCR8, on these cells. Administration of CCL1-Ig during EAE enhanced the in vivo proliferation of these CCR8+ regulatory cells while inducing the expression of CD39, granzyme B, and IL-10, resulting in the efficacious suppression of ongoing EAE. The critical role of the CCL1-CCR8 axis in Treg cells was further dissected through adoptive transfer studies using CCR8-/- mice. Collectively, we demonstrate the pivotal role of CCR8+ Treg cells in restraining immunity and highlight the potential clinical implications of this discovery.
Collapse
|
37
|
Keller CW, Lünemann JD. Autophagy and Autophagy-Related Proteins in CNS Autoimmunity. Front Immunol 2017; 8:165. [PMID: 28289410 PMCID: PMC5326760 DOI: 10.3389/fimmu.2017.00165] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 02/02/2017] [Indexed: 12/13/2022] Open
Abstract
Autophagy comprises a heterogeneous group of cellular pathways that enables eukaryotic cells to deliver cytoplasmic constituents for lysosomal degradation, to recycle nutrients, and to survive during starvation. In addition to these primordial functions, autophagy has emerged as a key mechanism in orchestrating innate and adaptive immune responses and to shape CD4+ T cell immunity through delivery of peptides to major histocompatibility complex (MHC) class II-containing compartments (MIICs). Individual autophagy proteins additionally modulate expression of MHC class I molecules for CD8+ T cell activation. The emergence and expansion of autoreactive CD4+ and CD8+ T cells are considered to play a key role in the pathogenesis of multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis. Expression of the essential autophagy-related protein 5 (Atg5), which supports T lymphocyte survival and proliferation, is increased in T cells isolated from blood or brain tissues from patients with relapsing-remitting MS. Whether Atgs contribute to the activation of autoreactive T cells through autophagy-mediated antigen presentation is incompletely understood. Here, we discuss the complex functions of autophagy proteins and pathways in regulating T cell immunity and its potential role in the development and progression of MS.
Collapse
Affiliation(s)
- Christian W Keller
- Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zürich , Zürich , Switzerland
| | - Jan D Lünemann
- Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zürich, Zürich, Switzerland; Department of Neurology, University Hospital Zürich, Zürich, Switzerland
| |
Collapse
|
38
|
Brod SA. Ingested (oral) rituximab inhibits EAE. Cytokine 2016; 85:177-83. [DOI: 10.1016/j.cyto.2016.06.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 06/22/2016] [Accepted: 06/23/2016] [Indexed: 12/31/2022]
|
39
|
Increase of Alternatively Activated Antigen Presenting Cells in Active Experimental Autoimmune Encephalomyelitis. J Neuroimmune Pharmacol 2016; 11:721-732. [PMID: 27423192 DOI: 10.1007/s11481-016-9696-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 06/27/2016] [Indexed: 12/31/2022]
Abstract
The importance of CD11c+ antigen-presenting cells (APCs) in the pathogenesis of experimental autoimmune encephalomyelitis (EAE) is well accepted and the gate keeper function of perivascular CD11c+ APCs has been demonstrated. CD11c can be expressed by APCs from external sources or by central nervous system (CNS) resident APCs such as microglia. Yet, changes in the gene expression pattern of CNS CD11c+ APCs during disease are still unclear and differentially expressed genes might play a decisive role in EAE progression. Due to their low numbers in the diseased brain and due to the absence of considerable numbers in the healthy CNS, analysis of CNS CD11c+ cells is technically difficult. To ask whether the CD11c+ APC population contributes to remission of EAE disease, we used Illumina deep mRNA sequencing (RNA-Seq) and quantitative real time polymerase chain reaction (qRT-PCR) analyses to identify the transcriptome of CD11c+ APCs during disease course. We identified a battery of genes that were significantly regulated during the exacerbation of the disease compared to remission and relapse. Three of these genes, Arginase-1, Chi3l3 and Ms4a8a, showed a higher expression at the exacerbation than at later time points during the disease, both in SJL/J and in C57BL/6 mice, and could be attributed to alternatively activated APCs. Expression of Arginase-1, Chi3l3 and Ms4a8a genes was linked to the disease phase of EAE rather than to disease score. Expression of these genes suggested that APCs resembling alternatively activated macrophages are involved during the first wave of neuroinflammation and can be directly associated with the disease progress.
Collapse
|
40
|
Lopes Pinheiro MA, Kamermans A, Garcia-Vallejo JJ, van Het Hof B, Wierts L, O'Toole T, Boeve D, Verstege M, van der Pol SM, van Kooyk Y, de Vries HE, Unger WW. Internalization and presentation of myelin antigens by the brain endothelium guides antigen-specific T cell migration. eLife 2016; 5. [PMID: 27336724 PMCID: PMC4959842 DOI: 10.7554/elife.13149] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 06/22/2016] [Indexed: 11/13/2022] Open
Abstract
Trafficking of myelin-reactive CD4(+) T-cells across the brain endothelium, an essential step in the pathogenesis of multiple sclerosis (MS), is suggested to be an antigen-specific process, yet which cells provide this signal is unknown. Here we provide direct evidence that under inflammatory conditions, brain endothelial cells (BECs) stimulate the migration of myelin-reactive CD4(+) T-cells by acting as non-professional antigen presenting cells through the processing and presentation of myelin-derived antigens in MHC-II. Inflamed BECs internalized myelin, which was routed to endo-lysosomal compartment for processing in a time-dependent manner. Moreover, myelin/MHC-II complexes on inflamed BECs stimulated the trans-endothelial migration of myelin-reactive Th1 and Th17 2D2 cells, while control antigen loaded BECs did not stimulate T-cell migration. Furthermore, blocking the interaction between myelin/MHC-II complexes and myelin-reactive T-cells prevented T-cell transmigration. These results demonstrate that endothelial cells derived from the brain are capable of enhancing antigen-specific T cell recruitment.
Collapse
Affiliation(s)
- Melissa A Lopes Pinheiro
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Alwin Kamermans
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Juan J Garcia-Vallejo
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Bert van Het Hof
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Laura Wierts
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Tom O'Toole
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Daniël Boeve
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Marleen Verstege
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Susanne Ma van der Pol
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Wendy Wj Unger
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
41
|
Piermattei A, Migliara G, Di Sante G, Foti M, Hayrabedyan SB, Papagna A, Geloso MC, Corbi M, Valentini M, Sgambato A, Delogu G, Constantin G, Ria F. Toll-Like Receptor 2 Mediates In Vivo Pro- and Anti-inflammatory Effects of Mycobacterium Tuberculosis and Modulates Autoimmune Encephalomyelitis. Front Immunol 2016; 7:191. [PMID: 27252700 PMCID: PMC4878199 DOI: 10.3389/fimmu.2016.00191] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 05/02/2016] [Indexed: 12/20/2022] Open
Abstract
Mycobacteria display pro- and anti-inflammatory effects in human and experimental pathology. We show here that both effects are mediated by Toll-like receptor 2 (Tlr2), by exploiting a previously characterized Tlr2 variant (Met82Ile). Tlr2 82ile promoted self-specific proinflammatory polarization as well as expansion of ag-specific FoxP3(+) Tregs, while Tlr2 82met impairs the expansion of Tregs and reduces the production of IFN-γ and IL-17 proinflammatory cytokines. Preferential dimerization with Tlr1 or Tlr6 could not explain these differences. In silico, we showed that Tlr2 variant Met82Ile modified the binding pocket for peptidoglycans and participated directly to a putative binding pocket for sugars and cadherins. The distinct pro- and anti-inflammatory actions impacted severity, extent of remission, and distribution of the lesions within the central nervous system of experimental autoimmune encephalomyelitis. Thus, Tlr2 has a janus function in vivo as mediator of the role of bacterial products in balancing pro- and anti-inflammatory immune responses.
Collapse
Affiliation(s)
- Alessia Piermattei
- Laboratory of Immunology, Institute of General Pathology, Università Cattolica del Sacro Cuore , Rome , Italy
| | - Giuseppe Migliara
- Laboratory of Immunology, Institute of General Pathology, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Public Health and Infectious Diseases, University "La Sapienza", Rome, Italy
| | - Gabriele Di Sante
- Laboratory of Immunology, Institute of General Pathology, Università Cattolica del Sacro Cuore, Rome, Italy; Institute of Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maria Foti
- Molecular Medicine and Immunology Laboratory, Genopolis Consortium, University of "Milano Bicocca" , Milan , Italy
| | - Soren Bohos Hayrabedyan
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences , Sofia , Bulgaria
| | - Angela Papagna
- Molecular Medicine and Immunology Laboratory, Genopolis Consortium, University of "Milano Bicocca" , Milan , Italy
| | - Maria Concetta Geloso
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore , Rome , Italy
| | - Maddalena Corbi
- Institute of General Pathology, Università Cattolica del Sacro Cuore , Rome , Italy
| | - Mariagrazia Valentini
- Laboratory of Immunology, Institute of General Pathology, Università Cattolica del Sacro Cuore , Rome , Italy
| | - Alessandro Sgambato
- Institute of General Pathology, Università Cattolica del Sacro Cuore , Rome , Italy
| | - Giovanni Delogu
- Institute of Microbiology, Università Cattolica del Sacro Cuore , Rome , Italy
| | - Gabriela Constantin
- Section of General Pathology, Department of Medicine, University of Verona , Verona , Italy
| | - Francesco Ria
- Laboratory of Immunology, Institute of General Pathology, Università Cattolica del Sacro Cuore , Rome , Italy
| |
Collapse
|
42
|
Abstract
Experimental autoimmune encephalitis (EAE), the animal model of multiple sclerosis (MS), has provided significant insight into the mechanisms that initiate and drive autoimmunity. Several central nervous system proteins and peptides have been used to induce disease, in a number of different mouse strains, to model the diverse clinical presentations of MS. In this chapter, we detail the materials and methods used to induce active and adoptive EAE. We focus on disease induction in the SJL/J, C57BL/6, and BALB/c mouse strains, using peptides derived from proteolipid protein, myelin basic protein, and myelin oligodendrocyte glycoprotein. We also include a protocol for the isolation of leukocytes from the spinal cord and brain for flow cytometric analysis.
Collapse
Affiliation(s)
- Rachael L Terry
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | | | | |
Collapse
|
43
|
Prinz J, Karacivi A, Stormanns ER, Recks MS, Kuerten S. Time-Dependent Progression of Demyelination and Axonal Pathology in MP4-Induced Experimental Autoimmune Encephalomyelitis. PLoS One 2015; 10:e0144847. [PMID: 26658811 PMCID: PMC4676607 DOI: 10.1371/journal.pone.0144847] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 11/24/2015] [Indexed: 11/21/2022] Open
Abstract
Background Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS) characterized by inflammation, demyelination and axonal pathology. Myelin basic protein/proteolipid protein (MBP-PLP) fusion protein MP4 is capable of inducing chronic experimental autoimmune encephalomyelitis (EAE) in susceptible mouse strains mirroring diverse histopathological and immunological hallmarks of MS. Limited availability of human tissue underscores the importance of animal models to study the pathology of MS. Methods Twenty-two female C57BL/6 (B6) mice were immunized with MP4 and the clinical development of experimental autoimmune encephalomyelitis (EAE) was observed. Methylene blue-stained semi-thin and ultra-thin sections of the lumbar spinal cord were assessed at the peak of acute EAE, three months (chronic EAE) and six months after onset of EAE (long-term EAE). The extent of lesional area and inflammation were analyzed in semi-thin sections on a light microscopic level. The magnitude of demyelination and axonal damage were determined using electron microscopy. Emphasis was put on the ventrolateral tract (VLT) of the spinal cord. Results B6 mice demonstrated increasing demyelination and severe axonal pathology in the course of MP4-induced EAE. In addition, mitochondrial swelling and a decrease in the nearest neighbor neurofilament distance (NNND) as early signs of axonal damage were evident with the onset of EAE. In semi-thin sections we observed the maximum of lesional area in the chronic state of EAE while inflammation was found to a similar extent in acute and chronic EAE. In contrast to the well-established myelin oligodendrocyte glycoprotein (MOG) model, disease stages of MP4-induced EAE could not be distinguished by assessing the extent of parenchymal edema or the grade of inflammation. Conclusions Our results complement our previous ultrastructural studies of B6 EAE models and suggest that B6 mice immunized with different antigens constitute useful instruments to study the diverse histopathological aspects of MS.
Collapse
MESH Headings
- Animals
- Axons/pathology
- Axons/ultrastructure
- Demyelinating Diseases
- Disease Models, Animal
- Disease Progression
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Female
- Humans
- Immunization
- Lumbar Vertebrae/pathology
- Lumbar Vertebrae/ultrastructure
- Mice
- Mice, Inbred C57BL
- Microtomy
- Mitochondria/pathology
- Mitochondria/ultrastructure
- Mitochondrial Swelling
- Multiple Sclerosis/immunology
- Multiple Sclerosis/pathology
- Multiple Sclerosis/physiopathology
- Myelin Basic Protein/administration & dosage
- Myelin Proteolipid Protein/administration & dosage
- Myelin Sheath/pathology
- Myelin Sheath/ultrastructure
- Recombinant Fusion Proteins/administration & dosage
- Severity of Illness Index
- Time Factors
Collapse
Affiliation(s)
- Johanna Prinz
- Department of Anatomy I, University of Cologne, Joseph-Stelzmann-Str. 9, 50931, Cologne, Germany
| | - Aylin Karacivi
- Department of Anatomy I, University of Cologne, Joseph-Stelzmann-Str. 9, 50931, Cologne, Germany
| | - Eva R. Stormanns
- Department of Anatomy I, University of Cologne, Joseph-Stelzmann-Str. 9, 50931, Cologne, Germany
| | - Mascha S. Recks
- Department of Anatomy II, University of Cologne, Joseph-Stelzmann-Str. 9, 50931, Cologne, Germany
| | - Stefanie Kuerten
- Department of Anatomy and Cell Biology, University of Würzburg, Koellikerstraße 6, 97070, Würzburg, Germany
- * E-mail:
| |
Collapse
|
44
|
Brod SA. Ingested (oral) anti-IL-12/23 inhibits EAE. J Neurol Sci 2015; 361:19-25. [PMID: 26810510 DOI: 10.1016/j.jns.2015.12.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 11/17/2015] [Accepted: 12/07/2015] [Indexed: 02/05/2023]
Abstract
BACKGROUND Blocking the activity of IL-12/23 can inhibit autoimmune diseases such as psoriasis. OBJECTIVE We examined whether an antibody against IL-12/23, ustekinumab (UTZ) (Stelera®), used clinically in psoriasis would have similar anti-inflammatory effects in EAE after oral administration. DESIGN/METHODS B6 mice were immunized with MOG peptide 35-55 and gavaged with isotype IgG control or UTZ during ongoing disease. Splenocytes, CD4(+) T cells or macrophages/monocyte lineage cells (CD11b(+)) from control fed or UTZ fed mice were adoptively transferred into active MOG peptide 35-55 immunized recipient mice during ongoing disease. Actively fed and recipient mice were examined for disease inhibition, inflammation, and cytokine responses. RESULTS Ingested (oral) UTZ inhibited ongoing disease and decreased inflammation. Adoptively transferred cells from UTZ fed donors protected against actively induced disease and decreased inflammation. Oral UTZ decreased pro-inflammatory cytokines Th1-like cytokines IL-2, IL-12, IFN-γ, IL-17 (Teff) and TNF-α in UTZ fed mice and increased counter-regulatory cytokines IL-4, IL-10 and IL-13 in recipients of donor cells from UTZ fed mice. CONCLUSIONS Ingested (orally administered) UTZ can inhibit disease, CNS inflammation, decrease pro-inflammatory Th1-like and Th17 cytokines and increase Th2-like anti-inflammatory cytokines.
Collapse
Affiliation(s)
- Staley A Brod
- Department of Neurology, Division of MS/Neuroimmunology, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226, USA.
| |
Collapse
|
45
|
Do not judge a cell by its cover--diversity of CNS resident, adjoining and infiltrating myeloid cells in inflammation. Semin Immunopathol 2015; 37:591-605. [PMID: 26251238 DOI: 10.1007/s00281-015-0520-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 07/27/2015] [Indexed: 12/24/2022]
Abstract
Specialized populations of tissue-resident myeloid cells inhabit every organ of the body. While many of these populations appear similar morphologically and phenotypically, they exhibit great functional diversity. The central nervous system (CNS), as an immune privileged organ, possesses a unique tissue-resident macrophage population, the microglia, as well as numerous myeloid cell subsets at its boarders and barriers in CNS-adjoining tissues, namely the meninges, the perivascular space, and the choroid plexus. Recent research has added much to our knowledge about microglia, whereas the populations of CNS-surrounding phagocytes are just starting to be appreciated. As guardians of CNS homeostasis, these myeloid cells perform immune surveillance and immune modulatory tasks in health and disease. As such, microglia and CNS-surrounding antigen-presenting cells have been shown to be crucially involved not only in the initiation and progression but also resolution of multiple sclerosis (MS). MS and its rodent model, experimental autoimmune encephalomyelitis, are autoimmune inflammatory demyelinating CNS pathologies. While some crucial aspects of the disease pathogenesis have been solved, much of the complex involvement and interplay of the innate immune compartment remains yet to be clarified. Here, we will discuss the current understanding of the scope of phenotypes and functions of myeloid cells involved in CNS neuroinflammation.
Collapse
|
46
|
Procaccini C, De Rosa V, Pucino V, Formisano L, Matarese G. Animal models of Multiple Sclerosis. Eur J Pharmacol 2015; 759:182-91. [PMID: 25823807 PMCID: PMC7094661 DOI: 10.1016/j.ejphar.2015.03.042] [Citation(s) in RCA: 222] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 01/30/2015] [Accepted: 03/12/2015] [Indexed: 12/26/2022]
Abstract
Multiple Sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) which involves a complex interaction between immune system and neural cells. Animal modeling has been critical for addressing MS pathogenesis. The three most characterized animal models of MS are (1) the experimental autoimmune/allergic encephalomyelitis (EAE); (2) the virally-induced chronic demyelinating disease, known as Theiler׳s murine encephalomyelitis virus (TMEV) infection and (3) the toxin-induced demyelination. All these models, in a complementary way, have allowed to reach a good knowledge of the pathogenesis of MS. Specifically, EAE is the model which better reflects the autoimmune pathogenesis of MS and is extremely useful to study potential experimental treatments. Furthermore, both TMEV and toxin-induced demyelination models are suitable for characterizing the role of the axonal injury/repair and the remyelination process in MS. In conclusion, animal models, despite their limitations, remain the most useful instrument for implementing the study of MS.
Collapse
MESH Headings
- Animals
- Cardiovirus Infections/pathology
- Cardiovirus Infections/virology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/etiology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Humans
- Mice
- Mice, Transgenic
- Multiple Sclerosis/etiology
- Multiple Sclerosis/genetics
- Multiple Sclerosis/immunology
- Multiple Sclerosis/pathology
- Theilovirus/pathogenicity
Collapse
Affiliation(s)
- Claudio Procaccini
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR) c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131 Napoli, Italy
| | - Veronica De Rosa
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR) c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131 Napoli, Italy; Unità di NeuroImmunologia, IRCCS Fondazione Santa Lucia, 00143 Roma, Italy
| | - Valentina Pucino
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli Federico II, 80131 Napoli, Italy
| | - Luigi Formisano
- Divisione di Farmacologia, Dipartimento di Scienze e Tecnologie, Università degli Studi del Sannio, 82100 Benevento, Italy
| | - Giuseppe Matarese
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, Baronissi Campus, 84081 Baronissi, Salerno, Italy; IRCCS Multimedica, 20138 Milano, Italy.
| |
Collapse
|
47
|
Huang X, Wu H, Lu Q. The mechanisms and applications of T cell vaccination for autoimmune diseases: a comprehensive review. Clin Rev Allergy Immunol 2015; 47:219-33. [PMID: 25096807 DOI: 10.1007/s12016-014-8439-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Autoimmune diseases (ADs) are a spectrum of diseases originating from loss of immunologic self-tolerance and T cell abnormal autoreactivity, causing organ damage and death. However, the pathogenic mechanism of ADs remains unclear. The current treatments of ADs include nonsteroidal anti-inflammatory drugs (NSAIDS), antimalarials, corticosteroids, immunosuppressive drugs, and biological therapies. With the need to prevent side effects resulting from current treatments and acquire better clinical remission, developing a novel pharmaceutical treatment is extremely urgent. The concept of T cell vaccination (TCV) has been raised as the finding that immunization with attenuated autoreactive T cells is capable of inducing T cell-dependent inhibition of autoimmune responses. TCV may act as an approach to control unwanted adaptive immune response through eliminating the autoreactive T cells. Over the past decades, the effect of TCV has been justified in several animal models of autoimmune diseases including experimental autoimmune encephalomyelitis (EAE), murine autoimmune diabetes in nonobese diabetic (NOD) mice, collagen-induced arthritis (CIA), and so on. Meanwhile, clinical trials of TCV have confirmed the safety and efficacy in corresponding autoimmune diseases ranging from multiple sclerosis (MS) to systemic lupus erythematosus (SLE). This review aims to summarize the ongoing experimental and clinical trials and elucidate possible molecule mechanisms of TCV.
Collapse
Affiliation(s)
- Xin Huang
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, Second Xiangya Hospital, Central South University, #139 Renmin Middle Rd, Changsha, 410011, Hunan, People's Republic of China
| | | | | |
Collapse
|
48
|
Recks MS, Grether NB, van der Broeck F, Ganscher A, Wagner N, Henke E, Ergün S, Schroeter M, Kuerten S. Four different synthetic peptides of proteolipid protein induce a distinct antibody response in MP4-induced experimental autoimmune encephalomyelitis. Clin Immunol 2015; 159:93-106. [PMID: 25959684 DOI: 10.1016/j.clim.2015.04.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 04/30/2015] [Indexed: 01/26/2023]
Abstract
Here we studied the autoantibody specificity elicited by proteolipid protein (PLP) in MP4-induced experimental autoimmune encephalomyelitis, a mouse model of multiple sclerosis (MS). In C57BL/6 (B6) mice, antibodies were induced by immunization with one of the two extracellular and by the intracellular PLP domain. Antibodies against extracellular PLP were myelin-reactive in oligodendrocyte cultures and induced mild spinal cord demyelination upon transfer into B cell-deficient J(H)T mice. Remarkably, also antibodies against intracellular PLP showed binding to intact oligodendrocytes and were capable of inducing myelin pathology upon transfer into J(H)T mice. In MP4-immunized mice peptide-specific T(H)1/T(H)17 responses were mainly directed against the extracellular PLP domains, but also involved the intracellular epitopes. These data suggest that both extracellular and intracellular epitopes of PLP contribute to the pathogenesis of MP4-induced EAE already in the setting of intact myelin. It remains to be elucidated if this concept also applies to MS itself.
Collapse
Affiliation(s)
- Mascha S Recks
- Department of Anatomy II (Neuroanatomy), University of Cologne, Kerpener Straβe 62, 50924 Cologne, Germany
| | - Nicolai B Grether
- Department of Anatomy and Cell Biology, University of Wuerzburg, Koellikerstr. 6, 97070 Wuerzburg, Germany
| | | | - Alla Ganscher
- Department of Anatomy and Cell Biology, University of Wuerzburg, Koellikerstr. 6, 97070 Wuerzburg, Germany
| | - Nicole Wagner
- Department of Anatomy and Cell Biology, University of Wuerzburg, Koellikerstr. 6, 97070 Wuerzburg, Germany
| | - Erik Henke
- Department of Anatomy and Cell Biology, University of Wuerzburg, Koellikerstr. 6, 97070 Wuerzburg, Germany
| | - Süleyman Ergün
- Department of Anatomy and Cell Biology, University of Wuerzburg, Koellikerstr. 6, 97070 Wuerzburg, Germany
| | - Michael Schroeter
- Department of Neurology, University of Cologne, Kerpener Str. 62, 50924 Cologne, Germany
| | - Stefanie Kuerten
- Department of Anatomy and Cell Biology, University of Wuerzburg, Koellikerstr. 6, 97070 Wuerzburg, Germany.
| |
Collapse
|
49
|
Parker Harp CR, Archambault AS, Sim J, Ferris ST, Mikesell RJ, Koni PA, Shimoda M, Linington C, Russell JH, Wu GF. B cell antigen presentation is sufficient to drive neuroinflammation in an animal model of multiple sclerosis. THE JOURNAL OF IMMUNOLOGY 2015; 194:5077-84. [PMID: 25895531 DOI: 10.4049/jimmunol.1402236] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 03/20/2015] [Indexed: 11/19/2022]
Abstract
B cells are increasingly regarded as integral to the pathogenesis of multiple sclerosis, in part as a result of the success of B cell-depletion therapy. Multiple B cell-dependent mechanisms contributing to inflammatory demyelination of the CNS have been explored using experimental autoimmune encephalomyelitis (EAE), a CD4 T cell-dependent animal model for multiple sclerosis. Although B cell Ag presentation was suggested to regulate CNS inflammation during EAE, direct evidence that B cells can independently support Ag-specific autoimmune responses by CD4 T cells in EAE is lacking. Using a newly developed murine model of in vivo conditional expression of MHC class II, we reported previously that encephalitogenic CD4 T cells are incapable of inducing EAE when B cells are the sole APC. In this study, we find that B cells cooperate with dendritic cells to enhance EAE severity resulting from myelin oligodendrocyte glycoprotein (MOG) immunization. Further, increasing the precursor frequency of MOG-specific B cells, but not the addition of soluble MOG-specific Ab, is sufficient to drive EAE in mice expressing MHCII by B cells alone. These data support a model in which expansion of Ag-specific B cells during CNS autoimmunity amplifies cognate interactions between B and CD4 T cells and have the capacity to independently drive neuroinflammation at later stages of disease.
Collapse
Affiliation(s)
- Chelsea R Parker Harp
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110
| | - Angela S Archambault
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110
| | - Julia Sim
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110
| | - Stephen T Ferris
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Robert J Mikesell
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110
| | - Pandelakis A Koni
- Cancer Immunology, Inflammation, and Tolerance Program, Cancer Center and Department of Medicine, Georgia Regents University, Augusta, GA 30912
| | - Michiko Shimoda
- Department of Dermatology, University of California at Davis School of Medicine, Sacramento, CA 95817; and
| | | | - John H Russell
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110
| | - Gregory F Wu
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110;
| |
Collapse
|
50
|
Pharmaceutical integrated stress response enhancement protects oligodendrocytes and provides a potential multiple sclerosis therapeutic. Nat Commun 2015; 6:6532. [PMID: 25766071 PMCID: PMC4360920 DOI: 10.1038/ncomms7532] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 02/05/2015] [Indexed: 01/21/2023] Open
Abstract
Oligodendrocyte death contributes to the pathogenesis of the inflammatory demyelinating disease multiple sclerosis (MS). Nevertheless, current MS therapies are mainly immunomodulatory and have demonstrated limited ability to inhibit MS progression. Protection of oligodendrocytes is therefore a desirable strategy for alleviating disease. Here we demonstrate that enhancement of the integrated stress response using the FDA-approved drug guanabenz increases oligodendrocyte survival in culture and prevents hypomyelination in cerebellar explants in the presence of interferon-γ, a pro-inflammatory cytokine implicated in MS pathogenesis. In vivo, guanabenz treatment protects against oligodendrocyte loss caused by CNS-specific expression of interferon-γ. In a mouse model of MS, experimental autoimmune encephalomyelitis, guanabenz alleviates clinical symptoms, which correlates with increased oligodendrocyte survival and diminished CNS CD4+ T cell accumulation. Moreover, guanabenz ameliorates relapse in relapsing-remitting experimental autoimmune encephalomyelitis. Our results provide support for a MS therapy that enhances the integrated stress response to protect oligodendrocytes against the inflammatory CNS environment. Current multiple sclerosis treatments focus on prevention of immune attack on oligodendrocytes and myelin. Here the authors show a different strategy to ameliorate disease in several mouse models, protecting oligodendrocytes from inflammation-induced death with an FDA-approved drug, guanabenz.
Collapse
|