1
|
Phan NM, Nguyen TL, Min DK, Kim J. Mesoporous polydopamine nanoparticle-based tolerogenic vaccine induces antigen-specific immune tolerance to prevent and treat autoimmune multiple sclerosis. Biomaterials 2025; 316:122997. [PMID: 39662275 DOI: 10.1016/j.biomaterials.2024.122997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 10/24/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
Multiple sclerosis (MS) is a chronic neurological disorder derived from autoreactive immune system attacking the protective myelin sheath that surrounds nerves in the central nervous system (CNS). Here, a tolerogenic nanovaccine for generating an antigen-specific immune tolerance for treating MS is proposed. It consisted of a mesoporous polydopamine (mPDA) nanoparticle, characterized by high reactive oxygen species (ROS)-scavenging property, loaded with MS-derived autoantigen. Intravenous vaccination of autoantigen-loaded mPDA could induce tolerogenic dendritic cells (DCs) with low expression of co-stimulatory molecules while presenting peptide epitopes. The tolerogenic DCs induced peripheral regulatory T-cells (Tregs), thereby reducing infiltration of autoreactive CD4+ T-cells and inflammatory antigen-presenting cells (APCs) into the CNS. In MS-mimicking mouse model, the tolerogenic nanovaccine prevented MS development in the early therapeutic setup and exhibited an enhanced recovery from complete paralysis in the late therapeutic model. The current platform could be exploited to treat other autoimmune diseases where disease-dependent autoantigen peptides are delivered.
Collapse
Affiliation(s)
- Ngoc Man Phan
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Thanh Loc Nguyen
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Dong Kwang Min
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Jaeyun Kim
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea; Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea; Department of MetaBioHealth, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea; Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea; Institute of Quantum Biophysics (IQB), Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
2
|
Rudebeck S, Eyre M, Lim M. Neuropsychological outcomes in pediatric MOGAD: clinical practice and future research. Child Neuropsychol 2025:1-15. [PMID: 40240317 DOI: 10.1080/09297049.2025.2489697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 04/01/2025] [Indexed: 04/18/2025]
Abstract
Myelin Oligodendrocyte Glycoprotein Antibody-Associated Disorder (MOGAD) is a recently identified demyelinating condition affecting children and adults. Its impact on children's cognitive outcomes remains poorly understood but is a growing area of interest due to potential long-term implications. A systematic PubMed search was conducted to identify English-language studies that assessed cognition in individuals under 18 with MOGAD using neuropsychological tests, screening tools, or questionnaires. Children with MOGAD, particularly those with phenotypes such as AcuteDisseminated Encephalomyelitis (ADEM) and Neuromyelitis Optica SpectrumDisorder (NMOSD), often exhibit impairments in intellectual functioning, memory, processing speed, and working memory. However, some children maintain cognitive performance within the normal range. Cognitive difficulties are linked to disease relapses and may develop over time, although brain lesions do not consistently correlate with cognitive outcomes. Current studies, limited by small sample sizes, indicate that children with MOGAD are at risk for cognitive impairments. Regular neuropsychological monitoring is essential for pediatric MOGADpatients to identify and address cognitive challenges early, mitigating risks of academic and occupational underachievement. Multicentre multinational studies are needed to understand the cognitive profile of MOGAD better and assess the influence of disease-related variables on cognitive outcomes.
Collapse
Affiliation(s)
- Sarah Rudebeck
- Department of Neuropsychology, Kings College Hospital, London, UK
| | - Michael Eyre
- Children's Neurosciences, Evelina London Children's Hospital at Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Ming Lim
- Children's Neurosciences, Evelina London Children's Hospital at Guy's and St Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|
3
|
Hughes EP, Syage AR, Mehrabad EM, Lane TE, Spike BT, Tantin D. OCA-B promotes pathogenic maturation of stem-like CD4 + T cells and autoimmune demyelination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.11.29.569210. [PMID: 38076925 PMCID: PMC10705450 DOI: 10.1101/2023.11.29.569210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Stem-like T cells selectively contribute to autoimmunity, but the activities that promote their pathogenicity are incompletely understood. Here, we identify the transcription coregulator OCA-B as a driver of the pathogenic maturation of stem-like CD4 + T cell to promote autoimmune demyelination. Using two human multiple sclerosis (MS) datasets, we show that POU2AF1 , the gene encoding OCA-B, is elevated in CD4 + T cells from MS patients. We show that T cell-intrinsic OCA-B loss protects mice from experimental autoimmune encephalomyelitis (EAE) while preserving responses to viral CNS infection. In EAE models driven by antigen reencounter, OCA-B deletion nearly eliminates CNS infiltration, proinflammatory cytokine production and clinical disease. OCA-B-expressing CD4 + T cells of mice primed with autoantigen express an encephalitogenic gene program and preferentially confer disease. In a relapsing-remitting EAE model, OCA-B loss protects mice specifically at relapse. During remission, OCA-B promotes the expression of Tcf7 , Slamf6 , and Sell in proliferating CNS T cell populations. At relapse timepoints, OCA-B loss results in both the accumulation of an immunomodulatory CD4 + T cell population expressing Ccr9 and Bach2 , and loss of pro-inflammatory gene expression from Th17 cells. These results identify OCA-B as a driver of pathogenic CD4 + T cells.
Collapse
|
4
|
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
|
5
|
Üremiş MM, Üremiş N, Gül M, Gül S, Çiğremiş Y, Durhan M, Türköz Y. Acrylamide, Applied During Pregnancy and Postpartum Period in Offspring Rats, Significantly Disrupted Myelination by Decreasing the Levels of Myelin-Related Proteins: MBP, MAG, and MOG. Neurochem Res 2024; 49:617-635. [PMID: 37989894 DOI: 10.1007/s11064-023-04053-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 08/06/2023] [Accepted: 10/19/2023] [Indexed: 11/23/2023]
Abstract
Acrylamide (ACR) is a colorless, odorless, and water-soluble solid molecule. In addition to being an important industrial material, ACR is found in fried and baked carbohydrate-rich foods. ACR is regarded as a typical axonal neurotoxin that induces neuropathy. The brain is protected from oxidative damage by vitamin E, which is regarded as the most powerful fat-soluble antioxidant vitamin. This study aimed to reveal the toxic effect of ACR on the development of myelin in the brain at the molecular level and to examine whether Vitamin E has a neuroprotective effect on the harmful effect of ACR. The study was started by dividing 40 pregnant rats into 4 groups and after lactation, the study was continued with offspring rats (females and males offspring rats) from each group. Offspring rats were equally divided into Control, Vitamin E, ACR, ACR + Vitamin E groups. Following the ACR administration, the Water Maze test was applied to evaluate cognitive function. To evaluate the level of demyelination and remyelination, MBP, MAG, and MOG proteins and mRNA levels were performed. In addition, the degeneration of myelin and glial cells was examined by immunohistochemistry and electron microscopic analysis. Analysis results showed that ACR administration decreased gene and protein levels of myelin-related proteins MBP, MAG, and MOG. The findings were confirmed by histopathological, immunohistochemical, and microscopic examinations. The application of vitamin E improved this negative effect of ACR. It has been observed that ACR may play a role in the pathogenesis of myelin-related neurodegenerative diseases by causing demyelination during gestation, lactation, and post-lactation. In addition, it has been understood that vitamin E supports myelination as a strong neuroprotective vitamin against the toxicity caused by ACR. Our research results suggest that acrylamide may play a role in the etiopathogenesis of demyelinating diseases such as multiple sclerosis in humans since fast-food-type nutrition is very common today and people are chronically exposed to acrylamide.
Collapse
Affiliation(s)
- Muhammed Mehdi Üremiş
- Department of Medical Biochemistry, Medical Faculty, Inonu University, Malatya, Turkey
| | - Nuray Üremiş
- Department of Medical Biochemistry, Medical Faculty, Inonu University, Malatya, Turkey
| | - Mehmet Gül
- Department of Histology and Embryology, Medical Faculty, Inonu University, Malatya, Turkey
| | - Semir Gül
- Department of Histology and Embryology, Medical Faculty, Inonu University, Malatya, Turkey
| | - Yılmaz Çiğremiş
- Department of Medical Biology and Genetics, Medical Faculty, Inonu University, Malatya, Turkey
| | - Merve Durhan
- Department of Medical Biology and Genetics, Medical Faculty, Inonu University, Malatya, Turkey
| | - Yusuf Türköz
- Department of Medical Biochemistry, Medical Faculty, Inonu University, Malatya, Turkey.
| |
Collapse
|
6
|
Amin M, Al-Iedani O, Lea RA, Brilot F, Maltby VE, Lechner-Scott J. A longitudinal analysis of brain volume changes in myelin oligodendrocyte glycoprotein antibody-associated disease. J Neuroimaging 2024; 34:78-85. [PMID: 38018386 DOI: 10.1111/jon.13175] [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: 08/15/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND AND PURPOSE Myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD) is a relapsing demyelinating condition. There are several cross-sectional studies showing evidence of brain atrophy in people with MOGAD (pwMOGAD), but longitudinal brain volumetric assessment is still an unmet need. Current recommendations do not include monitoring with MRI and assume distinct attacks. Evidence of ongoing axon loss will have diagnostic and therapeutic implications. In this study, we assessed brain volume changes in pwMOGAD over a mean follow-up period of 2 years and compared this to changes in people with multiple sclerosis (pwMS). METHODS This is a retrospective single-center study over a 7-year period from 2014 to 2021. MRI brain scans at the time of diagnosis and follow-up in remission were collected from 14 Caucasian pwMOGAD, confirmed by serum myelin oligodendrocyte glycoprotein immunoglobulin G antibody presence, detected by live cell-based assays. Total brain volume (TBV), white matter (WM), gray matter (GM), and demyelinating lesion volumes were assessed automatically using the Statistical Parametric Mapping and FMRIB automated segmentation tools. MRI brain scans at diagnosis and follow-up on remission were collected from 32-matched pwMS for comparison. Statistical analysis was done using analysis of variance. RESULTS There is evidence of TBV loss, affecting particularly GM, over an approximately 2-year follow-up period in pwMOGAD (p < .05), comparable to pwMS. WM and lesion volume change over the same period were not statistically significant (p > .1). CONCLUSION We found evidence of loss of GM and TBV over time in pwMOGAD, similar to pwMS, although the WM and lesion volumes were unchanged.
Collapse
Affiliation(s)
- Mohammad Amin
- Nepean Hospital, Kingswood, New South Wales, Australia
- Department of Neurology, John Hunter Hospital, New Lambton Heights, New South Wales, Australia
| | - Oun Al-Iedani
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Immune Health Program, Hunter Medical Research Institute, New Lambton, New South Wales, Australia
| | - Rodney A Lea
- Immune Health Program, Hunter Medical Research Institute, New Lambton, New South Wales, Australia
- Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Fabienne Brilot
- Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
- Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Vicki E Maltby
- Department of Neurology, John Hunter Hospital, New Lambton Heights, New South Wales, Australia
- Immune Health Program, Hunter Medical Research Institute, New Lambton, New South Wales, Australia
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
| | - Jeannette Lechner-Scott
- Department of Neurology, John Hunter Hospital, New Lambton Heights, New South Wales, Australia
- Immune Health Program, Hunter Medical Research Institute, New Lambton, New South Wales, Australia
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
| |
Collapse
|
7
|
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
|
8
|
Phan NM, Nguyen TL, Shin H, Trinh TA, Kim J. ROS-Scavenging Lignin-Based Tolerogenic Nanoparticle Vaccine for Treatment of Multiple Sclerosis. ACS NANO 2023; 17:24696-24709. [PMID: 38051295 DOI: 10.1021/acsnano.3c04497] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Multiple sclerosis (MS) is a demyelinating autoimmune disease, in which the immune system attacks myelin. Although systemic immunosuppressive agents have been used to treat MS, long-term treatment with these drugs causes undesirable side effects such as altered glucose metabolism, insomnia, and hypertension. Herein, we propose a tolerogenic therapeutic vaccine to treat MS based on lignin nanoparticles (LNP) with intrinsic reactive oxygen species (ROS)-scavenging capacity derived from their phenolic moieties. The LNP loaded with autoantigens of MS allowed for inducing tolerogenic DCs with low-level expression of costimulatory molecules while presenting antigenic peptides. Intravenous injection of an LNP-based tolerogenic vaccine into an experimental autoimmune encephalomyelitis (EAE) model led to durable antigen-specific immune tolerance via inducing regulatory T cells (Tregs). Autoreactive T helper type 1 cells, T helper type 17 cells, and inflammatory antigen presentation cells (APCs) were suppressed in the central nervous system (CNS), ameliorating ongoing MS in early and late disease states. Additionally, the incorporation of dexamethasone into an LNP-based tolerogenic nanovaccine could further improve the recovery of EAE mice in the severe chronic stage. As lignin is the most abundant biomass and waste byproduct in the pulping industry, a lignin-based tolerogenic vaccine could be a novel, cost-effective, high-value vaccine platform with potent therapeutic efficiency in treating autoimmune diseases.
Collapse
Affiliation(s)
- Ngoc Man Phan
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Thanh Loc Nguyen
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Hyunsu Shin
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Thuy An Trinh
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Jaeyun Kim
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Institute of Quantum Biophysics (IQB), Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
9
|
Mehmood A, Song S, Du X, Yan H, Wang X, Guo L, Li B. mRNA expression profile reveals differentially expressed genes in splenocytes of experimental autoimmune encephalomyelitis model. Int J Exp Pathol 2023; 104:247-257. [PMID: 37427716 PMCID: PMC10500171 DOI: 10.1111/iep.12488] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/04/2023] [Accepted: 06/18/2023] [Indexed: 07/11/2023] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a mouse model that can be used to investigate aetiology, pathogenesis, and treatment approaches for multiple sclerosis (MS). A novel integrated bioinformatics approach was used to understand the involvement of differentially expressed genes (DEGs) in the spleen of EAE mice through data mining of existing microarray and RNA-seq datasets. We screened differentially expressed mRNAs using mRNA expression profile data of EAE spleens taken from Gene Expression Omnibus (GEO). Functional and pathway enrichment analyses of DEGs were performed by Database for Annotation, Visualization, and Integrated Discovery (DAVID). Subsequently, the DEGs-encoded protein-protein interaction (PPI) network was constructed. The 784 DEGs in GSE99300 A.SW PP-EAE mice spleen mRNA profiles, 859 DEGs in GSE151701 EAE mice spleen mRNA profiles, and 646 DEGs in GSE99300 SJL/J PP-EAE mice spleen mRNA profiles were explored. Functional enrichment of 55 common DEGs among 3 sub-datasets revealed several immune-related terms, such as neutrophil extravasation, leucocyte migration, antimicrobial humoral immune response mediated by an antimicrobial peptide, toll-like receptor 4 bindings, IL-17 signalling pathway, and TGF-beta signalling pathway. In the screening of 10 hub genes, including MPO, ELANE, CTSG, LTF, LCN2, SELP, CAMP, S100A9, ITGA2B, and PRTN3, and in choosing and validating the 5 DEGs, including ANK1, MBOAT2, SLC25A21, SLC43A1, and SOX6, the results showed that SLC43A1 and SOX6 were significantly decreased in EAE mice spleen. Thus this study offers a list of genes expressed in the spleen that might play a key role in the pathogenesis of EAE.
Collapse
Affiliation(s)
- Arshad Mehmood
- Department of NeurologyThe Second Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
- Key Laboratory of Neurology of Hebei ProvinceShijiazhuangHebeiChina
| | - Shuang Song
- Department of NeurologyThe Second Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
- Key Laboratory of Neurology of Hebei ProvinceShijiazhuangHebeiChina
| | - Xiaochen Du
- Department of NeurologyThe Second Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
- Key Laboratory of Neurology of Hebei ProvinceShijiazhuangHebeiChina
| | - Hongjing Yan
- Department of NeurologyThe Second Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
- Key Laboratory of Neurology of Hebei ProvinceShijiazhuangHebeiChina
| | - Xuan Wang
- Department of NeurologyThe Second Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
- Key Laboratory of Neurology of Hebei ProvinceShijiazhuangHebeiChina
| | - Li Guo
- Department of NeurologyThe Second Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
- Key Laboratory of Neurology of Hebei ProvinceShijiazhuangHebeiChina
| | - Bin Li
- Department of NeurologyThe Second Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
- Key Laboratory of Neurology of Hebei ProvinceShijiazhuangHebeiChina
| |
Collapse
|
10
|
Jayaraman S, Jayaraman A. Impact of histone modifier-induced protection against autoimmune encephalomyelitis on multiple sclerosis treatment. Front Neurol 2022; 13:980758. [PMID: 36313502 PMCID: PMC9614082 DOI: 10.3389/fneur.2022.980758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/09/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis is a progressive demyelinating central nervous system disorder with unknown etiology. The condition has heterogeneous presentations, including relapsing-remitting multiple sclerosis and secondary and primary progressive multiple sclerosis. The genetic and epigenetic mechanisms underlying these various forms of multiple sclerosis remain elusive. Many disease-modifying therapies approved for multiple sclerosis are broad-spectrum immunomodulatory drugs that reduce relapses but do not halt the disease progression or neuroaxonal damage. Some are also associated with many severe side effects, including fatalities. Improvements in disease-modifying treatments especially for primary progressive multiple sclerosis remain an unmet need. Several experimental animal models are available to decipher the mechanisms involved in multiple sclerosis. These models help us decipher the advantages and limitations of novel disease-modifying therapies for multiple sclerosis.
Collapse
Affiliation(s)
- Sundararajan Jayaraman
- Department of Surgery, University of Illinois College of Medicine, Peoria, IL, United States
| | | |
Collapse
|
11
|
Benlaribi R, Gou Q, Takaba H. Thymic self-antigen expression for immune tolerance and surveillance. Inflamm Regen 2022; 42:28. [PMID: 36056452 PMCID: PMC9440513 DOI: 10.1186/s41232-022-00211-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 04/27/2022] [Indexed: 11/10/2022] Open
Abstract
T cells are a group of lymphocytes that play a central role in the immune system, notably, eliminating pathogens and attacking cancer while being tolerant of the self. Elucidating how immune tolerance is ensured has become a significant research issue for understanding the pathogenesis of autoimmune diseases as well as cancer immunity. T cell immune tolerance is established mainly in the thymic medulla by the removal of self-responsive T cells and the generation of regulatory T cells, this process depends mainly on the expression of a variety of tissue restricted antigens (TRAs) by medullary thymic epithelial cells (mTECs). The expression of TRAs is known to be regulated by at least two independent factors, Fezf2 and Aire, which play non-redundant and complementary roles by different mechanisms. In this review, we introduce the molecular logic of thymic self-antigen expression that underlies T cell selection for the prevention of autoimmunity and the establishment of immune surveillance.
Collapse
Affiliation(s)
- Rayene Benlaribi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Qiao Gou
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Takaba
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
12
|
Li Y, Liu X, Wang J, Pan C, Tang Z. Clinical Features and Imaging Findings of Myelin Oligodendrocyte Glycoprotein-IgG-Associated Disorder (MOGAD). Front Aging Neurosci 2022; 14:850743. [PMID: 35370624 PMCID: PMC8965323 DOI: 10.3389/fnagi.2022.850743] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 02/08/2022] [Indexed: 01/14/2023] Open
Abstract
Myelin oligodendrocyte glycoprotein-IgG-associated disorder (MOGAD) is a nervous system (NS) demyelination disease and a newly recognized distinct disease complicated with various diseases or symptoms; however, MOGAD was once considered a subset of neuromyelitis optica spectrum disorder (NMOSD). The detection of MOG-IgG has been greatly improved by the cell-based assay test method. In one study, 31% of NMOSD patients with negative aquaporin-4 (AQP-4) antibody were MOG-IgG positive. MOGAD occurs in approximately the fourth decade of a person’s life without a markedly female predominance. Usually, optic neuritis (ON), myelitis or acute disseminated encephalomyelitis (ADEM) encephalitis are the typical symptoms of MOGAD. MOG-IgG have been found in patients with peripheral neuropathy, teratoma, COVID-19 pneumonia, etc. Some studies have revealed the presence of brainstem lesions, encephalopathy or cortical encephalitis. Attention should be given to screening patients with atypical symptoms. Compared to NMOSD, MOGAD generally responds well to immunotherapy and has a good functional prognosis. Approximately 44-83% of patients undergo relapsing episodes within 8 months, which mostly involve the optic nerve, and persistently observed MOG-IgG and severe clinical performance may indicate a polyphasic course of illness. Currently, there is a lack of clinical randomized controlled trials on the treatment and prognosis of MOGAD. The purpose of this review is to discuss the clinical manifestations, imaging features, outcomes and prognosis of MOGAD.
Collapse
|
13
|
Abstract
Animal models with high translational validity are essential tools in understanding disease pathogenesis and in the development of therapeutic strategies. Multiple sclerosis (MS) is an autoimmune demyelinating disease of the central nervous system characterized by progressive neurological deficits and socioeconomic burden. Experimental autoimmune encephalomyelitis (EAE) is the most extensively utilized animal model of MS, with well-characterized rodent and non-human primate variants. The EAE model is typically induced by either active immunization with myelin-derived proteins or peptides in adjuvant or by passive transfer of activated myelin-specific CD4+ T lymphocytes. To date, the EAE model has been an essential tool in the development of at least seven U.S. Food and Drug Administration (FDA)-approved immunomodulatory drugs for the treatment of MS, including glatiramer acetate, fingolimod, and natalizumab. However, the translational validity of the EAE model is frequently compromised due to poor study design, inconsistent clinical scoring endpoints, and inappropriate statistical calculations. No single animal model accurately reflects the complexity of human MS pathogenesis. Beyond EAE, multiple additional animal models are described, including Theiler's murine encephalomyelitis virus and cuprizone-induced demyelination, which facilitate the study of pathogen-induced CNS autoimmunity and remyelination, respectively. This overview summarizes several of the most frequently used animal models of MS and highlights key factors that significantly influence the experimental outcome and affect translational validity. © 2021 Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Paul Smith
- Incyte Research Institute, Wilmington, Delaware
| |
Collapse
|
14
|
Bai P, Zhang M, Yuan J, Zhu R, Li N. A comparison of the effects of rituximab versus other immunotherapies for MOG-IgG-associated central nervous system demyelination: A meta-analysis. Mult Scler Relat Disord 2021; 53:103044. [PMID: 34091176 DOI: 10.1016/j.msard.2021.103044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/11/2021] [Accepted: 05/18/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Myelin oligodendrocyte glycoprotein (MOG) antibody disease (MOG-AD) is now recognised as a nosological entity with specific clinical and paraclinical features to aid early diagnosis. Rituximab (RTX) is a chimeric monoclonal antibody directed against CD20 epitope expressed on pre-B and mature B cells and is used to treat B-cell-derived lymphoid neoplasms and antibody-mediated autoimmune diseases. In this review, we performed a meta-analysis to evaluate RTX efficacy and assessed the treatment efficacies based on relapse rates. METHODS This study was conducted according to the PRISMA (Preferred Reporting Items for Systemic review and Meta-Analysis) statement. We searched for publications on the PubMed, Embase, Cochrane Library, clinical trials up to December 2020. We compiled 5 studies, Meta-analysis forest plots was conducted for the ARR ratio change pre and post-treatment between rituximab and other disease modifying drugs. A sensitivity analysis was performed with mean difference (MD) of the efficacy of RTX versus other immunotherapies and subgroup analysis was also performed based on site of study. RESULTS A meta-analysis of 5 studies with 239 participants was conducted. Patients have received rituximab were recorded in 82 of 239 (34.31%). The mean difference of ARR ratio of rituximab therapy versus other immunotherapies was 0.16 (95%CI, -0.15 to 0.47). No studies found to significantly affect heterogeneity. No major differences occurred in 9.2% of China patients (95% CI: -0.20-1.86; I2=0%) and 90.8% of non- China patients (95% CI: -0.24-0.42; I2=0%). Meanwhile there was no significant subgroup difference (p = 0.18) between them. CONCLUSION RTX reduces the relapse frequency in most patients with MOG antibody disease, but there is no differences between rituximab and other immunotherapies in MOG antibody disease. Future a large multicenter randomized controlled clinical trial to thoroughly characterize the efficacy of rituximab for MOG antibody disease is necessary.
Collapse
Affiliation(s)
- Peng Bai
- Department of Neurology, Inner Mongolia People's Hospital No.20 of Zhaowuda Road, Hohhot 010017, Inner Mongolia, People's Republic of China.
| | - Meini Zhang
- Department of Neurology, First Hospital of Shanxi Medical University, No. 85 Jiefangnan Road, Taiyuan 030001, Shanxi, People's Republic of China.
| | - Jun Yuan
- Department of Neurology, Inner Mongolia People's Hospital No.20 of Zhaowuda Road, Hohhot 010017, Inner Mongolia, People's Republic of China
| | - Runxiu Zhu
- Department of Neurology, Inner Mongolia People's Hospital No.20 of Zhaowuda Road, Hohhot 010017, Inner Mongolia, People's Republic of China
| | - Na Li
- Department of Neurology, Inner Mongolia People's Hospital No.20 of Zhaowuda Road, Hohhot 010017, Inner Mongolia, People's Republic of China
| |
Collapse
|
15
|
Xu J, Liu L, Xiong J, Zhang L, Huang P, Tang L, Xiao Y, Li X, Li J, Luo Y, Li H, Mao D, Liu L. The Clinical, Radiologic, and Prognostic Differences Between Pediatric and Adult Patients With Myelin Oligodendrocyte Glycoprotein Antibody-Associated Encephalomyelitis. Front Neurol 2021; 12:679430. [PMID: 34093424 PMCID: PMC8173107 DOI: 10.3389/fneur.2021.679430] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 04/16/2021] [Indexed: 11/24/2022] Open
Abstract
Purpose: To evaluate the clinical differences between pediatric and adult patients with myelin oligodendrocyte glycoprotein antibody-associated encephalomyelitis (MOG-EM). Methods: We retrospectively reviewed the clinical features of pediatric and adult patients with MOG-EM in our center between November 2015 and October 2020. Results: Twenty-eight pediatric patients and 25 adults were admitted to our study. Bilateral optic neuritis (BON) was the most common initial phenotype in the pediatric group but less common in the adult group (28.57 vs. 0%, p = 0.0119). Almost half of the adult patients presented with neuromyelitis optica spectrum disease (NMOSD), which was less prevalent among the pediatrics (48 vs. 21.43%, p = 0.0414). Visual impairment was the most common symptom in both groups during the initial attack (pediatric group, 39.29%; adult group, 64%) and throughout the full course (pediatric group, 57.14%; adult group, 72%). More pediatric patients suffered from fever than adult patients at onset (pediatric group, 28.57%; adult group, 4%; p = 0.0442) and throughout the full course (pediatric group, 39.29%; adult group, 12%; p = 0.0245). Multiple patchy lesions in subcortical white matter (pediatric group, 40.74%; adult group, 45%), periventricular (pediatric group, 25.93%; adult group, 35%), infratentorial (pediatric group, 18.52%; adult group, 30%) and deep gray matter (pediatric group, 25.93%; adult group, 20%) were frequent in all cases, no significant difference was found between the two groups, while bilateral optic nerve involvement was more frequent in pediatric group (61.54 vs. 14.29%, p = 0.0042) and unilateral optic nerve involvement was higher in adult group (64.29 vs. 15.38%, p = 0.0052). At the last follow-up, adult patients had a higher average EDSS score (median 1.0, range 0–3) than pediatrics (median 0.0, range 0–3), though not significant (p = 0.0752). Patients aged 0–9 years (61.54%) and 10–18 years (70%), and patients presenting with encephalitis/meningoencephalitis (100%) and ADEM (75%) were more likely to recover fully. Conclusions: Visual impairment was the dominant symptom in both pediatric and adult patients, while fever was more frequent in pediatric patients. Data suggested that BON and bilateral optic nerve involvement were more common in pediatric cases whereas NMOSD and unilateral optic nerve involvement were more prevalent in adults. The younger patients and patients presenting with encephalitis/meningoencephalitis and ADEM tended to recover better.
Collapse
Affiliation(s)
- Jie Xu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lingjuan Liu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jie Xiong
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lu Zhang
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Peng Huang
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Li Tang
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yangyang Xiao
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xingfang Li
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jian Li
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yingying Luo
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Huiling Li
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Dingan Mao
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Liqun Liu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
16
|
Baker D, Nutma E, O'Shea H, Cooke A, Orian JM, Amor S. Autoimmune encephalomyelitis in NOD mice is not initially a progressive multiple sclerosis model. Ann Clin Transl Neurol 2019; 6:1362-1372. [PMID: 31402611 PMCID: PMC6689692 DOI: 10.1002/acn3.792] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/12/2019] [Accepted: 04/23/2019] [Indexed: 01/31/2023] Open
Abstract
Objective Despite progress in treating relapsing multiple sclerosis (MS), effective inhibition of nonrelapsing progressive MS is an urgent, unmet, clinical need. Animal models of MS, such as experimental autoimmune encephalomyelitis (EAE), provide valuable tools to examine the mechanisms contributing to disease and may be important for developing rational therapeutic approaches for treatment of progressive MS. It has been suggested that myelin oligodendrocyte glycoprotein (MOG) peptide residues 35‐55 (MOG35‐55)‐induced EAE in nonobese diabetic (NOD) mice resembles secondary progressive MS. The objective was to determine whether the published data merits such claims. Methods Induction and monitoring of EAE in NOD mice and literature review. Results It is evident that the NOD mouse model lacks validity as a progressive MS model as the individual course seems to be an asynchronous, relapsing‐remitting neurodegenerative disease, characterized by increasingly poor recovery from relapse. The seemingly progressive course seen in group means of clinical score is an artifact of data handling and interpretation. Interpretation Although MOG35‐55‐induced EAE in NOD mice may provide some clues about approaches to block neurodegeneration associated with the inflammatory penumbra as lesions form, it should not be used to justify trials in people with nonactive, progressive MS. This adds further support to the view that drug studies in animals should universally adopt transparent raw data deposition as part of the publication process, such that claims can adequately be interrogated. This transparency is important if animal‐based science is to remain a credible part of translational research in MS.
Collapse
Affiliation(s)
- David Baker
- BartsMS, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, United Kingdom
| | - Erik Nutma
- Department of Pathology, Amsterdam UMC, Location VUmc, Amsterdam, 1081HV, The Netherlands
| | - Helen O'Shea
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, United Kingdom
| | - Anne Cooke
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, United Kingdom
| | - Jacqueline M Orian
- La Trobe Institute of Molecular Sciences La Trobe University, Bundoora, Victoria, 3086, Australia
| | - Sandra Amor
- BartsMS, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, United Kingdom.,Department of Pathology, Amsterdam UMC, Location VUmc, Amsterdam, 1081HV, The Netherlands
| |
Collapse
|
17
|
Dingman R, Balu-Iyer SV. Immunogenicity of Protein Pharmaceuticals. J Pharm Sci 2019; 108:1637-1654. [PMID: 30599169 PMCID: PMC6720129 DOI: 10.1016/j.xphs.2018.12.014] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 02/07/2023]
Abstract
Protein therapeutics have drastically changed the landscape of treatment for many diseases by providing a regimen that is highly specific and lacks many off-target toxicities. The clinical utility of many therapeutic proteins has been undermined by the potential development of unwanted immune responses against the protein, limiting their efficacy and negatively impacting its safety profile. This review attempts to provide an overview of immunogenicity of therapeutic proteins, including immune mechanisms and factors influencing immunogenicity, impact of immunogenicity, preclinical screening methods, and strategies to mitigate immunogenicity.
Collapse
Affiliation(s)
- Robert Dingman
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214
| | - Sathy V Balu-Iyer
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214.
| |
Collapse
|
18
|
Selek S, Esrefoglu M, Meral I, Bulut H, Caglar HG, Sonuc G, Yildiz C, Teloglu ES, Dogan N, Yuce B, Tiftik E, Bayindir N. Effects of Oenothera biennis L. and Hypericum perforatum L. extracts on some central nervous system myelin proteins, brain histopathology and oxidative stress in mice with experimental autoimmune encephalomyelitis. Biotech Histochem 2019; 94:75-83. [PMID: 30957550 DOI: 10.1080/10520295.2018.1482001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
We investigated the effects of Oenothera biennis L. and Hypericum perforatum L. extracts on brain tissue histopathology, myelin oligodendrocyte glycoprotein (MOG), myelin basic protein (MBP), total antioxidant status (TAS), total oxidant status (TOS) and oxidative stress index (OSI) in mice with experimental autoimmune encephalomyelitis (EAE). Forty-seven C57BL/6J mice were divided into the following groups: multiple sclerosis (MS), control (healthy mice), MS + H. perforatum treated (MS + HP), MS + O. biennis treated (MS + OB). All groups except the control group were immunized by EAE methods. Two weeks after the immunization, the mice in the MS + HP group were fed normal food containing 18 - 21 g/kg H. perforatum extract, the mice in MS + OB group were fed normal food containing 18 - 21 g/kg O. biennis extract, and the mice in control and MS groups were fed normal food for six weeks. Brain tissue samples were collected from all mice for histopathological and biochemical analysis. Clinical signs of the disease were scored using functional systems scores (FSS) daily. The H. perforatum and O. biennis extracts ameliorated the increased brain tissue MOG and MBP values for animals with MS. H. perforatum and O. biennis extract decreased the TOS and OSI values for brain tissue and increased TAS levels in brain tissue of animals with MS. In addition, H. perforatum and O. biennis extracts decreased the clinical signs at the end of the experiment compared to the beginning of extract administration. We found that myelin was lost in MS group vs. control group. H. perforatum and O. biennis extract treatments decreased the amount of myelin loss in the MS + HP and MS + OB groups. We also observed amyloid deposition on vascular walls, in the cytoplasm of the neurons and in the intercellular space in the MS group. O. biennis and H. perforatum treated groups exhibited neither abnormal amyloid deposition nor obvious cell infiltration. The beneficial effects of O. biennis and H. perforatum for attenuating myelin loss and amyloid deposition suggest their therapeutic utility for treatment of MS.
Collapse
Affiliation(s)
- S Selek
- a Departments of Medical Biochemistry , Bezmialem Vakif University , Istanbul , Turkey
| | - M Esrefoglu
- b Histology and Embryology , Bezmialem Vakif University , Istanbul , Turkey
| | - I Meral
- c Physiology Faculty of Medicine , Bezmialem Vakif University , Istanbul , Turkey
| | - H Bulut
- a Departments of Medical Biochemistry , Bezmialem Vakif University , Istanbul , Turkey
| | - H G Caglar
- a Departments of Medical Biochemistry , Bezmialem Vakif University , Istanbul , Turkey
| | - G Sonuc
- d School of Medicine , Bezmialem Vakif University , Istanbul , Turkey
| | - C Yildiz
- d School of Medicine , Bezmialem Vakif University , Istanbul , Turkey
| | - E S Teloglu
- d School of Medicine , Bezmialem Vakif University , Istanbul , Turkey
| | - N Dogan
- d School of Medicine , Bezmialem Vakif University , Istanbul , Turkey
| | - B Yuce
- d School of Medicine , Bezmialem Vakif University , Istanbul , Turkey
| | - E Tiftik
- d School of Medicine , Bezmialem Vakif University , Istanbul , Turkey
| | - N Bayindir
- b Histology and Embryology , Bezmialem Vakif University , Istanbul , Turkey
| |
Collapse
|
19
|
Djedovic N, Mansilla MJ, Jevtić B, Navarro-Barriuso J, Saksida T, Martínez-Cáceres EM, Miljković Ð. Ethyl Pyruvate Induces Tolerogenic Dendritic Cells. Front Immunol 2019; 10:157. [PMID: 30792716 PMCID: PMC6374627 DOI: 10.3389/fimmu.2019.00157] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 01/17/2019] [Indexed: 01/03/2023] Open
Abstract
Dendritic cells (DC) are professional antigen presenting cells that have a key role in shaping the immune response. Tolerogenic DC (tolDC) have immuno-regulatory properties and they are a promising prospective therapy for multiple sclerosis and other autoimmune diseases. Ethyl pyruvate (EP) is a redox analog of dimethyl fumarate (Tecfidera), a drug for multiple sclerosis treatment. We have recently shown that EP ameliorates experimental autoimmune encephalomyelitis, a multiple sclerosis murine model. Here, we expanded our study to its tolerogenic effects on DC. Phenotypic analysis has shown that DC obtained from mice or humans reduce expression of molecules required for T cell activation such as CD86, CD83, and HLA-DR under the influence of EP, while CD11c expression and viability of DC are not affected. Furthermore, EP-treated DC restrain proliferation and modulate cytokine production of allogeneic lymphocytes. These results demonstrate that EP has the ability to direct DC toward tolDC.
Collapse
Affiliation(s)
- Neda Djedovic
- Department of Immunology, Institute for Biological Research "Siniša Stanković" University of Belgrade, Belgrade, Serbia
| | - María José Mansilla
- Immunology Division, Germans Trias i Pujol University Hospital and Research Institute, Badalona, Spain.,Department of Cellular Biology, Physiology, and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Bojan Jevtić
- Department of Immunology, Institute for Biological Research "Siniša Stanković" University of Belgrade, Belgrade, Serbia
| | - Juan Navarro-Barriuso
- Immunology Division, Germans Trias i Pujol University Hospital and Research Institute, Badalona, Spain.,Department of Cellular Biology, Physiology, and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Tamara Saksida
- Department of Immunology, Institute for Biological Research "Siniša Stanković" University of Belgrade, Belgrade, Serbia
| | - Eva M Martínez-Cáceres
- Immunology Division, Germans Trias i Pujol University Hospital and Research Institute, Badalona, Spain.,Department of Cellular Biology, Physiology, and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Ðorđe Miljković
- Department of Immunology, Institute for Biological Research "Siniša Stanković" University of Belgrade, Belgrade, Serbia
| |
Collapse
|
20
|
Abstract
Current multiple sclerosis (MS) therapies are effective in reducing relapse rate, short-term measures of disability, and magnetic resonance imaging (MRI) measures of inflammation in relapsing remitting MS (RRMS), whereas in progressive/degenerative disease phases these medications are of little or no benefit. Therefore, the development of new therapies aimed at reversing neurodegeneration is of great interest. Remyelination, which is usually a spontaneous endogenous process, is achieved when myelin-producing oligodendrocytes are generated from oligodendrocyte precursor cells (OPCs). Even though these precursor cells are abundant in MS brains, their regeneration capacity is limited. Enhancing the generation of myelin-producing cells is therefore a major focus of MS research. Here we present an overview of the different advancements in the field of remyelination, including suitable animal models for testing remyelination therapies, approved medications with a proposed role in regeneration, myelin repair treatments under investigation in clinical trials, as well as future therapeutics aimed at facilitating myelin repair.
Collapse
Affiliation(s)
- David Kremer
- Department of Neurology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany
| | - Rainer Akkermann
- Department of Neurology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany
| | - Patrick Küry
- Department of Neurology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany
| | - Ranjan Dutta
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio-44195
| |
Collapse
|
21
|
Wynford-Thomas R, Jacob A, Tomassini V. Neurological update: MOG antibody disease. J Neurol 2018; 266:1280-1286. [PMID: 30569382 PMCID: PMC6469662 DOI: 10.1007/s00415-018-9122-2] [Citation(s) in RCA: 164] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 11/09/2018] [Accepted: 11/12/2018] [Indexed: 12/25/2022]
Abstract
Myelin oligodendrocyte glycoprotein (MOG) antibody disease (MOG-AD) is now recognised as a nosological entity with specific clinical and paraclinical features to aid early diagnosis. Although no age group is exempt, median age of onset is within the fourth decade of life, with optic neuritis being the most frequent presenting phenotype. Disease course can be either monophasic or relapsing, with subsequent relapses most commonly involving the optic nerve. Residual disability develops in 50-80% of patients, with transverse myelitis at onset being the most significant predictor of long-term outcome. Recent advances in MOG antibody testing offer improved sensitivity and specificity. To avoid misdiagnosis, MOG antibody testing should be undertaken in selected cases presenting clinical and paraclinical features that are felt to be in keeping with MOG-AD, using a validated cell-based assay. MRI characteristics can help in differentiating MOG-AD from other neuroinflammatory disorders, including multiple sclerosis and neuromyelitis optica. Cerebrospinal fluid oligoclonal bands are uncommon. Randomised control trials are limited, but observational open-label experience suggests a role for high-dose steroids and plasma exchange in the treatment of acute attacks, and for immunosuppressive therapies, such as steroids, oral immunosuppressants and rituximab as maintenance treatment.
Collapse
Affiliation(s)
- Ray Wynford-Thomas
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University School of Medicine, University Hospital of Wales, Heath Park, Cardiff, CF14 4XN, UK.,Helen Durham Centre for Neuroinflammation, University Hospital of Wales, Cardiff, UK
| | - Anu Jacob
- Walton Centre NHS Foundation Trust, Liverpool, UK
| | - Valentina Tomassini
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University School of Medicine, University Hospital of Wales, Heath Park, Cardiff, CF14 4XN, UK. .,Helen Durham Centre for Neuroinflammation, University Hospital of Wales, Cardiff, UK. .,Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff, UK.
| |
Collapse
|
22
|
The possible anti-apoptotic and antioxidant effects of acetyl l-carnitine as an add-on therapy on a relapsing-remitting model of experimental autoimmune encephalomyelitis in rats. Biomed Pharmacother 2018; 103:1302-1311. [DOI: 10.1016/j.biopha.2018.04.173] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/22/2018] [Accepted: 04/23/2018] [Indexed: 01/02/2023] Open
|
23
|
Wang SS, Zhang Z, Zhu TB, Chu SF, He WB, Chen NH. Myelin injury in the central nervous system and Alzheimer's disease. Brain Res Bull 2018; 140:162-168. [PMID: 29730417 DOI: 10.1016/j.brainresbull.2018.05.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/18/2018] [Accepted: 05/02/2018] [Indexed: 12/20/2022]
Abstract
Myelin is a membrane wrapped around the axon of the nerve cell, which is composed of the mature oligodendrocytes. The role of myelin is to insulate and prevent the nerve electrical impulses from the axon of the neurons to the axons of the other neurons, which is essential for the proper functioning of the nervous system. Minor changes in myelin thickness could lead to substantial changes in conduction speed and may thus alter neural circuit function. Demyelination is the myelin damage, which characterized by the loss of nerve sheath and the relative fatigue of the neuronal sheath and axon. Studies have shown that myelin injury may be closely related to neurodegenerative diseases and may be an early diagnostic criteria and therapeutic target. Thus this review summarizes the recent result of pathologic effect and signal pathways of myelin injury in neurodegenerative diseases, especially the Alzheimer's disease to provide new and effective therapeutic targets.
Collapse
Affiliation(s)
- Sha-Sha Wang
- School of Basic Medicine, Shanxi University of Traditional Chinese Medicine, Taiyuan 030619, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Tian-Bi Zhu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Shi-Feng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wen-Bin He
- School of Basic Medicine, Shanxi University of Traditional Chinese Medicine, Taiyuan 030619, China
| | - Nai-Hong Chen
- School of Basic Medicine, Shanxi University of Traditional Chinese Medicine, Taiyuan 030619, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
24
|
Bjelobaba I, Begovic-Kupresanin V, Pekovic S, Lavrnja I. Animal models of multiple sclerosis: Focus on experimental autoimmune encephalomyelitis. J Neurosci Res 2018; 96:1021-1042. [PMID: 29446144 DOI: 10.1002/jnr.24224] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 01/15/2018] [Accepted: 01/25/2018] [Indexed: 12/15/2022]
Abstract
Multiple sclerosis (MS) is a chronic, progressive disorder of the central nervous system (CNS) that affects more than two million people worldwide. Several animal models resemble MS pathology; the most employed are experimental autoimmune encephalomyelitis (EAE) and toxin- and/or virus-induced demyelination. In this review we will summarize our knowledge on the utility of different animal models in MS research. Although animal models cannot replicate the complexity and heterogeneity of the MS pathology, they have proved to be useful for the development of several drugs approved for treatment of MS patients. This review focuses on EAE because it represents both clinical and pathological features of MS. During the past decades, EAE has been effective in illuminating various pathological processes that occur during MS, including inflammation, CNS penetration, demyelination, axonopathy, and neuron loss mediated by immune cells.
Collapse
Affiliation(s)
- Ivana Bjelobaba
- Institute for Biological Research "Sinisa Stankovic," Department of Neurobiology, University of Belgrade, Belgrade, Serbia
| | | | - Sanja Pekovic
- Institute for Biological Research "Sinisa Stankovic," Department of Neurobiology, University of Belgrade, Belgrade, Serbia
| | - Irena Lavrnja
- Institute for Biological Research "Sinisa Stankovic," Department of Neurobiology, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
25
|
Jurynczyk M, Geraldes R, Probert F, Woodhall MR, Waters P, Tackley G, DeLuca G, Chandratre S, Leite MI, Vincent A, Palace J. Distinct brain imaging characteristics of autoantibody-mediated CNS conditions and multiple sclerosis. Brain 2017; 140:617-627. [DOI: 10.1093/brain/aww350] [Citation(s) in RCA: 170] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 11/20/2016] [Indexed: 12/25/2022] Open
|
26
|
Kulkarni P, Yellanki S, Medishetti R, Sriram D, Saxena U, Yogeeswari P. Novel Zebrafish EAE model: A quick in vivo screen for multiple sclerosis. Mult Scler Relat Disord 2017; 11:32-39. [DOI: 10.1016/j.msard.2016.11.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 11/07/2016] [Accepted: 11/26/2016] [Indexed: 12/14/2022]
|
27
|
Sahraian MA, Owji M, Naser Moghadasi A. Concomitant multiple sclerosis and another autoimmune disease: Does the clinical course change? Clin Neurol Neurosurg 2016; 150:92-95. [DOI: 10.1016/j.clineuro.2016.09.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Revised: 08/27/2016] [Accepted: 09/02/2016] [Indexed: 01/27/2023]
|
28
|
Breser ML, Lino AC, Motrich RD, Godoy GJ, Demengeot J, Rivero VE. Regulatory T cells control strain specific resistance to Experimental Autoimmune Prostatitis. Sci Rep 2016; 6:33097. [PMID: 27624792 PMCID: PMC5022010 DOI: 10.1038/srep33097] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 08/19/2016] [Indexed: 12/18/2022] Open
Abstract
Susceptibility to autoimmune diseases results from the encounter of a complex and long evolved genetic context with a no less complex and changing environment. Major actors in maintaining health are regulatory T cells (Treg) that primarily dampen a large subset of autoreactive lymphocytes escaping thymic negative selection. Here, we directly asked whether Treg participate in defining susceptibility and resistance to Experimental Autoimmune Prostatitis (EAP). We analyzed three common laboratory strains of mice presenting with different susceptibility to autoimmune prostatitis upon immunization with prostate proteins. The NOD, the C57BL/6 and the BALB/c mice that can be classified along a disease score ranging from severe, mild and to undetectable, respectively. Upon mild and transient depletion of Treg at the induction phase of EAP, each model showed an increment along this score, most remarkably with the BALB/c mice switching from a resistant to a susceptible phenotype. We further show that disease associates with the upregulation of CXCR3 expression on effector T cells, a process requiring IFNγ. Together with recent advances on environmental factors affecting Treg, these findings provide a likely cellular and molecular explanation to the recent rise in autoimmune diseases incidence.
Collapse
Affiliation(s)
- Maria L Breser
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, Ciudad Universitaria, 5016, Córdoba, Argentina
| | | | - Ruben D Motrich
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, Ciudad Universitaria, 5016, Córdoba, Argentina
| | - Gloria J Godoy
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, Ciudad Universitaria, 5016, Córdoba, Argentina
| | | | - Virginia E Rivero
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, Ciudad Universitaria, 5016, Córdoba, Argentina
| |
Collapse
|
29
|
Nissen JC, Tsirka SE. Tuftsin-driven experimental autoimmune encephalomyelitis recovery requires neuropilin-1. Glia 2016; 64:923-36. [PMID: 26880314 DOI: 10.1002/glia.22972] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 12/31/2015] [Accepted: 01/12/2016] [Indexed: 01/01/2023]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is an animal model of demyelinating autoimmune disease, such as multiple sclerosis (MS), which is characterized by central nervous system white matter lesions, microglial activation, and peripheral T-cell infiltration secondary to blood-brain barrier disruption. We have previously shown that treatment with tuftsin, a tetrapeptide generated from IgG proteolysis, dramatically improves disease symptoms in EAE. Here, we report that microglial expression of Neuropilin-1 (Nrp1) is required for tuftsin-driven amelioration of EAE symptoms. Nrp1 ablation in microglia blocks microglial signaling and polarization to the anti-inflammatory M2 phenotype, and ablation in either the microglia or immunosuppressive regulatory T cells (Tregs) reduces extended functional contacts between them and Treg activation, implicating a role for microglia in the activation process, and more generally, how immune surveillance is conducted in the CNS. Taken together, our findings delineate the mechanistic action of tuftsin as a candidate therapeutic against immune-mediated demyelinating lesions.
Collapse
Affiliation(s)
- Jillian C Nissen
- Program in Molecular and Cellular Pharmacology, Department of Pharmacological Sciences, Stony Brook University, New York, New York
| | - Stella E Tsirka
- Program in Molecular and Cellular Pharmacology, Department of Pharmacological Sciences, Stony Brook University, New York, New York
| |
Collapse
|
30
|
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
|
31
|
Pacini G, Ieronymaki M, Nuti F, Sabatino G, Larregola M, Aharoni R, Papini AM, Rovero P. Epitope mapping of anti-myelin oligodendrocyte glycoprotein (MOG) antibodies in a mouse model of multiple sclerosis: microwave-assisted synthesis of the peptide antigens and ELISA screening. J Pept Sci 2015; 22:52-8. [PMID: 26663200 DOI: 10.1002/psc.2839] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 10/28/2015] [Accepted: 10/28/2015] [Indexed: 01/06/2023]
Abstract
The role of pathologic auto-antibodies against myelin oligodendrocyte glycoprotein (MOG) in multiple sclerosis is a highly controversial matter. As the use of animal models may enable to unravel the molecular mechanisms of the human disorder, numerous studies on multiple sclerosis are carried out using experimental autoimmune encephalomyelitis (EAE). In particular, the most extensively used EAE model is obtained by immunizing C57BL/6 mice with the immunodominant peptide MOG(35-55). In this scenario, we analyzed the anti-MOG antibody response in this model using the recombinant refolded extracellular domain of the protein, MOG(1-117). To assess the presence of a B-cell intramolecular epitope spreading mechanism, we tested also five synthetic peptides mapping the 1-117 sequence of MOG, including MOG(35-55). For this purpose, we cloned, expressed in Escherichia coli and on-column refolded MOG(1-117), and we applied an optimized microwave-assisted solid-phase synthetic strategy to obtain the designed peptide sequences. Subsequently, we set up a solid-phase immunoenzymatic assay testing both naïve and EAE mice sera and using MOG protein and peptides as antigenic probes. The results obtained disclose an intense IgG antibody response against both the recombinant protein and the immunizing peptide, while no response was observed against the other synthetic fragments, thus excluding the presence of an intramolecular epitope spreading mechanism. Furthermore, as the properly refolded recombinant probe is able to bind antibodies with greater efficiency compared with MOG(35-55), we hypothesize the presence of both linear and conformational epitopes on MOG(35-55) sequence.
Collapse
Affiliation(s)
- Giulia Pacini
- French-Italian Interdepartmental Laboratory of Peptide and Protein Chemistry and Biology - PeptLab, Florence, Italy and Cergy-Pontoise, France.,Department NeuroFarBa, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Via Ugo Schiff 6, Sesto Fiorentino, Florence, I-50019, Italy
| | - Matthaia Ieronymaki
- French-Italian Interdepartmental Laboratory of Peptide and Protein Chemistry and Biology - PeptLab, Florence, Italy and Cergy-Pontoise, France.,Department NeuroFarBa, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Via Ugo Schiff 6, Sesto Fiorentino, Florence, I-50019, Italy.,Laboratoire de Chimie Biologique EA4505, University of Cergy-Pontoise, 5 mail Gay-Lussac Neuville-sur-Oise, Cergy-Pontoise, 95000, France
| | - Francesca Nuti
- French-Italian Interdepartmental Laboratory of Peptide and Protein Chemistry and Biology - PeptLab, Florence, Italy and Cergy-Pontoise, France.,Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3/13, Sesto Fiorentino, Florence, I-50019, Italy
| | - Giuseppina Sabatino
- French-Italian Interdepartmental Laboratory of Peptide and Protein Chemistry and Biology - PeptLab, Florence, Italy and Cergy-Pontoise, France.,Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3/13, Sesto Fiorentino, Florence, I-50019, Italy
| | - Maud Larregola
- French-Italian Interdepartmental Laboratory of Peptide and Protein Chemistry and Biology - PeptLab, Florence, Italy and Cergy-Pontoise, France.,Laboratoire de Chimie Biologique EA4505, University of Cergy-Pontoise, 5 mail Gay-Lussac Neuville-sur-Oise, Cergy-Pontoise, 95000, France
| | - Rina Aharoni
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Anna Maria Papini
- French-Italian Interdepartmental Laboratory of Peptide and Protein Chemistry and Biology - PeptLab, Florence, Italy and Cergy-Pontoise, France.,Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3/13, Sesto Fiorentino, Florence, I-50019, Italy.,Laboratoire de Chimie Biologique EA4505, University of Cergy-Pontoise, 5 mail Gay-Lussac Neuville-sur-Oise, Cergy-Pontoise, 95000, France
| | - Paolo Rovero
- French-Italian Interdepartmental Laboratory of Peptide and Protein Chemistry and Biology - PeptLab, Florence, Italy and Cergy-Pontoise, France.,Department NeuroFarBa, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Via Ugo Schiff 6, Sesto Fiorentino, Florence, I-50019, Italy
| |
Collapse
|
32
|
Experimental autoimmune encephalomyelitis development is aggravated by Candida albicans infection. J Immunol Res 2015; 2015:635052. [PMID: 25969836 PMCID: PMC4417602 DOI: 10.1155/2015/635052] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 01/19/2015] [Accepted: 01/22/2015] [Indexed: 12/31/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory/autoimmune disease of the central nervous system (CNS) mainly mediated by myelin specific T cells. It is widely believed that environmental factors, including fungal infections, contribute to disease induction or evolution. Even though Candida infection among MS patients has been described, the participation of this fungus in this pathology is not clear. The purpose of this work was to evaluate the effect of a Candida albicans infection on experimental autoimmune encephalomyelitis (EAE) that is a widely accepted model to study MS. Female C57BL/6 mice were infected with C. albicans and 3 days later, animals were submitted to EAE induction by immunization with myelin oligodendrocyte glycoprotein. Previous infection increased the clinical score and also the body weight loss. EAE aggravation was associated with expansion of peripheral CD4+ T cells and production of high levels of TNF-α, IFN-γ IL-6, and IL-17 by spleen and CNS cells. In addition to yeast and hyphae, fungus specific T cells were found in the CNS. These findings suggest that C. albicans infection before EAE induction aggravates EAE, and possibly MS, mainly by CNS dissemination and local induction of encephalitogenic cytokines. Peripheral production of encephalitogenic cytokines could also contribute to disease aggravation.
Collapse
|
33
|
Haines JD, Herbin O, de la Hera B, Vidaurre OG, Moy GA, Sun Q, Fung HYJ, Albrecht S, Alexandropoulos K, McCauley D, Chook YM, Kuhlmann T, Kidd GJ, Shacham S, Casaccia P. Nuclear export inhibitors avert progression in preclinical models of inflammatory demyelination. Nat Neurosci 2015; 18:511-20. [PMID: 25706475 PMCID: PMC4522902 DOI: 10.1038/nn.3953] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 01/22/2015] [Indexed: 12/13/2022]
Abstract
Axonal damage has been associated with aberrant protein trafficking. We examined a newly characterized class of compounds that target nucleo-cytoplasmic shuttling by binding to the catalytic groove of the nuclear export protein XPO1 (also known as CRM1, chromosome region maintenance protein 1). Oral administration of reversible CRM1 inhibitors in preclinical murine models of demyelination significantly attenuated disease progression, even when started after the onset of paralysis. Clinical efficacy was associated with decreased proliferation of immune cells, characterized by nuclear accumulation of cell cycle inhibitors, and preservation of cytoskeletal integrity even in demyelinated axons. Neuroprotection was not limited to models of demyelination, but was also observed in another mouse model of axonal damage (that is, kainic acid injection) and detected in cultured neurons after knockdown of Xpo1, the gene encoding CRM1. A proteomic screen for target molecules revealed that CRM1 inhibitors in neurons prevented nuclear export of molecules associated with axonal damage while retaining transcription factors modulating neuroprotection.
Collapse
MESH Headings
- Acrylamides/administration & dosage
- Acrylamides/pharmacokinetics
- Acrylamides/pharmacology
- Active Transport, Cell Nucleus/drug effects
- Animals
- Axons/drug effects
- Axons/metabolism
- Axons/pathology
- Cell Nucleus/metabolism
- Cells, Cultured
- Disease Models, Animal
- Disease Progression
- Drug Evaluation, Preclinical
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Female
- Karyopherins/antagonists & inhibitors
- Karyopherins/genetics
- Karyopherins/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neuroprotective Agents/administration & dosage
- Neuroprotective Agents/pharmacokinetics
- Neuroprotective Agents/pharmacology
- Proteomics
- Rats
- Rats, Sprague-Dawley
- Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Thiazoles/administration & dosage
- Thiazoles/pharmacokinetics
- Thiazoles/pharmacology
- Treatment Outcome
- Exportin 1 Protein
Collapse
Affiliation(s)
- Jeffery D. Haines
- Department of Neuroscience and Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Olivier Herbin
- Department of Medicine, Division of Clinical Immunology, The Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Belén de la Hera
- Department of Neuroscience and Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Oscar G. Vidaurre
- Department of Neuroscience and Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gregory A. Moy
- Department of Neuroscience and Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Qingxiang Sun
- Department of Pharmacology, University of Texas Southwestern Medical Center, 6001 Forest Park Road, Dallas, TX 75390-9041
| | - Ho Yee Joyce Fung
- Department of Pharmacology, University of Texas Southwestern Medical Center, 6001 Forest Park Road, Dallas, TX 75390-9041
| | - Stephanie Albrecht
- Institute of Neuropathology, University Hospital Münster, Pottkamp 2, 48149 Münster, Germany
| | - Konstantina Alexandropoulos
- Department of Medicine, Division of Clinical Immunology, The Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dilara McCauley
- Karyopharm Therapeutics, 2 Mercer Road, Natick, MA 01760, USA
| | - Yuh Min Chook
- Department of Pharmacology, University of Texas Southwestern Medical Center, 6001 Forest Park Road, Dallas, TX 75390-9041
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Münster, Pottkamp 2, 48149 Münster, Germany
| | - Grahame J. Kidd
- Department of Neurosciences, Cleveland Clinic, 4500 Euclid Ave, Cleveland, OH, 44195, USA
| | - Sharon Shacham
- Karyopharm Therapeutics, 2 Mercer Road, Natick, MA 01760, USA
| | - Patrizia Casaccia
- Department of Neuroscience and Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
34
|
Waters P, Woodhall M, O'Connor KC, Reindl M, Lang B, Sato DK, Juryńczyk M, Tackley G, Rocha J, Takahashi T, Misu T, Nakashima I, Palace J, Fujihara K, Leite MI, Vincent A. MOG cell-based assay detects non-MS patients with inflammatory neurologic disease. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2015; 2:e89. [PMID: 25821844 PMCID: PMC4370386 DOI: 10.1212/nxi.0000000000000089] [Citation(s) in RCA: 307] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 01/20/2015] [Indexed: 11/29/2022]
Abstract
Objective: To optimize sensitivity and disease specificity of a myelin oligodendrocyte glycoprotein (MOG) antibody assay. Methods: Consecutive sera (n = 1,109) sent for aquaporin-4 (AQP4) antibody testing were screened for MOG antibodies (Abs) by cell-based assays using either full-length human MOG (FL-MOG) or the short-length form (SL-MOG). The Abs were initially detected by Alexa Fluor goat anti-human IgG (H + L) and subsequently by Alexa Fluor mouse antibodies to human IgG1. Results: When tested at 1:20 dilution, 40/1,109 sera were positive for AQP4-Abs, 21 for SL-MOG, and 180 for FL-MOG. Only one of the 40 AQP4-Ab–positive sera was positive for SL-MOG-Abs, but 10 (25%) were positive for FL-MOG-Abs (p = 0.0069). Of equal concern, 48% (42/88) of sera from controls (patients with epilepsy) were positive by FL-MOG assay. However, using an IgG1-specific secondary antibody, only 65/1,109 (5.8%) sera were positive on FL-MOG, and AQP4-Ab– positive and control sera were negative. IgM reactivity accounted for the remaining anti-human IgG (H + L) positivity toward FL-MOG. The clinical diagnoses were obtained in 33 FL-MOG–positive patients, blinded to the antibody data. IgG1-Abs to FL-MOG were associated with optic neuritis (n = 11), AQP4-seronegative neuromyelitis optica spectrum disorder (n = 4), and acute disseminated encephalomyelitis (n = 1). All 7 patients with probable multiple sclerosis (MS) were MOG-IgG1 negative. Conclusions: The limited disease specificity of FL-MOG-Abs identified using Alexa Fluor goat anti-human IgG (H + L) is due in part to detection of IgM-Abs. Use of the FL-MOG and restricting to IgG1-Abs substantially improves specificity for non-MS demyelinating diseases. Classification of evidence: This study provides Class II evidence that the presence of serum IgG1- MOG-Abs in AQP4-Ab–negative patients distinguishes non-MS CNS demyelinating disorders from MS (sensitivity 24%, 95% confidence interval [CI] 9%–45%; specificity 100%, 95% CI 88%–100%).
Collapse
Affiliation(s)
- Patrick Waters
- Nuffield Department of Clinical Neurosciences (P.W., M.W., B.L., M.J., G.T., J.R., J.P., M.I.L., A.V.), John Radcliffe Hospital, Oxford, UK; Department of Neurology (K.C.O.), Yale School of Medicine, New Haven, CT; Clinical Department of Neurology (M.R.), Innsbruck Medical University, Innsbruck, Austria; Department of Neurology (D.K.S., I.N.) and Department of Multiple Sclerosis Therapeutics (T.M., K.F.) Tohoku University School of Medicine, Sendai, Japan; and Department of Neurology (T.T.), Yonezawa National Hospital, Yonezawa, Japan
| | - Mark Woodhall
- Nuffield Department of Clinical Neurosciences (P.W., M.W., B.L., M.J., G.T., J.R., J.P., M.I.L., A.V.), John Radcliffe Hospital, Oxford, UK; Department of Neurology (K.C.O.), Yale School of Medicine, New Haven, CT; Clinical Department of Neurology (M.R.), Innsbruck Medical University, Innsbruck, Austria; Department of Neurology (D.K.S., I.N.) and Department of Multiple Sclerosis Therapeutics (T.M., K.F.) Tohoku University School of Medicine, Sendai, Japan; and Department of Neurology (T.T.), Yonezawa National Hospital, Yonezawa, Japan
| | - Kevin C O'Connor
- Nuffield Department of Clinical Neurosciences (P.W., M.W., B.L., M.J., G.T., J.R., J.P., M.I.L., A.V.), John Radcliffe Hospital, Oxford, UK; Department of Neurology (K.C.O.), Yale School of Medicine, New Haven, CT; Clinical Department of Neurology (M.R.), Innsbruck Medical University, Innsbruck, Austria; Department of Neurology (D.K.S., I.N.) and Department of Multiple Sclerosis Therapeutics (T.M., K.F.) Tohoku University School of Medicine, Sendai, Japan; and Department of Neurology (T.T.), Yonezawa National Hospital, Yonezawa, Japan
| | - Markus Reindl
- Nuffield Department of Clinical Neurosciences (P.W., M.W., B.L., M.J., G.T., J.R., J.P., M.I.L., A.V.), John Radcliffe Hospital, Oxford, UK; Department of Neurology (K.C.O.), Yale School of Medicine, New Haven, CT; Clinical Department of Neurology (M.R.), Innsbruck Medical University, Innsbruck, Austria; Department of Neurology (D.K.S., I.N.) and Department of Multiple Sclerosis Therapeutics (T.M., K.F.) Tohoku University School of Medicine, Sendai, Japan; and Department of Neurology (T.T.), Yonezawa National Hospital, Yonezawa, Japan
| | - Bethan Lang
- Nuffield Department of Clinical Neurosciences (P.W., M.W., B.L., M.J., G.T., J.R., J.P., M.I.L., A.V.), John Radcliffe Hospital, Oxford, UK; Department of Neurology (K.C.O.), Yale School of Medicine, New Haven, CT; Clinical Department of Neurology (M.R.), Innsbruck Medical University, Innsbruck, Austria; Department of Neurology (D.K.S., I.N.) and Department of Multiple Sclerosis Therapeutics (T.M., K.F.) Tohoku University School of Medicine, Sendai, Japan; and Department of Neurology (T.T.), Yonezawa National Hospital, Yonezawa, Japan
| | - Douglas K Sato
- Nuffield Department of Clinical Neurosciences (P.W., M.W., B.L., M.J., G.T., J.R., J.P., M.I.L., A.V.), John Radcliffe Hospital, Oxford, UK; Department of Neurology (K.C.O.), Yale School of Medicine, New Haven, CT; Clinical Department of Neurology (M.R.), Innsbruck Medical University, Innsbruck, Austria; Department of Neurology (D.K.S., I.N.) and Department of Multiple Sclerosis Therapeutics (T.M., K.F.) Tohoku University School of Medicine, Sendai, Japan; and Department of Neurology (T.T.), Yonezawa National Hospital, Yonezawa, Japan
| | - Maciej Juryńczyk
- Nuffield Department of Clinical Neurosciences (P.W., M.W., B.L., M.J., G.T., J.R., J.P., M.I.L., A.V.), John Radcliffe Hospital, Oxford, UK; Department of Neurology (K.C.O.), Yale School of Medicine, New Haven, CT; Clinical Department of Neurology (M.R.), Innsbruck Medical University, Innsbruck, Austria; Department of Neurology (D.K.S., I.N.) and Department of Multiple Sclerosis Therapeutics (T.M., K.F.) Tohoku University School of Medicine, Sendai, Japan; and Department of Neurology (T.T.), Yonezawa National Hospital, Yonezawa, Japan
| | - George Tackley
- Nuffield Department of Clinical Neurosciences (P.W., M.W., B.L., M.J., G.T., J.R., J.P., M.I.L., A.V.), John Radcliffe Hospital, Oxford, UK; Department of Neurology (K.C.O.), Yale School of Medicine, New Haven, CT; Clinical Department of Neurology (M.R.), Innsbruck Medical University, Innsbruck, Austria; Department of Neurology (D.K.S., I.N.) and Department of Multiple Sclerosis Therapeutics (T.M., K.F.) Tohoku University School of Medicine, Sendai, Japan; and Department of Neurology (T.T.), Yonezawa National Hospital, Yonezawa, Japan
| | - Joao Rocha
- Nuffield Department of Clinical Neurosciences (P.W., M.W., B.L., M.J., G.T., J.R., J.P., M.I.L., A.V.), John Radcliffe Hospital, Oxford, UK; Department of Neurology (K.C.O.), Yale School of Medicine, New Haven, CT; Clinical Department of Neurology (M.R.), Innsbruck Medical University, Innsbruck, Austria; Department of Neurology (D.K.S., I.N.) and Department of Multiple Sclerosis Therapeutics (T.M., K.F.) Tohoku University School of Medicine, Sendai, Japan; and Department of Neurology (T.T.), Yonezawa National Hospital, Yonezawa, Japan
| | - Toshiyuki Takahashi
- Nuffield Department of Clinical Neurosciences (P.W., M.W., B.L., M.J., G.T., J.R., J.P., M.I.L., A.V.), John Radcliffe Hospital, Oxford, UK; Department of Neurology (K.C.O.), Yale School of Medicine, New Haven, CT; Clinical Department of Neurology (M.R.), Innsbruck Medical University, Innsbruck, Austria; Department of Neurology (D.K.S., I.N.) and Department of Multiple Sclerosis Therapeutics (T.M., K.F.) Tohoku University School of Medicine, Sendai, Japan; and Department of Neurology (T.T.), Yonezawa National Hospital, Yonezawa, Japan
| | - Tatsuro Misu
- Nuffield Department of Clinical Neurosciences (P.W., M.W., B.L., M.J., G.T., J.R., J.P., M.I.L., A.V.), John Radcliffe Hospital, Oxford, UK; Department of Neurology (K.C.O.), Yale School of Medicine, New Haven, CT; Clinical Department of Neurology (M.R.), Innsbruck Medical University, Innsbruck, Austria; Department of Neurology (D.K.S., I.N.) and Department of Multiple Sclerosis Therapeutics (T.M., K.F.) Tohoku University School of Medicine, Sendai, Japan; and Department of Neurology (T.T.), Yonezawa National Hospital, Yonezawa, Japan
| | - Ichiro Nakashima
- Nuffield Department of Clinical Neurosciences (P.W., M.W., B.L., M.J., G.T., J.R., J.P., M.I.L., A.V.), John Radcliffe Hospital, Oxford, UK; Department of Neurology (K.C.O.), Yale School of Medicine, New Haven, CT; Clinical Department of Neurology (M.R.), Innsbruck Medical University, Innsbruck, Austria; Department of Neurology (D.K.S., I.N.) and Department of Multiple Sclerosis Therapeutics (T.M., K.F.) Tohoku University School of Medicine, Sendai, Japan; and Department of Neurology (T.T.), Yonezawa National Hospital, Yonezawa, Japan
| | - Jacqueline Palace
- Nuffield Department of Clinical Neurosciences (P.W., M.W., B.L., M.J., G.T., J.R., J.P., M.I.L., A.V.), John Radcliffe Hospital, Oxford, UK; Department of Neurology (K.C.O.), Yale School of Medicine, New Haven, CT; Clinical Department of Neurology (M.R.), Innsbruck Medical University, Innsbruck, Austria; Department of Neurology (D.K.S., I.N.) and Department of Multiple Sclerosis Therapeutics (T.M., K.F.) Tohoku University School of Medicine, Sendai, Japan; and Department of Neurology (T.T.), Yonezawa National Hospital, Yonezawa, Japan
| | - Kazuo Fujihara
- Nuffield Department of Clinical Neurosciences (P.W., M.W., B.L., M.J., G.T., J.R., J.P., M.I.L., A.V.), John Radcliffe Hospital, Oxford, UK; Department of Neurology (K.C.O.), Yale School of Medicine, New Haven, CT; Clinical Department of Neurology (M.R.), Innsbruck Medical University, Innsbruck, Austria; Department of Neurology (D.K.S., I.N.) and Department of Multiple Sclerosis Therapeutics (T.M., K.F.) Tohoku University School of Medicine, Sendai, Japan; and Department of Neurology (T.T.), Yonezawa National Hospital, Yonezawa, Japan
| | - M Isabel Leite
- Nuffield Department of Clinical Neurosciences (P.W., M.W., B.L., M.J., G.T., J.R., J.P., M.I.L., A.V.), John Radcliffe Hospital, Oxford, UK; Department of Neurology (K.C.O.), Yale School of Medicine, New Haven, CT; Clinical Department of Neurology (M.R.), Innsbruck Medical University, Innsbruck, Austria; Department of Neurology (D.K.S., I.N.) and Department of Multiple Sclerosis Therapeutics (T.M., K.F.) Tohoku University School of Medicine, Sendai, Japan; and Department of Neurology (T.T.), Yonezawa National Hospital, Yonezawa, Japan
| | - Angela Vincent
- Nuffield Department of Clinical Neurosciences (P.W., M.W., B.L., M.J., G.T., J.R., J.P., M.I.L., A.V.), John Radcliffe Hospital, Oxford, UK; Department of Neurology (K.C.O.), Yale School of Medicine, New Haven, CT; Clinical Department of Neurology (M.R.), Innsbruck Medical University, Innsbruck, Austria; Department of Neurology (D.K.S., I.N.) and Department of Multiple Sclerosis Therapeutics (T.M., K.F.) Tohoku University School of Medicine, Sendai, Japan; and Department of Neurology (T.T.), Yonezawa National Hospital, Yonezawa, Japan
| |
Collapse
|
35
|
Homo-β-amino acid containing MBP(85-99) analogs alleviate experimental autoimmune encephalomyelitis. Sci Rep 2015; 5:8205. [PMID: 25644378 PMCID: PMC4314633 DOI: 10.1038/srep08205] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 01/13/2015] [Indexed: 11/20/2022] Open
Abstract
MBP(85–99), an immuno-dominant epitope of myelin basic protein which binds to the major histocompatibility complex haplotype HLA-DR2 is widely implicated in the pathogenesis of multiple sclerosis. J5, an antagonist of MBP(85–99), that blocks the binding of MBP(85–99) to soluble HLA-DR2b much more efficiently than glatiramer acetate (a random copolymer comprising major MHC and T-cell receptor contact residues), was transformed into analogs with superior biological half-lives and antagonistic-activities by substitution of some of its residues with homo-β-amino acids. S18, the best analog obtained ameliorated symptoms of experimental autoimmune encephalomyelitis at least twice more effectively than glatiramer acetate or J5. S18 displayed marked resistance to proteolysis in-vitro; biological impact of which was evident in the form of delayed clinical onset of disease and prolonged therapeutic-benefits. Besides active suppression of MBP(85–99)-reactive CD4+ T-cells in-vitro and in-vivo S18 treatment also generated IL-4 producing CD4+ T-cell clones, through which protective effect could be transferred passively.
Collapse
|
36
|
Dang PT, Bui Q, D'Souza CS, Orian JM. Modelling MS: Chronic-Relapsing EAE in the NOD/Lt Mouse Strain. Curr Top Behav Neurosci 2015; 26:143-177. [PMID: 26126592 DOI: 10.1007/7854_2015_378] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Modelling complex disorders presents considerable challenges, and multiple sclerosis (MS) is no exception to this rule. The aetiology of MS is unknown, and its pathophysiology is poorly understood. Moreover, the last two decades have witnessed a dramatic revision of the long-held view of MS as an inflammatory demyelinating white matter disease. Instead, it is now regarded as a global central nervous system (CNS) disorder with a neurodegenerative component. Currently, there is no animal model recapitulating MS immunopathogenesis. Available models are based on autoimmune-mediated demyelination, denoted experimental autoimmune encephalomyelitis (EAE) or virally or chemically induced demyelination. Of these, the EAE model has been the most commonly used. It has been extensively improved since its first description and now exists as a number of variants, including genetically modified and humanized versions. Nonetheless, EAE is a distinct disease, and each variant models only certain facets of MS. Whilst the search for more refined MS models must continue, it is important to further explore where mechanisms underlying EAE provide proof-of-principle for those driving MS pathogenesis. EAE variants generated with the myelin component myelin oligodendrocyte glycoprotein (MOG) have emerged as the preferred ones, because in this particular variant disease is associated with both T- and B-cell effector mechanisms, together with demyelination. MOG-induced EAE in the non-obese diabetic (NOD) mouse strain exhibits a chronic-relapsing EAE clinical profile and high disease incidence. We describe the generation of this variant, its contribution to the understanding of MS immune and pathogenetic mechanisms and potential for evaluation of candidate therapies.
Collapse
Affiliation(s)
- Phuc T Dang
- Department of Biochemistry and La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Quyen Bui
- Department of Biochemistry and La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Claretta S D'Souza
- Department of Biochemistry and La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Jacqueline M Orian
- Department of Biochemistry and La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia.
| |
Collapse
|
37
|
Dang AK, Jain RW, Craig HC, Kerfoot SM. B cell recognition of myelin oligodendrocyte glycoprotein autoantigen depends on immunization with protein rather than short peptide, while B cell invasion of the CNS in autoimmunity does not. J Neuroimmunol 2015; 278:73-84. [DOI: 10.1016/j.jneuroim.2014.12.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 11/12/2014] [Accepted: 12/08/2014] [Indexed: 10/24/2022]
|
38
|
B cell epitope spreading: mechanisms and contribution to autoimmune diseases. Immunol Lett 2014; 163:56-68. [PMID: 25445494 DOI: 10.1016/j.imlet.2014.11.001] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 11/03/2014] [Accepted: 11/05/2014] [Indexed: 01/03/2023]
Abstract
While a variety of factors act to trigger or initiate autoimmune diseases, the process of epitope spreading is an important contributor in their development. Epitope spreading is a diversification of the epitopes recognized by the immune system. This process happens to both T and B cells, with this review focusing on B cells. Such spreading can progress among multiple epitopes on a single antigen, or from one antigenic molecule to another. Systemic lupus erythematosus, multiple sclerosis, pemphigus, bullous pemphigoid and other autoimmune diseases, are all influenced by intermolecular and intramolecular B cell epitope spreading. Endocytic processing, antigen presentation, and somatic hypermutation act as molecular mechanisms that assist in driving epitope spreading and broadening the immune response in autoimmune diseases. The purpose of this review is to summarize our current understanding of B cell epitope spreading with regard to autoimmunity, how it contributes during the progression of various autoimmune diseases, and treatment options available.
Collapse
|
39
|
Flügel A, Schläger C, Lühder F, Odoardi F. Autoimmune disease in the brain--how to spot the culprits and how to keep them in check. J Neurol Sci 2014; 311 Suppl 1:S3-11. [PMID: 22206764 DOI: 10.1016/s0022-510x(11)70002-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Current concepts attribute an early and central role for auto-aggressive, myelin-specific T-lymphocytes in the pathogenesis of multiple sclerosis. This view emerged from immunological and pathological findings in experimental autoimmune encephalitis, an animal model characterised by pathological lesions closely resembling the ones found in multiple sclerosis. Furthermore, therapeutic strategies targeting the functions of these encephalitogenic T cells which attenuate their pathogenicity such as glatiramer acetate or anti-VLA4 antibody treatments represent proven approaches in multiple sclerosis. Nonetheless, all therapies evaluated to date either insufficiently dampen down inflammation or completely block immune processes. For this reason, there is a need to identify new therapeutic targets. We have employed live intravital two-photon microscopy to learn more about the behaviour of T cells during the preclinical phase of EAE, when T cells acquire the properties required to invade their target organ. Furthermore, we were able to identify an unexpected locomotive behaviour of T cells at the blood-brain barrier, which occurs immediately before diapedesis and the induction of paralytic disease. Such studies might open new avenues for the treatment of CNS autoimmune diseases. Multiple sclerosis is considered to be an autoimmune disease in which self-reactive T cells enter the central nervous system (CNS) and create an inflammatory milieu that destroys myelin and neurons. Immunomodulatory strategies for the treatment of multiple sclerosis target this process by attempting to inactivate these auto-aggressive T cells. However, so far, these strategies have failed to extinguish disease activity completely. For this reason, there is a need to understand in more detail the mechanisms by which T cells become encephalitogenic, how they enter the nervous system, and what the signals are that guide them along this path. If these processes could be better understood, it may be possible to design more effective and specific therapies for multiple sclerosis. This article will give a brief overview about our recent findings obtained using intravital imaging of autoaggressive effector T cells in an experimental model of multiple sclerosis. This new technological approach might help to fill some gaps in the understanding of autoimmune pathogenesis of multiple sclerosis.
Collapse
Affiliation(s)
- Alexander Flügel
- Institute for Multiple Sclerosis Research, Department of Neuroimmunology, Gemeinnützige Hertie-Stiftung and University Medical Centre Göttingen, Göttingen, Germany
| | | | | | | |
Collapse
|
40
|
Meyers L, Groover CJ, Douglas J, Lee S, Brand D, Levin MC, Gardner LA. A role for Apolipoprotein A-I in the pathogenesis of multiple sclerosis. J Neuroimmunol 2014; 277:176-85. [PMID: 25468275 DOI: 10.1016/j.jneuroim.2014.10.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 10/20/2014] [Accepted: 10/24/2014] [Indexed: 01/13/2023]
Abstract
Apolipoprotein A1 (Apo A-I), the most abundant component of high-density lipoprotein (HDL), is an anti-inflammatory molecule, yet its potential role in the pathogenesis of multiple sclerosis (MS) has not been fully investigated. In this study, Western blot analyses of human plasma showed differential Apo A-I expression in healthy controls compared to MS patients. Further, primary progressive MS patients had less plasma Apo A-I than other forms of MS. Using experimental allergic encephalomyelitis (EAE) as a model for MS, Apo A-I deficient mice exhibited worse clinical disease and more neurodegeneration concurrent with increased levels of pro-inflammatory cytokines compared to wild-type animals. These data suggest that Apo A-I plays a role in the pathogenesis of EAE, a model for MS, creating the possibility for agents that increase Apo A-I levels as potential therapies for MS.
Collapse
Affiliation(s)
| | | | | | - Sangmin Lee
- Research Service VAMC, Memphis, TN 38104, United States; Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - David Brand
- Research Service VAMC, Memphis, TN 38104, United States
| | - Michael C Levin
- Research Service VAMC, Memphis, TN 38104, United States; Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Lidia A Gardner
- Research Service VAMC, Memphis, TN 38104, United States; Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, United States.
| |
Collapse
|
41
|
Single β³-amino acid substitutions to MOG peptides suppress the development of experimental autoimmune encephalomyelitis. J Neuroimmunol 2014; 277:67-76. [PMID: 25454728 DOI: 10.1016/j.jneuroim.2014.09.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 09/17/2014] [Accepted: 09/30/2014] [Indexed: 12/12/2022]
Abstract
CD4(+) T-cells play a key role in the pathogenesis of multiple sclerosis (MS). Altered peptide ligands capable of modulating T-cell autoreactivity are considered a promising strategy for development of antigen-specific therapies for MS. Since peptides are inherently unstable, the current study explored single β-amino acid substitution as a means of stabilizing an epitope of myelin oligodendrocyte glycoprotein. β-Amino acid substitution at position 44, the major T-cell receptor contact residue, increased the half-life of active metabolites. Vaccination with one altered peptide, MOG44βF, conferred protection from EAE, decreased T-cell autoreactivity and pro-inflammatory cytokine production. Additional studies using MOG44βF in an oral treatment regimen, administered after EAE induction, also attenuated disease severity. Thus, altered peptides such as those reported here may lead to the development of novel and more specific treatments for MS.
Collapse
|
42
|
Shetty A, Gupta SG, Varrin-Doyer M, Weber MS, Prod'homme T, Molnarfi N, Ji N, Nelson PA, Patarroyo JC, Schulze-Topphoff U, Fogal SE, Forsthuber T, Sobel RA, Bernard CCA, Slavin AJ, Zamvil SS. Immunodominant T-cell epitopes of MOG reside in its transmembrane and cytoplasmic domains in EAE. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2014; 1:e22. [PMID: 25340074 PMCID: PMC4202928 DOI: 10.1212/nxi.0000000000000022] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/26/2014] [Indexed: 01/15/2023]
Abstract
Objective: Studies evaluating T-cell recognition of myelin oligodendrocyte glycoprotein (MOG) in multiple sclerosis (MS) and its model, experimental autoimmune encephalomyelitis (EAE), have focused mostly on its 117 amino acid (aa) extracellular domain, especially peptide (p) 35-55. We characterized T-cell responses to the entire 218 aa MOG sequence, including its transmembrane and cytoplasmic domains. Methods: T-cell recognition in mice was examined using overlapping peptides and intact full-length mouse MOG. EAE was evaluated by peptide immunization and by adoptive transfer of MOG epitope-specific T cells. Frequency of epitope-specific T cells was examined by ELISPOT. Results: Three T-cell determinants of MOG were discovered in its transmembrane and cytoplasmic domains, p119–132, p181–195, and p186–200. Transmembrane MOG p119-132 induced clinical EAE, CNS inflammation, and demyelination as potently as p35-55 in C57BL/6 mice and other H-2b strains. p119-128 contained its minimal encephalitogenic epitope. p119-132 did not cause disease in EAE-susceptible non-H-2b strains, including Biozzi, NOD, and PL/J. MOG p119-132–specific T cells produced Th1 and Th17 cytokines and transferred EAE to wild-type recipient mice. After immunization with full-length MOG, a significantly higher frequency of MOG-reactive T cells responded to p119-132 than to p35-55, demonstrating that p119-132 is an immunodominant encephalitogenic epitope. MOG p181-195 did not cause EAE, and MOG p181-195–specific T cells could not transfer EAE into wild-type or highly susceptible T- and B-cell–deficient mice. Conclusions: Transmembrane and cytoplasmic domains of MOG contain immunodominant T-cell epitopes in EAE. A CNS autoantigen can also contain nonpathogenic stimulatory T-cell epitopes. Recognition that a myelin antigen contains multiple encephalitogenic and nonencephalitogenic determinants may have implications for therapeutic development in MS.
Collapse
Affiliation(s)
- Aparna Shetty
- Department of Neurology and Program in Immunology (A.S., S.G.G., M.V.-D., T.P., N.M., P.A.N., J.C.P., U.S.-T., S.S.Z.), University of California, San Francisco; Department of Neuropathology and Department of Neurology (M.S.W.), University Medical Center, Georg-August University, Göttingen, Germany; Department of Immunology (N.J., T.F.), University of Texas at San Antonio; Boehringer Ingelheim (S.E.F., A.J.S.), Ridgefield, CT; Department of Pathology (R.A.S.), Stanford University, Stanford, CA; and Multiple Sclerosis Research Group (C.C.A.B.), Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia. S.G.G. is currently at the Institute for Immunity Transplantation and Infection, Stanford University, Stanford, CA. T.P. is currently at Momenta Pharmaceuticals, Cambridge, MA. N.M. is currently at the Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospital and the Department of Pathology and Immunology, Geneva Faculty of Medicine, University Medical Center, Geneva, Switzerland. J.C.P. is currently at Pfizer, Inc., Cambridge, MA
| | - Sheena G Gupta
- Department of Neurology and Program in Immunology (A.S., S.G.G., M.V.-D., T.P., N.M., P.A.N., J.C.P., U.S.-T., S.S.Z.), University of California, San Francisco; Department of Neuropathology and Department of Neurology (M.S.W.), University Medical Center, Georg-August University, Göttingen, Germany; Department of Immunology (N.J., T.F.), University of Texas at San Antonio; Boehringer Ingelheim (S.E.F., A.J.S.), Ridgefield, CT; Department of Pathology (R.A.S.), Stanford University, Stanford, CA; and Multiple Sclerosis Research Group (C.C.A.B.), Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia. S.G.G. is currently at the Institute for Immunity Transplantation and Infection, Stanford University, Stanford, CA. T.P. is currently at Momenta Pharmaceuticals, Cambridge, MA. N.M. is currently at the Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospital and the Department of Pathology and Immunology, Geneva Faculty of Medicine, University Medical Center, Geneva, Switzerland. J.C.P. is currently at Pfizer, Inc., Cambridge, MA
| | - Michel Varrin-Doyer
- Department of Neurology and Program in Immunology (A.S., S.G.G., M.V.-D., T.P., N.M., P.A.N., J.C.P., U.S.-T., S.S.Z.), University of California, San Francisco; Department of Neuropathology and Department of Neurology (M.S.W.), University Medical Center, Georg-August University, Göttingen, Germany; Department of Immunology (N.J., T.F.), University of Texas at San Antonio; Boehringer Ingelheim (S.E.F., A.J.S.), Ridgefield, CT; Department of Pathology (R.A.S.), Stanford University, Stanford, CA; and Multiple Sclerosis Research Group (C.C.A.B.), Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia. S.G.G. is currently at the Institute for Immunity Transplantation and Infection, Stanford University, Stanford, CA. T.P. is currently at Momenta Pharmaceuticals, Cambridge, MA. N.M. is currently at the Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospital and the Department of Pathology and Immunology, Geneva Faculty of Medicine, University Medical Center, Geneva, Switzerland. J.C.P. is currently at Pfizer, Inc., Cambridge, MA
| | - Martin S Weber
- Department of Neurology and Program in Immunology (A.S., S.G.G., M.V.-D., T.P., N.M., P.A.N., J.C.P., U.S.-T., S.S.Z.), University of California, San Francisco; Department of Neuropathology and Department of Neurology (M.S.W.), University Medical Center, Georg-August University, Göttingen, Germany; Department of Immunology (N.J., T.F.), University of Texas at San Antonio; Boehringer Ingelheim (S.E.F., A.J.S.), Ridgefield, CT; Department of Pathology (R.A.S.), Stanford University, Stanford, CA; and Multiple Sclerosis Research Group (C.C.A.B.), Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia. S.G.G. is currently at the Institute for Immunity Transplantation and Infection, Stanford University, Stanford, CA. T.P. is currently at Momenta Pharmaceuticals, Cambridge, MA. N.M. is currently at the Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospital and the Department of Pathology and Immunology, Geneva Faculty of Medicine, University Medical Center, Geneva, Switzerland. J.C.P. is currently at Pfizer, Inc., Cambridge, MA
| | - Thomas Prod'homme
- Department of Neurology and Program in Immunology (A.S., S.G.G., M.V.-D., T.P., N.M., P.A.N., J.C.P., U.S.-T., S.S.Z.), University of California, San Francisco; Department of Neuropathology and Department of Neurology (M.S.W.), University Medical Center, Georg-August University, Göttingen, Germany; Department of Immunology (N.J., T.F.), University of Texas at San Antonio; Boehringer Ingelheim (S.E.F., A.J.S.), Ridgefield, CT; Department of Pathology (R.A.S.), Stanford University, Stanford, CA; and Multiple Sclerosis Research Group (C.C.A.B.), Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia. S.G.G. is currently at the Institute for Immunity Transplantation and Infection, Stanford University, Stanford, CA. T.P. is currently at Momenta Pharmaceuticals, Cambridge, MA. N.M. is currently at the Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospital and the Department of Pathology and Immunology, Geneva Faculty of Medicine, University Medical Center, Geneva, Switzerland. J.C.P. is currently at Pfizer, Inc., Cambridge, MA
| | - Nicolas Molnarfi
- Department of Neurology and Program in Immunology (A.S., S.G.G., M.V.-D., T.P., N.M., P.A.N., J.C.P., U.S.-T., S.S.Z.), University of California, San Francisco; Department of Neuropathology and Department of Neurology (M.S.W.), University Medical Center, Georg-August University, Göttingen, Germany; Department of Immunology (N.J., T.F.), University of Texas at San Antonio; Boehringer Ingelheim (S.E.F., A.J.S.), Ridgefield, CT; Department of Pathology (R.A.S.), Stanford University, Stanford, CA; and Multiple Sclerosis Research Group (C.C.A.B.), Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia. S.G.G. is currently at the Institute for Immunity Transplantation and Infection, Stanford University, Stanford, CA. T.P. is currently at Momenta Pharmaceuticals, Cambridge, MA. N.M. is currently at the Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospital and the Department of Pathology and Immunology, Geneva Faculty of Medicine, University Medical Center, Geneva, Switzerland. J.C.P. is currently at Pfizer, Inc., Cambridge, MA
| | - Niannian Ji
- Department of Neurology and Program in Immunology (A.S., S.G.G., M.V.-D., T.P., N.M., P.A.N., J.C.P., U.S.-T., S.S.Z.), University of California, San Francisco; Department of Neuropathology and Department of Neurology (M.S.W.), University Medical Center, Georg-August University, Göttingen, Germany; Department of Immunology (N.J., T.F.), University of Texas at San Antonio; Boehringer Ingelheim (S.E.F., A.J.S.), Ridgefield, CT; Department of Pathology (R.A.S.), Stanford University, Stanford, CA; and Multiple Sclerosis Research Group (C.C.A.B.), Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia. S.G.G. is currently at the Institute for Immunity Transplantation and Infection, Stanford University, Stanford, CA. T.P. is currently at Momenta Pharmaceuticals, Cambridge, MA. N.M. is currently at the Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospital and the Department of Pathology and Immunology, Geneva Faculty of Medicine, University Medical Center, Geneva, Switzerland. J.C.P. is currently at Pfizer, Inc., Cambridge, MA
| | - Patricia A Nelson
- Department of Neurology and Program in Immunology (A.S., S.G.G., M.V.-D., T.P., N.M., P.A.N., J.C.P., U.S.-T., S.S.Z.), University of California, San Francisco; Department of Neuropathology and Department of Neurology (M.S.W.), University Medical Center, Georg-August University, Göttingen, Germany; Department of Immunology (N.J., T.F.), University of Texas at San Antonio; Boehringer Ingelheim (S.E.F., A.J.S.), Ridgefield, CT; Department of Pathology (R.A.S.), Stanford University, Stanford, CA; and Multiple Sclerosis Research Group (C.C.A.B.), Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia. S.G.G. is currently at the Institute for Immunity Transplantation and Infection, Stanford University, Stanford, CA. T.P. is currently at Momenta Pharmaceuticals, Cambridge, MA. N.M. is currently at the Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospital and the Department of Pathology and Immunology, Geneva Faculty of Medicine, University Medical Center, Geneva, Switzerland. J.C.P. is currently at Pfizer, Inc., Cambridge, MA
| | - Juan C Patarroyo
- Department of Neurology and Program in Immunology (A.S., S.G.G., M.V.-D., T.P., N.M., P.A.N., J.C.P., U.S.-T., S.S.Z.), University of California, San Francisco; Department of Neuropathology and Department of Neurology (M.S.W.), University Medical Center, Georg-August University, Göttingen, Germany; Department of Immunology (N.J., T.F.), University of Texas at San Antonio; Boehringer Ingelheim (S.E.F., A.J.S.), Ridgefield, CT; Department of Pathology (R.A.S.), Stanford University, Stanford, CA; and Multiple Sclerosis Research Group (C.C.A.B.), Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia. S.G.G. is currently at the Institute for Immunity Transplantation and Infection, Stanford University, Stanford, CA. T.P. is currently at Momenta Pharmaceuticals, Cambridge, MA. N.M. is currently at the Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospital and the Department of Pathology and Immunology, Geneva Faculty of Medicine, University Medical Center, Geneva, Switzerland. J.C.P. is currently at Pfizer, Inc., Cambridge, MA
| | - Ulf Schulze-Topphoff
- Department of Neurology and Program in Immunology (A.S., S.G.G., M.V.-D., T.P., N.M., P.A.N., J.C.P., U.S.-T., S.S.Z.), University of California, San Francisco; Department of Neuropathology and Department of Neurology (M.S.W.), University Medical Center, Georg-August University, Göttingen, Germany; Department of Immunology (N.J., T.F.), University of Texas at San Antonio; Boehringer Ingelheim (S.E.F., A.J.S.), Ridgefield, CT; Department of Pathology (R.A.S.), Stanford University, Stanford, CA; and Multiple Sclerosis Research Group (C.C.A.B.), Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia. S.G.G. is currently at the Institute for Immunity Transplantation and Infection, Stanford University, Stanford, CA. T.P. is currently at Momenta Pharmaceuticals, Cambridge, MA. N.M. is currently at the Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospital and the Department of Pathology and Immunology, Geneva Faculty of Medicine, University Medical Center, Geneva, Switzerland. J.C.P. is currently at Pfizer, Inc., Cambridge, MA
| | - Stephen E Fogal
- Department of Neurology and Program in Immunology (A.S., S.G.G., M.V.-D., T.P., N.M., P.A.N., J.C.P., U.S.-T., S.S.Z.), University of California, San Francisco; Department of Neuropathology and Department of Neurology (M.S.W.), University Medical Center, Georg-August University, Göttingen, Germany; Department of Immunology (N.J., T.F.), University of Texas at San Antonio; Boehringer Ingelheim (S.E.F., A.J.S.), Ridgefield, CT; Department of Pathology (R.A.S.), Stanford University, Stanford, CA; and Multiple Sclerosis Research Group (C.C.A.B.), Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia. S.G.G. is currently at the Institute for Immunity Transplantation and Infection, Stanford University, Stanford, CA. T.P. is currently at Momenta Pharmaceuticals, Cambridge, MA. N.M. is currently at the Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospital and the Department of Pathology and Immunology, Geneva Faculty of Medicine, University Medical Center, Geneva, Switzerland. J.C.P. is currently at Pfizer, Inc., Cambridge, MA
| | - Thomas Forsthuber
- Department of Neurology and Program in Immunology (A.S., S.G.G., M.V.-D., T.P., N.M., P.A.N., J.C.P., U.S.-T., S.S.Z.), University of California, San Francisco; Department of Neuropathology and Department of Neurology (M.S.W.), University Medical Center, Georg-August University, Göttingen, Germany; Department of Immunology (N.J., T.F.), University of Texas at San Antonio; Boehringer Ingelheim (S.E.F., A.J.S.), Ridgefield, CT; Department of Pathology (R.A.S.), Stanford University, Stanford, CA; and Multiple Sclerosis Research Group (C.C.A.B.), Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia. S.G.G. is currently at the Institute for Immunity Transplantation and Infection, Stanford University, Stanford, CA. T.P. is currently at Momenta Pharmaceuticals, Cambridge, MA. N.M. is currently at the Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospital and the Department of Pathology and Immunology, Geneva Faculty of Medicine, University Medical Center, Geneva, Switzerland. J.C.P. is currently at Pfizer, Inc., Cambridge, MA
| | - Raymond A Sobel
- Department of Neurology and Program in Immunology (A.S., S.G.G., M.V.-D., T.P., N.M., P.A.N., J.C.P., U.S.-T., S.S.Z.), University of California, San Francisco; Department of Neuropathology and Department of Neurology (M.S.W.), University Medical Center, Georg-August University, Göttingen, Germany; Department of Immunology (N.J., T.F.), University of Texas at San Antonio; Boehringer Ingelheim (S.E.F., A.J.S.), Ridgefield, CT; Department of Pathology (R.A.S.), Stanford University, Stanford, CA; and Multiple Sclerosis Research Group (C.C.A.B.), Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia. S.G.G. is currently at the Institute for Immunity Transplantation and Infection, Stanford University, Stanford, CA. T.P. is currently at Momenta Pharmaceuticals, Cambridge, MA. N.M. is currently at the Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospital and the Department of Pathology and Immunology, Geneva Faculty of Medicine, University Medical Center, Geneva, Switzerland. J.C.P. is currently at Pfizer, Inc., Cambridge, MA
| | - Claude C A Bernard
- Department of Neurology and Program in Immunology (A.S., S.G.G., M.V.-D., T.P., N.M., P.A.N., J.C.P., U.S.-T., S.S.Z.), University of California, San Francisco; Department of Neuropathology and Department of Neurology (M.S.W.), University Medical Center, Georg-August University, Göttingen, Germany; Department of Immunology (N.J., T.F.), University of Texas at San Antonio; Boehringer Ingelheim (S.E.F., A.J.S.), Ridgefield, CT; Department of Pathology (R.A.S.), Stanford University, Stanford, CA; and Multiple Sclerosis Research Group (C.C.A.B.), Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia. S.G.G. is currently at the Institute for Immunity Transplantation and Infection, Stanford University, Stanford, CA. T.P. is currently at Momenta Pharmaceuticals, Cambridge, MA. N.M. is currently at the Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospital and the Department of Pathology and Immunology, Geneva Faculty of Medicine, University Medical Center, Geneva, Switzerland. J.C.P. is currently at Pfizer, Inc., Cambridge, MA
| | - Anthony J Slavin
- Department of Neurology and Program in Immunology (A.S., S.G.G., M.V.-D., T.P., N.M., P.A.N., J.C.P., U.S.-T., S.S.Z.), University of California, San Francisco; Department of Neuropathology and Department of Neurology (M.S.W.), University Medical Center, Georg-August University, Göttingen, Germany; Department of Immunology (N.J., T.F.), University of Texas at San Antonio; Boehringer Ingelheim (S.E.F., A.J.S.), Ridgefield, CT; Department of Pathology (R.A.S.), Stanford University, Stanford, CA; and Multiple Sclerosis Research Group (C.C.A.B.), Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia. S.G.G. is currently at the Institute for Immunity Transplantation and Infection, Stanford University, Stanford, CA. T.P. is currently at Momenta Pharmaceuticals, Cambridge, MA. N.M. is currently at the Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospital and the Department of Pathology and Immunology, Geneva Faculty of Medicine, University Medical Center, Geneva, Switzerland. J.C.P. is currently at Pfizer, Inc., Cambridge, MA
| | - Scott S Zamvil
- Department of Neurology and Program in Immunology (A.S., S.G.G., M.V.-D., T.P., N.M., P.A.N., J.C.P., U.S.-T., S.S.Z.), University of California, San Francisco; Department of Neuropathology and Department of Neurology (M.S.W.), University Medical Center, Georg-August University, Göttingen, Germany; Department of Immunology (N.J., T.F.), University of Texas at San Antonio; Boehringer Ingelheim (S.E.F., A.J.S.), Ridgefield, CT; Department of Pathology (R.A.S.), Stanford University, Stanford, CA; and Multiple Sclerosis Research Group (C.C.A.B.), Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia. S.G.G. is currently at the Institute for Immunity Transplantation and Infection, Stanford University, Stanford, CA. T.P. is currently at Momenta Pharmaceuticals, Cambridge, MA. N.M. is currently at the Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospital and the Department of Pathology and Immunology, Geneva Faculty of Medicine, University Medical Center, Geneva, Switzerland. J.C.P. is currently at Pfizer, Inc., Cambridge, MA
| |
Collapse
|
43
|
Cytokine production profiles in chronic relapsing-remitting experimental autoimmune encephalomyelitis: IFN-γ and TNF-α are important participants in the first attack but not in the relapse. J Neurol Sci 2014; 340:117-22. [PMID: 24655735 DOI: 10.1016/j.jns.2014.02.039] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2013] [Revised: 01/27/2014] [Accepted: 02/24/2014] [Indexed: 11/20/2022]
Abstract
Multiple sclerosis (MS) is a chronic demyelinating disease often displaying a relapsing-remitting course of neurological manifestations that is mimicked by experimental autoimmune encephalomyelitis (EAE) in animal models of MS. In particular, NOD mice immunized with myelin oligodendrocyte glycoprotein peptide 35-55 develop chronic relapsing-remitting EAE (CREAE). To elucidate the mechanisms that cause MS relapse, we investigated the histopathology and cytokine production of spleen cells and mRNA expression levels in the central nervous system (CNS) of CREAE mice. During the first attack, inflammatory cell infiltration around small vessels and in the subarachnoid space was observed in the spinal cord. Spleen cell production and mRNA expression in the CNS of several cytokines, including IFN-γ, TNF-α, IL-6, IL-17, and CC chemokine ligand 2 (CCL2), were higher in CREAE mice than in controls. Afterwards, parenchymal infiltration and demyelination were observed histologically in the spinal cord and corresponded with the more severe clinical symptoms of the first and second relapses. IL-17 and CCL2, but not IFN-γ, TNF-α, or IL-6, were also produced by spleen cells during recurrences. Our results suggested that the immune mechanisms in relapses were different from those in the first attack for CREAE. Further investigation of CREAE mechanisms may provide important insights into successful therapies for human relapsing-remitting MS.
Collapse
|
44
|
Dagley LF, Emili A, Purcell AW. Application of quantitative proteomics technologies to the biomarker discovery pipeline for multiple sclerosis. Proteomics Clin Appl 2014; 7:91-108. [PMID: 23112123 DOI: 10.1002/prca.201200104] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 10/04/2012] [Accepted: 10/11/2012] [Indexed: 11/08/2022]
Abstract
Multiple sclerosis is an inflammatory-mediated demyelinating disorder most prevalent in young Caucasian adults. The various clinical manifestations of the disease present several challenges in the clinic in terms of diagnosis, monitoring disease progression and response to treatment. Advances in MS-based proteomic technologies have revolutionized the field of biomarker research and paved the way for the identification and validation of disease-specific markers. This review focuses on the novel candidates discovered by the application of quantitative proteomics to relevant disease-affected tissues in both the human context and within the animal model of the disease known as experimental autoimmune encephalomyelitis. The role of targeted MS approaches for biomarker validation studies, such as multiple reaction monitoring will also be discussed.
Collapse
Affiliation(s)
- Laura F Dagley
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | | | | |
Collapse
|
45
|
Wang CK, Gruber CW, Cemazar M, Siatskas C, Tagore P, Payne N, Sun G, Wang S, Bernard CC, Craik DJ. Molecular grafting onto a stable framework yields novel cyclic peptides for the treatment of multiple sclerosis. ACS Chem Biol 2014; 9:156-63. [PMID: 24147816 PMCID: PMC3898541 DOI: 10.1021/cb400548s] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
![]()
Multiple sclerosis (MS) is an inflammatory
disease of the central
nervous system (CNS) and is characterized by the destruction of myelin
and axons leading to progressive disability. Peptide epitopes from
CNS proteins, such as myelin oligodendrocyte glycoprotein (MOG), possess
promising immunoregulatory potential for treating MS; however, their
instability and poor bioavailability is a major impediment for their
use clinically. To overcome this problem, we used molecular grafting
to incorporate peptide sequences from the MOG35–55 epitope onto a cyclotide, which is a macrocyclic peptide scaffold
that has been shown to be intrinsically stable. Using this approach,
we designed novel cyclic peptides that retained the structure and
stability of the parent scaffold. One of the grafted peptides, MOG3,
displayed potent ability to prevent disease development in a mouse
model of MS. These results demonstrate the potential of bioengineered
cyclic peptides for the treatment of MS.
Collapse
Affiliation(s)
- Conan K. Wang
- Institute
for Molecular Bioscience, University of Queensland, Brisbane, QLD 4072, Australia
| | - Christian W. Gruber
- Institute
for Molecular Bioscience, University of Queensland, Brisbane, QLD 4072, Australia
- Center
for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, Vienna 1090, Austria
| | - Maša Cemazar
- Institute
for Molecular Bioscience, University of Queensland, Brisbane, QLD 4072, Australia
| | - Christopher Siatskas
- Multiple
Sclerosis Research Group, Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Prascilla Tagore
- Institute
for Molecular Bioscience, University of Queensland, Brisbane, QLD 4072, Australia
| | - Natalie Payne
- Multiple
Sclerosis Research Group, Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Guizhi Sun
- Multiple
Sclerosis Research Group, Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Shunhe Wang
- Multiple
Sclerosis Research Group, Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Claude C. Bernard
- Multiple
Sclerosis Research Group, Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - David J. Craik
- Institute
for Molecular Bioscience, University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
46
|
Cross-reactivity between human cytomegalovirus peptide 981-1003 and myelin oligodendroglia glycoprotein peptide 35-55 in experimental autoimmune encephalomyelitis in Lewis rats. Biochem Biophys Res Commun 2014; 443:1118-23. [PMID: 24388990 DOI: 10.1016/j.bbrc.2013.12.122] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 12/23/2013] [Indexed: 11/21/2022]
Abstract
Multiple sclerosis (MS) has been documented to have various clinical and pathological presentations. However the underlying mechanisms remain unknown. Viral infections may play a certain role in the etiopathogenesis of MS. This study was designed to explore whether different phospholipid peptides and viral mimic peptides induce antigen specific lesion in experimental autoimmune encephalomyelitis (EAE), an MS animal model. In the present study, Lewis rats immunized with myelin basic protein (MBP) 82-99 or MBP68-86 exhibited clinical signs of EAE and inflammatory infiltrates throughout CNS. Immunization with myelin oligodendroglia glycoprotein (MOG) 35-55 also induced inflammatory infiltrates in spinal cords. Although cytomegalovirus (CMV) 981-1003 failed to induce clinical signs of EAE and inflammatory infiltrates, immunological examination revealed that CMV981-1003 cross-reacted with serum from rats immunized with MOG35-55, and vice versa. Further, MOG35-55 triggered CMV981-1003 specific lymphocytes recruitment in spleen. Together these, this study provides certain evidences for various pathological manifestations of EAE and the linkage of viral mimic peptides with phospholipid peptides. Molecular mimicry may be an explanation the pathogenesis of MS.
Collapse
|
47
|
Litwak SA, Payne NL, Campanale N, Ozturk E, Lee JY, Petratos S, Siatskas C, Bakhuraysah M, Bernard CCA. Nogo-receptor 1 deficiency has no influence on immune cell repertoire or function during experimental autoimmune encephalomyelitis. PLoS One 2013; 8:e82101. [PMID: 24339996 PMCID: PMC3855334 DOI: 10.1371/journal.pone.0082101] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2013] [Accepted: 10/30/2013] [Indexed: 12/03/2022] Open
Abstract
The potential role of Nogo-66 Receptor 1 (NgR1) on immune cell phenotypes and their activation during neuroinflammatory diseases such as multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE), is unclear. To further understand the function of this receptor on haematopoietically-derived cells, phenotypic and functional analyses were performed using NgR1-deficient (ngr1-/-) animals. Flow cytometry-based phenotypic analyses performed on blood, spleen, thymus, lymph nodes, bone marrow and central nervous-system (CNS)-infiltrating blood cells revealed no immunological defects in naïve ngr1-/- animals versus wild-type littermate (WTLM) controls. EAE was induced by either recombinant myelin oligodendrocyte glycoprotein (rMOG), a model in which B cells are considered to contribute pathogenically, or by MOG35–55 peptide, a B cell-independent model. We have demonstrated that in ngr1-/- mice injected with MOG35–55, a significant reduction in the severity of EAE correlated with reduced axonal damage present in the spinal cord when compared to their WTLM controls. However, despite a reduction in axonal damage observed in the CNS of ngr1-/- mice at the chronic stage of disease, no clinical differences could be attributed to a specific genotype when rMOG was used as the encephalitogen. Following MOG35–55-induction of EAE, we could not derive any major changes to the immune cell populations analyzed between ngr1-/- and WTLM mice. Collectively, these data demonstrate that NgR1 has little if any effects on the repertoire of immune cells, their activation and trafficking to the CNS.
Collapse
Affiliation(s)
- Sara A. Litwak
- Multiple Sclerosis Research Group, Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Natalie L. Payne
- Multiple Sclerosis Research Group, Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Naomi Campanale
- Multiple Sclerosis Research Group, Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Ezgi Ozturk
- Multiple Sclerosis Research Group, Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Jae Young Lee
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Steven Petratos
- Central Clinical School, Monash University, Prahran, Victoria, Australia
| | - Christopher Siatskas
- Multiple Sclerosis Research Group, Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Maha Bakhuraysah
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Claude C. A. Bernard
- Multiple Sclerosis Research Group, Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
- * E-mail:
| |
Collapse
|
48
|
Lalive PH, Benkhoucha M, Tran NL, Kreutzfeldt M, Merkler D, Santiago-Raber ML. TLR7 signaling exacerbates CNS autoimmunity through downregulation of Foxp3+ Treg cells. Eur J Immunol 2013; 44:46-57. [PMID: 24018482 DOI: 10.1002/eji.201242985] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 08/02/2013] [Accepted: 09/06/2013] [Indexed: 12/21/2022]
Abstract
The innate Toll-like receptor 7 (TLR7) detects infections by recognizing viral and bacterial single-stranded RNA. In addition to pathogen-derived RNA, immune cells expressing high levels of TLR7, such as B cells and dendritic cells (DCs), can be activated by self-RNA. During myelin-induced experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis, TLR7 expression is increased within the central nervous system (CNS). To define the contribution of TLR7 to the development of EAE, we evaluated the course of the disease in C57BL/6-Tlr7-deficient mice compared with that in WT mice and found that TLR7-deficient mice had decreased disease severity. This protection was associated with decreased myelin oligodendrocyte glycoprotein-specific T-cell activation by primed DCs, decreased circulating autoantibodies, attenuated inflammation within the CNS, and increased Foxp3(+) regulatory T cells in the periphery and in the CNS. In conclusion, we show that TLR7 is involved in the maintenance of autoimmunity in the pathogenesis of EAE.
Collapse
Affiliation(s)
- Patrice H Lalive
- Faculty of Medicine, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland; Division of Neurology, Department of Clinical Neurosciences, Neuroimmunology Laboratory, Geneva University Hospital, Geneva, Switzerland; Division of Laboratory Medicine, Department of Genetic and Laboratory Medicine, Geneva University Hospital, Geneva, Switzerland
| | | | | | | | | | | |
Collapse
|
49
|
Recks MS, Stormanns ER, Bader J, Arnhold S, Addicks K, Kuerten S. Early axonal damage and progressive myelin pathology define the kinetics of CNS histopathology in a mouse model of multiple sclerosis. Clin Immunol 2013; 149:32-45. [DOI: 10.1016/j.clim.2013.06.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Revised: 06/04/2013] [Accepted: 06/10/2013] [Indexed: 12/14/2022]
|
50
|
Subclinical CNS Inflammation as Response to a Myelin Antigen in Humanized Mice. J Neuroimmune Pharmacol 2013; 8:1037-47. [DOI: 10.1007/s11481-013-9466-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 04/22/2013] [Indexed: 12/25/2022]
|