1
|
Perdaens O, van Pesch V. Molecular Mechanisms of Immunosenescene and Inflammaging: Relevance to the Immunopathogenesis and Treatment of Multiple Sclerosis. Front Neurol 2022; 12:811518. [PMID: 35281989 PMCID: PMC8913495 DOI: 10.3389/fneur.2021.811518] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/27/2021] [Indexed: 12/18/2022] Open
Abstract
Aging is characterized, amongst other features, by a complex process of cellular senescence involving both innate and adaptive immunity, called immunosenescence and associated to inflammaging, a low-grade chronic inflammation. Both processes fuel each other and partially explain increasing incidence of cancers, infections, age-related autoimmunity, and vascular disease as well as a reduced response to vaccination. Multiple sclerosis (MS) is a lifelong disease, for which considerable progress in disease-modifying therapies (DMTs) and management has improved long-term survival. However, disability progression, increasing with age and disease duration, remains. Neurologists are now involved in caring for elderly MS patients, with increasing comorbidities. Aging of the immune system therefore has relevant implications for MS pathogenesis, response to DMTs and the risks mediated by these treatments. We propose to review current evidence regarding markers and molecular mechanisms of immunosenescence and their relevance to understanding MS pathogenesis. We will focus on age-related changes in the innate and adaptive immune system in MS and other auto-immune diseases, such as systemic lupus erythematosus and rheumatoid arthritis. The consequences of these immune changes on MS pathology, in interaction with the intrinsic aging process of central nervous system resident cells will be discussed. Finally, the impact of immunosenescence on disease evolution and on the safety and efficacy of current DMTs will be presented.
Collapse
Affiliation(s)
- Océane Perdaens
- Laboratory of Neurochemistry, Institute of Neuroscience, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Vincent van Pesch
- Laboratory of Neurochemistry, Institute of Neuroscience, Université catholique de Louvain (UCLouvain), Brussels, Belgium
- Department of Neurology, Cliniques universitaires Saint-Luc, Université catholique de Louvain (UCLouvain), Brussels, Belgium
- *Correspondence: Vincent van Pesch
| |
Collapse
|
2
|
Jacobs SAH, Muraro PA, Cencioni MT, Knowles S, Cole JH, Nicholas R. Worse Physical Disability Is Associated With the Expression of PD-1 on Inflammatory T-Cells in Multiple Sclerosis Patients With Older Appearing Brains. Front Neurol 2022; 12:801097. [PMID: 35069428 PMCID: PMC8770747 DOI: 10.3389/fneur.2021.801097] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 12/15/2021] [Indexed: 12/30/2022] Open
Abstract
Background: Magnetic Resonance Imaging (MRI) analysis method "brain-age" paradigm could offer an intuitive prognostic metric (brain-predicted age difference: brain-PAD) for disability in Multiple Sclerosis (MS), reflecting structural brain health adjusted for aging. Equally, cellular senescence has been reported in MS using T-cell biomarker CD8+CD57+. Objective: Here we explored links between MRI-derived brain-age and blood-derived cellular senescence. We examined the value of combining brain-PAD with CD8+CD57+(ILT2+PD-1+) T-cells when predicting disability score in MS and considered whether age-related biological mechanisms drive disability. Methods: Brain-age analysis was applied to T1-weighted MRI images. Disability was assessed and peripheral blood was examined for CD8+CD57+ T-cell phenotypes. Linear regression models were used, adjusted for sex, age and normalized brain volume. Results: We included 179 mainly relapsing-remitting MS patients. A high brain-PAD was associated with high physical disability (mean brain-PAD = +6.54 [5.12-7.95]). CD8+CD57+(ILT2+PD-1+) T-cell frequency was neither associated with disability nor with brain-PAD. Physical disability was predicted by the interaction between brain-PAD and CD8+CD57+ILT2+PD-1+ T-cell frequency (AR 2 = 0.196), yet without improvement compared to brain-PAD alone (AR 2 = 0.206; AICc = 1.8). Conclusion: Higher frequency of CD8+CD57+ILT2+PD-1+ T-cells in the peripheral blood in patients with an older appearing brain was associated with worse disability scores, suggesting a role of these cells in the development of disability in MS patients with poorer brain health.
Collapse
Affiliation(s)
- Sophie A. H. Jacobs
- Department of Computer Science, Centre for Medical Image Computing, University College London, London, United Kingdom
- Department of Neurology, Imperial College Healthcare, London, United Kingdom
| | - Paolo A. Muraro
- Department of Neurology, Imperial College Healthcare, London, United Kingdom
- Division of Clinical Neurology, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Maria T. Cencioni
- Division of Clinical Neurology, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Sarah Knowles
- Department of Neurology, Imperial College Healthcare, London, United Kingdom
| | - James H. Cole
- Department of Computer Science, Centre for Medical Image Computing, University College London, London, United Kingdom
| | - Richard Nicholas
- Department of Neurology, Imperial College Healthcare, London, United Kingdom
| |
Collapse
|
3
|
Hrastelj J, Andrews R, Loveless S, Morgan J, Bishop SM, Bray NJ, Williams NM, Robertson NP. CSF-resident CD4 + T-cells display a distinct gene expression profile with relevance to immune surveillance and multiple sclerosis. Brain Commun 2021; 3:fcab155. [PMID: 34761221 PMCID: PMC8574295 DOI: 10.1093/braincomms/fcab155] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 04/12/2021] [Accepted: 05/17/2021] [Indexed: 12/13/2022] Open
Abstract
The CNS has traditionally been considered an immune privileged site, but is now understood to have a system of immune surveillance, predominantly involving CD4+ T-cells. Identifying functional differences between CNS and blood CD4+ T-cells, therefore, have relevance to CNS immune surveillance as well as to neurological conditions, such as multiple sclerosis, in which CD4+ T-cells play a central role. Here, CD4+ T-cells were purified from CSF and blood from 21 patients with newly diagnosed treatment-naïve multiple sclerosis and 20 individuals with non-inflammatory disorders using fluorescence-activated cell sorting, and their transcriptomes were profiled by RNA sequencing. Paired comparisons between CD4+ T-cells from CSF and blood identified 5156 differentially expressed genes in controls and 4263 differentially expressed in multiple sclerosis patients at false discovery rate <5%. Differential expression analysis of CD4+ T-cells collected from the CSF highlighted genes involved in migration, activation, cholesterol biosynthesis and signalling, including those with known relevance to multiple sclerosis pathogenesis and treatment. Expression of markers of CD4+ T-cell subtypes suggested an increased proportion of Th1 and Th17 cells in CSF. Gene ontology terms significant only in multiple sclerosis were predominantly those involved in cellular proliferation. A two-way comparison of CSF versus blood CD4+ T-cells in multiple sclerosis compared with non-inflammatory disorder controls identified four significant genes at false discovery rate <5% (CYP51A1, LRRD1, YES1 and PASK), further implicating cholesterol biosynthesis and migration mechanisms. Analysis of CSF CD4+ T-cells in an extended cohort of multiple sclerosis cases (total N = 41) compared with non-inflammatory disorder controls (total N = 38) identified 140 differentially expressed genes at false discovery rate < 5%, many of which have known relevance to multiple sclerosis, including XBP1, BHLHE40, CD40LG, DPP4 and ITGB1. This study provides the largest transcriptomic analysis of purified cell subpopulations in CSF to date and has relevance for the understanding of CNS immune surveillance, as well as multiple sclerosis pathogenesis and treatment discovery.
Collapse
Affiliation(s)
- James Hrastelj
- Division of Psychological Medicine and Clinical
Neuroscience, Cardiff University, Cardiff CF14 4XW, UK
| | - Robert Andrews
- School of Medicine, Cardiff
University, Cardiff CF14 4XW, UK
| | - Samantha Loveless
- Division of Psychological Medicine and Clinical
Neuroscience, Cardiff University, Cardiff CF14 4XW, UK
| | - Joanne Morgan
- Division of Psychological Medicine and Clinical
Neuroscience, Cardiff University, Cardiff CF14 4XW, UK
| | - Stefan Mark Bishop
- European Cancer Stem Cell Research Institute,
Cardiff University, Cardiff CF24 4HQ, UK
| | - Nicholas J Bray
- Division of Psychological Medicine and Clinical
Neuroscience, Cardiff University, Cardiff CF14 4XW, UK
| | - Nigel M Williams
- Division of Psychological Medicine and Clinical
Neuroscience, Cardiff University, Cardiff CF14 4XW, UK
| | - Neil P Robertson
- Division of Psychological Medicine and Clinical
Neuroscience, Cardiff University, Cardiff CF14 4XW, UK
| |
Collapse
|
4
|
Amici SA, Osman W, Guerau-de-Arellano M. PRMT5 Promotes Cyclin E1 and Cell Cycle Progression in CD4 Th1 Cells and Correlates With EAE Severity. Front Immunol 2021; 12:695947. [PMID: 34168658 PMCID: PMC8217861 DOI: 10.3389/fimmu.2021.695947] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 05/21/2021] [Indexed: 12/11/2022] Open
Abstract
Multiple Sclerosis (MS) is a debilitating central nervous system disorder associated with inflammatory T cells. Activation and expansion of inflammatory T cells is thought to be behind MS relapses and influence disease severity. Protein arginine N-methyltransferase 5 (PRMT5) is a T cell activation-induced enzyme that symmetrically dimethylates proteins and promotes T cell proliferation. However, the mechanism behind PRMT5-mediated control of T cell proliferation and whether PRMT5 contributes to diseases severity is unclear. Here, we evaluated the role of PRMT5 on cyclin/cdk pairs and cell cycle progression, as well as PRMT5's link to disease severity in an animal model of relapsing-remitting MS. Treatment of T helper 1 (mTh1) cells with the selective PRMT5 inhibitor, HLCL65, arrested activation-induced T cell proliferation at the G1 stage of the cell cycle, suggesting PRMT5 promotes cell cycle progression in CD4+ T cells. The Cyclin E1/Cdk2 pair promoting G1/S progression was also decreased after PRMT5 inhibition, as was the phosphorylation of retinoblastoma. In the SJL mouse relapsing-remitting model of MS, the highest PRMT5 expression in central nervous system-infiltrating cells corresponded to peak and relapse timepoints. PRMT5 expression also positively correlated with increasing CD4 Th cell composition, disease severity and Cyclin E1 expression. These data indicate that PRMT5 promotes G1/S cell cycle progression and suggest that this effect influences disease severity and/or progression in the animal model of MS. Modulating PRMT5 levels may be useful for controlling T cell expansion in T cell-mediated diseases including MS.
Collapse
MESH Headings
- Animals
- Cell Cycle
- Cell Proliferation
- Cyclin E/metabolism
- Cyclin-Dependent Kinase 2/metabolism
- Disease Progression
- Encephalomyelitis, Autoimmune, Experimental/enzymology
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Genes, T-Cell Receptor
- Mice, Transgenic
- Multiple Sclerosis, Relapsing-Remitting/enzymology
- Multiple Sclerosis, Relapsing-Remitting/immunology
- Multiple Sclerosis, Relapsing-Remitting/pathology
- Oncogene Proteins/metabolism
- Phosphorylation
- Protein-Arginine N-Methyltransferases/metabolism
- Retinoblastoma Protein/metabolism
- Severity of Illness Index
- Signal Transduction
- Th1 Cells/enzymology
- Th1 Cells/immunology
- Th1 Cells/pathology
Collapse
Affiliation(s)
- Stephanie A. Amici
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Wissam Osman
- Discovery PREP Program, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Mireia Guerau-de-Arellano
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
5
|
Gross CC, Schulte-Mecklenbeck A, Madireddy L, Pawlitzki M, Strippel C, Räuber S, Krämer J, Rolfes L, Ruck T, Beuker C, Schmidt-Pogoda A, Lohmann L, Schneider-Hohendorf T, Hahn T, Schwab N, Minnerup J, Melzer N, Klotz L, Meuth SG, Meyer zu Hörste G, Baranzini SE, Wiendl H. Classification of neurological diseases using multi-dimensional cerebrospinal fluid analysis. Brain 2021; 144:2625-2634. [PMID: 33848319 PMCID: PMC8557345 DOI: 10.1093/brain/awab147] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/05/2021] [Accepted: 03/11/2021] [Indexed: 12/19/2022] Open
Abstract
Although CSF analysis routinely enables the diagnosis of neurological diseases, it is mainly used for the gross distinction between infectious, autoimmune inflammatory, and degenerative disorders of the CNS. To investigate, whether a multi-dimensional cellular blood and CSF characterization can support the diagnosis of clinically similar neurological diseases, we analysed 546 patients with autoimmune neuroinflammatory, degenerative, or vascular conditions in a cross-sectional retrospective study. By combining feature selection with dimensionality reduction and machine learning approaches we identified pan-disease parameters that were altered across all autoimmune neuroinflammatory CNS diseases and differentiated them from other neurological conditions and inter-autoimmunity classifiers that subdifferentiate variants of CNS-directed autoimmunity. Pan-disease as well as diseases-specific changes formed a continuum, reflecting clinical disease evolution. A validation cohort of 231 independent patients confirmed that combining multiple parameters into composite scores can assist the classification of neurological patients. Overall, we showed that the integrated analysis of blood and CSF parameters improves the differential diagnosis of neurological diseases, thereby facilitating early treatment decisions.
Collapse
Affiliation(s)
- Catharina C Gross
- Department of Neurology with Institute of Translational Neurology, University and University Hospital Münster, 48149 Münster, Germany
- Correspondence to: Heinz Wiendl Department of Neurology with Institute of Translational Neurology University and University Hospital Münster; Albert-Schweitzer-Campus 1A1 48149 Münster, Germany E-mail: Correspondence may also be addressed to: Catharina C. Gross E-mail:
| | - Andreas Schulte-Mecklenbeck
- Department of Neurology with Institute of Translational Neurology, University and University Hospital Münster, 48149 Münster, Germany
| | - Lohith Madireddy
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Marc Pawlitzki
- Department of Neurology with Institute of Translational Neurology, University and University Hospital Münster, 48149 Münster, Germany
| | - Christine Strippel
- Department of Neurology with Institute of Translational Neurology, University and University Hospital Münster, 48149 Münster, Germany
| | - Saskia Räuber
- Department of Neurology with Institute of Translational Neurology, University and University Hospital Münster, 48149 Münster, Germany
- Present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Julia Krämer
- Department of Neurology with Institute of Translational Neurology, University and University Hospital Münster, 48149 Münster, Germany
| | - Leoni Rolfes
- Department of Neurology with Institute of Translational Neurology, University and University Hospital Münster, 48149 Münster, Germany
| | - Tobias Ruck
- Department of Neurology with Institute of Translational Neurology, University and University Hospital Münster, 48149 Münster, Germany
- Present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Carolin Beuker
- Department of Neurology with Institute of Translational Neurology, University and University Hospital Münster, 48149 Münster, Germany
| | - Antje Schmidt-Pogoda
- Department of Neurology with Institute of Translational Neurology, University and University Hospital Münster, 48149 Münster, Germany
| | - Lisa Lohmann
- Department of Neurology with Institute of Translational Neurology, University and University Hospital Münster, 48149 Münster, Germany
| | - Tilman Schneider-Hohendorf
- Department of Neurology with Institute of Translational Neurology, University and University Hospital Münster, 48149 Münster, Germany
| | - Tim Hahn
- Department of Psychiatry, University of Münster, 48149 Münster, Germany
| | - Nicholas Schwab
- Department of Neurology with Institute of Translational Neurology, University and University Hospital Münster, 48149 Münster, Germany
| | - Jens Minnerup
- Department of Neurology with Institute of Translational Neurology, University and University Hospital Münster, 48149 Münster, Germany
| | - Nico Melzer
- Department of Neurology with Institute of Translational Neurology, University and University Hospital Münster, 48149 Münster, Germany
| | - Luisa Klotz
- Department of Neurology with Institute of Translational Neurology, University and University Hospital Münster, 48149 Münster, Germany
| | - Sven G Meuth
- Department of Neurology with Institute of Translational Neurology, University and University Hospital Münster, 48149 Münster, Germany
- Present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Gerd Meyer zu Hörste
- Department of Neurology with Institute of Translational Neurology, University and University Hospital Münster, 48149 Münster, Germany
| | - Sergio E Baranzini
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University and University Hospital Münster, 48149 Münster, Germany
- Correspondence to: Heinz Wiendl Department of Neurology with Institute of Translational Neurology University and University Hospital Münster; Albert-Schweitzer-Campus 1A1 48149 Münster, Germany E-mail: Correspondence may also be addressed to: Catharina C. Gross E-mail:
| |
Collapse
|
6
|
Hannikainen PA, Kosa P, Barbour C, Bielekova B. Extensive Healthy Donor Age/Gender Adjustments and Propensity Score Matching Reveal Physiology of Multiple Sclerosis Through Immunophenotyping. Front Neurol 2020; 11:565957. [PMID: 33329307 PMCID: PMC7732581 DOI: 10.3389/fneur.2020.565957] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 09/04/2020] [Indexed: 01/09/2023] Open
Abstract
Quantifying cell subpopulations in biological fluids aids in diagnosis and understanding of the mechanisms of injury. Although much has been learned from cerebrospinal fluid (CSF) flow cytometry in neuroimmunological disorders, such as multiple sclerosis (MS), previous studies did not contain enough healthy donors (HD) to derive age- and gender-related normative data and sufficient heterogeneity of other inflammatory neurological disease (OIND) controls to identify MS specific changes. The goals of this blinded training and validation study of MS patients and embedded controls, representing 1,240 prospectively acquired paired CSF/blood samples from 588 subjects was (1) to define physiological age-/gender-related changes in CSF cells, (2) to define/validate cellular abnormalities in blood and CSF of untreated MS through disease duration (DD) and determine which are MS-specific, and (3) to compare effect(s) of low-efficacy (i.e., interferon-beta [IFN-beta] and glatiramer acetate [GA]) and high-efficacy drugs (i.e., natalizumab, daclizumab, and ocrelizumab) on MS-related cellular abnormalities using propensity score matching. Physiological gender differences are less pronounced in the CSF compared to blood, and age-related changes suggest decreased immunosurveillance of CNS by activated HLA-DR+T cells associated with natural aging. Results from patient samples support the concept of MS being immunologically single disease evolving in time. Initially, peripherally activated innate and adaptive immune cells migrate into CSF to form MS lesions. With progression, T cells (CD8+ > CD4+), NK cells, and myeloid dendritic cells are depleted from blood as they continue to accumulate, together with B cells, in the CSF and migrate to CNS tissue, forming compartmentalized inflammation. All MS drugs inhibit non-physiological accumulation of immune cells in the CSF. Although low-efficacy drugs tend to normalize it, high-efficacy drugs overshoot some aspects of CSF physiology, suggesting impairment of CNS immunosurveillance. Comparable inhibition of MS-related CSF abnormalities advocates changes within CNS parenchyma responsible for differences in drug efficacy on MS disability progression. Video summarizing all results may become useful educational tool.
Collapse
Affiliation(s)
| | | | | | - Bibiana Bielekova
- Neuroimmunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
7
|
Anti-CD20 therapy depletes activated myelin-specific CD8 + T cells in multiple sclerosis. Proc Natl Acad Sci U S A 2019; 116:25800-25807. [PMID: 31748274 PMCID: PMC6926057 DOI: 10.1073/pnas.1915309116] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system. CD8+ T cells have been strongly implicated in MS pathogenesis, but it is unclear whether myelin is a CD8+ T cell autoantigenic target in MS. This study demonstrated that while myelin-specific CD8+ T cells are present at similar frequencies in untreated MS patients and healthy subjects, the proportion of memory and CD20-expressing myelin-specific CD8+ T cells was increased in MS patients, suggesting prior antigen encounter. This activated phenotype was reversible as the memory and CD20-expressing populations of certain myelin-specific CD8+ T cells were reduced following anti-CD20 treatment. CD8+ T cells are believed to play an important role in multiple sclerosis (MS), yet their role in MS pathogenesis remains poorly defined. Although myelin proteins are considered potential autoantigenic targets, prior studies of myelin-reactive CD8+ T cells in MS have relied on in vitro stimulation, thereby limiting accurate measurement of their ex vivo precursor frequencies and phenotypes. Peptide:MHC I tetramers were used to identify and validate 5 myelin CD8+ T cell epitopes, including 2 newly described determinants in humans. The validated tetramers were used to measure the ex vivo precursor frequencies and phenotypes of myelin-specific CD8+ T cells in the peripheral blood of untreated MS patients and HLA allele-matched healthy controls. In parallel, CD8+ T cell responses against immunodominant influenza epitopes were also measured. There were no differences in ex vivo frequencies of tetramer-positive myelin-specific CD8+ T cells between MS patients and control subjects. An increased proportion of myelin-specific CD8+ T cells in MS patients exhibited a memory phenotype and expressed CD20 compared to control subjects, while there were no phenotypic differences observed among influenza-specific CD8+ T cells. Longitudinal assessments were also measured in a subset of MS patients subsequently treated with anti-CD20 monoclonal antibody therapy. The proportion of memory and CD20+ CD8+ T cells specific for certain myelin but not influenza epitopes was significantly reduced following anti-CD20 treatment. This study, representing a characterization of unmanipulated myelin-reactive CD8+ T cells in MS, indicates these cells may be attractive targets in MS therapy.
Collapse
|
8
|
Webb LM, Narvaez Miranda J, Amici SA, Sengupta S, Nagy G, Guerau-de-Arellano M. NF-κB/mTOR/MYC Axis Drives PRMT5 Protein Induction After T Cell Activation via Transcriptional and Non-transcriptional Mechanisms. Front Immunol 2019; 10:524. [PMID: 30941147 PMCID: PMC6433977 DOI: 10.3389/fimmu.2019.00524] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 02/26/2019] [Indexed: 12/17/2022] Open
Abstract
Multiple sclerosis is an autoimmune disease of the central nervous system (CNS) mediated by CD4+ T cells and modeled via experimental autoimmune encephalomyelitis (EAE). Inhibition of PRMT5, the major Type II arginine methyltransferase, suppresses pathogenic T cell responses and EAE. PRMT5 is transiently induced in proliferating memory inflammatory Th1 cells and during EAE. However, the mechanisms driving PRMT5 protein induction and repression as T cells expand and return to resting is currently unknown. Here, we used naive mouse and memory mouse and human Th1/Th2 cells as models to identify mechanisms controlling PRMT5 protein expression in initial and recall T cell activation. Initial activation of naive mouse T cells resulted in NF-κB-dependent transient Prmt5 transcription and NF-κB, mTOR and MYC-dependent PRMT5 protein induction. In murine memory Th cells, transcription and miRNA loss supported PRMT5 induction to a lesser extent than in naive T cells. In contrast, NF-κB/MYC/mTOR-dependent non-transcriptional PRMT5 induction played a major role. These results highlight the importance of the NF-κB/mTOR/MYC axis in PRMT5-driven pathogenic T cell expansion and may guide targeted therapeutic strategies for MS.
Collapse
Affiliation(s)
- Lindsay M Webb
- Division of Medical Laboratory Science, Wexner Medical Center, School of Health and Rehabilitation Sciences, College of Medicine, The Ohio State University, Columbus, OH, United States.,Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, United States
| | - Janiret Narvaez Miranda
- Division of Medical Laboratory Science, Wexner Medical Center, School of Health and Rehabilitation Sciences, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Stephanie A Amici
- Division of Medical Laboratory Science, Wexner Medical Center, School of Health and Rehabilitation Sciences, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Shouvonik Sengupta
- Division of Medical Laboratory Science, Wexner Medical Center, School of Health and Rehabilitation Sciences, College of Medicine, The Ohio State University, Columbus, OH, United States.,Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, United States
| | - Gregory Nagy
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, United States
| | - Mireia Guerau-de-Arellano
- Division of Medical Laboratory Science, Wexner Medical Center, School of Health and Rehabilitation Sciences, College of Medicine, The Ohio State University, Columbus, OH, United States.,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States.,Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States.,Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
9
|
The influence and impact of ageing and immunosenescence (ISC) on adaptive immunity during multiple sclerosis (MS) and the animal counterpart experimental autoimmune encephalomyelitis (EAE). Ageing Res Rev 2018; 41:64-81. [PMID: 29101043 DOI: 10.1016/j.arr.2017.10.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 10/23/2017] [Accepted: 10/25/2017] [Indexed: 12/21/2022]
Abstract
The human ageing process encompasses mechanisms that effect a decline in homeostasis with increased susceptibility to disease and the development of chronic life-threatening illness. Increasing age affects the immune system which undergoes a progressive loss of efficiency, termed immunosenescence (ISC), to impact on quantitative and functional aspects of innate and adaptive immunity. The human demyelinating disease multiple sclerosis (MS) and the corresponding animal model experimental autoimmune encephalomyelitis (EAE) are strongly governed by immunological events that primarily involve the adaptive arm of the immune response. MS and EAE are frequently characterised by a chronic pathology and a protracted disease course which thereby creates the potential for exposure to the inherent, on-going effects and consequences of ISC. Collective evidence is presented to confirm the occurrence of established and unendorsed biological markers of ISC during the development of both diseases. Moreover, results are discussed from studies during the course of MS and EAE that reveal a premature upregulation of ISC-related biomarkers which indicates untimely alterations to the adaptive immune system. The effects of ISC and a prematurely aged immune system on autoimmune-associated neurodegenerative conditions such as MS and EAE are largely unknown but current evaluation of data justifies and encourages further investigation.
Collapse
|
10
|
La Rocca C, Carbone F, De Rosa V, Colamatteo A, Galgani M, Perna F, Lanzillo R, Brescia Morra V, Orefice G, Cerillo I, Florio C, Maniscalco GT, Salvetti M, Centonze D, Uccelli A, Longobardi S, Visconti A, Matarese G. Immunometabolic profiling of T cells from patients with relapsing-remitting multiple sclerosis reveals an impairment in glycolysis and mitochondrial respiration. Metabolism 2017; 77:39-46. [PMID: 29132538 PMCID: PMC5800394 DOI: 10.1016/j.metabol.2017.08.011] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 07/28/2017] [Accepted: 08/22/2017] [Indexed: 01/05/2023]
Abstract
BACKGROUND Metabolic reprogramming is shaped to support specific cell functions since cellular metabolism controls the final outcome of immune response. Multiple sclerosis (MS) is an autoimmune disease resulting from loss of immune tolerance against central nervous system (CNS) myelin. Metabolic alterations of T cells occurring during MS are not yet well understood and their studies could have relevance in the comprehension of the pathogenetic events leading to loss of immune tolerance to self and to develop novel therapeutic strategies aimed at limiting MS progression. METHODS AND RESULTS In this report, we observed that extracellular acidification rate (ECAR) and oxygen consumption rate (OCR), indicators of glycolysis and oxidative phosphorylation, respectively, were impaired during T cell activation in naïve-to-treatment relapsing remitting (RR)MS patients when compared with healthy controls. These results were also corroborated at biochemical level by a reduced expression of the glycolitic enzymes aldolase, enolase 1, hexokinase I, and by reduction of Krebs cycle enzymes dihydrolipoamide-S-acetyl transferase (DLAT) and dihydrolipoamide-S-succinyl transferase (DLST). Treatment of RRMS patients with interferon beta-1a (IFN beta-1a) was able to restore T cell glycolysis and mitochondrial respiration as well as the amount of the metabolic enzymes to a level comparable to that of healthy controls. These changes associated with an up-regulation of the glucose transporter-1 (GLUT-1), a key element in intracellular transport of glucose. CONCLUSIONS Our data suggest that T cells from RRMS patients display a reduced engagement of glycolysis and mitochondrial respiration, reversible upon IFN beta-1a treatment, thus suggesting an involvement of an altered metabolism in the pathogenesis of MS.
Collapse
Affiliation(s)
- Claudia La Rocca
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli, Italy
| | - Fortunata Carbone
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli, Italy
| | - Veronica De Rosa
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli, Italy
| | - Alessandra Colamatteo
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli, Italy
| | - Mario Galgani
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli, Italy
| | - Francesco Perna
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Roberta Lanzillo
- Dipartimento di Neuroscienze, Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Vincenzo Brescia Morra
- Dipartimento di Neuroscienze, Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Giuseppe Orefice
- Dipartimento di Neuroscienze, Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Ilaria Cerillo
- Dipartimento di Neuroscienze, Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Ciro Florio
- Dipartimento di Neurologia, Centro regionale di Sclerosi Multipla, Azienda Ospedaliera "A. Cardarelli", Napoli, Italy
| | - Giorgia Teresa Maniscalco
- Dipartimento di Neurologia, Centro regionale di Sclerosi Multipla, Azienda Ospedaliera "A. Cardarelli", Napoli, Italy
| | - Marco Salvetti
- Centro Neurologico Terapie Sperimentali, Dipartimento di Neuroscienze, Salute Mentale e Organi di Senso, Università degli Studi di Roma "La Sapienza", Roma, Italy; IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, Pozzilli (IS), Italy
| | - Diego Centonze
- IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, Pozzilli (IS), Italy; Unità di Neurologia, Dipartimento di Medicina dei Sistemi, Università degli Studi di Roma "Tor Vergata", Roma, Italy
| | - Antonio Uccelli
- Dipartimento di Neuroscienze, Riabilitazione, Oftalmologia, Genetica e Scienze Materno-infantili, Facoltà di Medicina e Chirurgia, Università degli Studi di Genova, Genova, Italy; Ospedale Policlinico San Martino IRCCS, Genova, Italy
| | | | | | - Giuseppe Matarese
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli, Italy; Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Napoli, Italy.
| |
Collapse
|
11
|
Pavelek Z, Vyšata O, Klímová B, Andrýs C, Vokurková D, Vališ M. Lymphocytes in the treatment with interferon beta-1 b. Mult Scler Relat Disord 2017; 18:29-32. [PMID: 29141817 DOI: 10.1016/j.msard.2017.08.020] [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] [Received: 06/30/2017] [Revised: 08/18/2017] [Accepted: 08/31/2017] [Indexed: 11/28/2022]
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic inflammatory demyelinating and neurodegenerative disease affecting the central nervous system. One of the basic medications for the treatment of a clinically isolated syndrome (CIS) or relapsing-remitting MS is interferon beta (INFβ). Although the exact mechanism of its effects is unknown, the medication has an anti-inflammatory and immunomodulatory effect. The goal of this study was to determine the characters which are affected in patients treated with INFβ. METHODS A total of 97 patients (25 males and 72 females) were included into the study. Patients were treated by INFβ 1-b (subcutaneous injection, 250µg, each other day). Clinical evaluations were performed by an attending neurologist. Peripheral blood samples were obtained just prior to treatment and 5 years after INFβ 1-b. Statistical analysis and processing of the obtained data were performed by using a comprehensive statistical software package MATLAB®. RESULTS A significant decrease of the observed parameters after 5 years' of treatment (significant at the 1% significance level) was found in the absolute and relative CD69 count, absolute cytotoxic/suppressor T lymphocyte count, absolute total leukocyte count, absolute natural killer cells count. A significant decrease of the observed parameters after 5 years' of treatment (significant at the 5% significance level) was found in the absolute lymphocyte count, relative cytotoxic/suppressor T lymphocyte count, relative CD3+CD69+ count and absolute CD8+CD38+ count. CONCLUSION The treatment with interferon beta reduces clinical exacerbations in multiple sclerosis (MS) through several known immunomodulatory mechanisms. However, the exact mechanism of effect of this medication is not known. This study presents some parameters that were affected by the long-term INFβ treatment.
Collapse
Affiliation(s)
- Zbyšek Pavelek
- Department of Neurology, Faculty of Medicine and University Hospital Hradec Králové, Charles University in Prague, Hradec Králové, Sokolská 581, 500 05, Czech Republic.
| | - Oldřich Vyšata
- Department of Neurology, Faculty of Medicine and University Hospital Hradec Králové, Charles University in Prague, Hradec Králové, Sokolská 581, 500 05, Czech Republic
| | - Blanka Klímová
- Department of Neurology, Faculty of Medicine and University Hospital Hradec Králové, Charles University in Prague, Hradec Králové, Sokolská 581, 500 05, Czech Republic
| | - Ctirad Andrýs
- Department of Clinical Immunology and Allergology, University Hospital Hradec Králové, Hradec Králové, Sokolská 581, 500 05, Czech Republic
| | - Doris Vokurková
- Department of Clinical Immunology and Allergology, University Hospital Hradec Králové, Hradec Králové, Sokolská 581, 500 05, Czech Republic
| | - Martin Vališ
- Department of Neurology, Faculty of Medicine and University Hospital Hradec Králové, Charles University in Prague, Hradec Králové, Sokolská 581, 500 05, Czech Republic
| |
Collapse
|
12
|
Hellberg S, Eklund D, Gawel DR, Köpsén M, Zhang H, Nestor CE, Kockum I, Olsson T, Skogh T, Kastbom A, Sjöwall C, Vrethem M, Håkansson I, Benson M, Jenmalm MC, Gustafsson M, Ernerudh J. Dynamic Response Genes in CD4+ T Cells Reveal a Network of Interactive Proteins that Classifies Disease Activity in Multiple Sclerosis. Cell Rep 2017; 16:2928-2939. [PMID: 27626663 DOI: 10.1016/j.celrep.2016.08.036] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 06/01/2016] [Accepted: 08/11/2016] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the CNS and has a varying disease course as well as variable response to treatment. Biomarkers may therefore aid personalized treatment. We tested whether in vitro activation of MS patient-derived CD4+ T cells could reveal potential biomarkers. The dynamic gene expression response to activation was dysregulated in patient-derived CD4+ T cells. By integrating our findings with genome-wide association studies, we constructed a highly connected MS gene module, disclosing cell activation and chemotaxis as central components. Changes in several module genes were associated with differences in protein levels, which were measurable in cerebrospinal fluid and were used to classify patients from control individuals. In addition, these measurements could predict disease activity after 2 years and distinguish low and high responders to treatment in two additional, independent cohorts. While further validation is needed in larger cohorts prior to clinical implementation, we have uncovered a set of potentially promising biomarkers.
Collapse
Affiliation(s)
- Sandra Hellberg
- Department of Clinical and Experimental Medicine, Linköping University, 581 83 Linköping, Sweden
| | - Daniel Eklund
- Department of Clinical and Experimental Medicine, Linköping University, 581 83 Linköping, Sweden.
| | - Danuta R Gawel
- The Centre for Individualised Medicine, Department of Clinical and Experimental Medicine, Linköping University, 581 83 Linköping, Sweden
| | - Mattias Köpsén
- The Centre for Individualised Medicine, Department of Clinical and Experimental Medicine, Linköping University, 581 83 Linköping, Sweden; Bioinformatics, Department of Physics, Chemistry and Biology, Linköping University, 581 83 Linköping, Sweden
| | - Huan Zhang
- The Centre for Individualised Medicine, Department of Clinical and Experimental Medicine, Linköping University, 581 83 Linköping, Sweden
| | - Colm E Nestor
- The Centre for Individualised Medicine, Department of Clinical and Experimental Medicine, Linköping University, 581 83 Linköping, Sweden
| | - Ingrid Kockum
- Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, 171 77 Linköping, Sweden
| | - Tomas Olsson
- Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, 171 77 Linköping, Sweden
| | - Thomas Skogh
- Department of Rheumatology and Department of Clinical and Experimental Medicine, Linköping University, 581 83 Linköping, Sweden
| | - Alf Kastbom
- Department of Rheumatology and Department of Clinical and Experimental Medicine, Linköping University, 581 83 Linköping, Sweden
| | - Christopher Sjöwall
- Department of Rheumatology and Department of Clinical and Experimental Medicine, Linköping University, 581 83 Linköping, Sweden
| | - Magnus Vrethem
- Department of Neurology and Department of Clinical and Experimental Medicine, Linköping University, 581 83 Linköping, Sweden
| | - Irene Håkansson
- Department of Neurology and Department of Clinical and Experimental Medicine, Linköping University, 581 83 Linköping, Sweden
| | - Mikael Benson
- The Centre for Individualised Medicine, Department of Clinical and Experimental Medicine, Linköping University, 581 83 Linköping, Sweden
| | - Maria C Jenmalm
- Department of Clinical and Experimental Medicine, Linköping University, 581 83 Linköping, Sweden
| | - Mika Gustafsson
- Bioinformatics, Department of Physics, Chemistry and Biology, Linköping University, 581 83 Linköping, Sweden.
| | - Jan Ernerudh
- Department of Clinical Immunology and Transfusion Medicine and Department of Clinical and Experimental Medicine, Linköping University, 581 83 Linköping, Sweden
| |
Collapse
|
13
|
Varghese S, Cotter M, Chevot F, Fergus C, Cunningham C, Mills KH, Connon SJ, Southern JM, Kelly VP. In vivo modification of tRNA with an artificial nucleobase leads to full disease remission in an animal model of multiple sclerosis. Nucleic Acids Res 2017; 45:2029-2039. [PMID: 28204548 PMCID: PMC5389723 DOI: 10.1093/nar/gkw847] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 08/10/2016] [Accepted: 09/03/2016] [Indexed: 01/07/2023] Open
Abstract
Queuine is a modified pyrrolopyrimidine nucleobase derived exclusively from bacteria. It post-transcriptionally replaces guanine 34 in transfer RNA isoacceptors for Asp, Asn, His and Tyr, in almost all eukaryotic organisms, through the activity of the ancient tRNA guanine transglycosylase (TGT) enzyme. tRNA hypomodification with queuine is a characteristic of rapidly-proliferating, non-differentiated cells. Autoimmune diseases, including multiple sclerosis, are characterised by the rapid expansion of T cells directed to self-antigens. Here, we demonstrate the potential medicinal relevance of targeting the modification of tRNA in the treatment of a chronic multiple sclerosis model—murine experimental autoimmune encephalomyelitis. Administration of a de novo designed eukaryotic TGT substrate (NPPDAG) led to an unprecedented complete reversal of clinical symptoms and a dramatic reduction of markers associated with immune hyperactivation and neuronal damage after five daily doses. TGT is essential for the therapeutic effect, since animals deficient in TGT activity were refractory to therapy. The data suggest that exploitation of the eukaryotic TGT enzyme is a promising approach for the treatment of multiple sclerosis.
Collapse
Affiliation(s)
- Sreeja Varghese
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Trinity College Dublin, Ireland
| | - Michelle Cotter
- School of Chemistry, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Trinity College Dublin, Ireland
| | - Franciane Chevot
- School of Chemistry, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Trinity College Dublin, Ireland
| | - Claire Fergus
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Trinity College Dublin, Ireland
| | - Colm Cunningham
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Trinity College Dublin, Ireland
| | - Kingston H Mills
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Trinity College Dublin, Ireland
| | - Stephen J Connon
- School of Chemistry, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Trinity College Dublin, Ireland
| | - John M Southern
- School of Chemistry, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Trinity College Dublin, Ireland
| | - Vincent P Kelly
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Trinity College Dublin, Ireland
| |
Collapse
|
14
|
Webb LM, Amici SA, Jablonski KA, Savardekar H, Panfil AR, Li L, Zhou W, Peine K, Karkhanis V, Bachelder EM, Ainslie KM, Green PL, Li C, Baiocchi RA, Guerau-de-Arellano M. PRMT5-Selective Inhibitors Suppress Inflammatory T Cell Responses and Experimental Autoimmune Encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2017; 198:1439-1451. [PMID: 28087667 DOI: 10.4049/jimmunol.1601702] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 12/15/2016] [Indexed: 12/22/2022]
Abstract
In the autoimmune disease multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE), expansion of pathogenic, myelin-specific Th1 cell populations drives active disease; selectively targeting this process may be the basis for a new therapeutic approach. Previous studies have hinted at a role for protein arginine methylation in immune responses, including T cell-mediated autoimmunity and EAE. However, a conclusive role for the protein arginine methyltransferase (PRMT) enzymes that catalyze these reactions has been lacking. PRMT5 is the main PRMT responsible for symmetric dimethylation of arginine residues of histones and other proteins. PRMT5 drives embryonic development and cancer, but its role in T cells, if any, has not been investigated. In this article, we show that PRMT5 is an important modulator of CD4+ T cell expansion. PRMT5 was transiently upregulated during maximal proliferation of mouse and human memory Th cells. PRMT5 expression was regulated upstream by the NF-κB pathway, and it promoted IL-2 production and proliferation. Blocking PRMT5 with novel, highly selective small molecule PRMT5 inhibitors severely blunted memory Th expansion, with preferential suppression of Th1 cells over Th2 cells. In vivo, PRMT5 blockade efficiently suppressed recall T cell responses and reduced inflammation in delayed-type hypersensitivity and clinical disease in EAE mouse models. These data implicate PRMT5 in the regulation of adaptive memory Th cell responses and suggest that PRMT5 inhibitors may be a novel therapeutic approach for T cell-mediated inflammatory disease.
Collapse
Affiliation(s)
- Lindsay M Webb
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, College of Medicine, The Ohio State University, Columbus, OH 43210.,Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, OH 43210
| | - Stephanie A Amici
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, College of Medicine, The Ohio State University, Columbus, OH 43210
| | - Kyle A Jablonski
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, College of Medicine, The Ohio State University, Columbus, OH 43210
| | - Himanshu Savardekar
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, College of Medicine, The Ohio State University, Columbus, OH 43210
| | - Amanda R Panfil
- College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210
| | - Linsen Li
- Division of Medicinal Chemistry and Pharmacology, College of Pharmacy, The Ohio State University, Columbus OH 43210
| | - Wei Zhou
- Division of Medicinal Chemistry and Pharmacology, College of Pharmacy, The Ohio State University, Columbus OH 43210
| | - Kevin Peine
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599
| | - Vrajesh Karkhanis
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Eric M Bachelder
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599
| | - Kristy M Ainslie
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599
| | - Patrick L Green
- College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210
| | - Chenglong Li
- Division of Medicinal Chemistry and Pharmacology, College of Pharmacy, The Ohio State University, Columbus OH 43210
| | - Robert A Baiocchi
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Mireia Guerau-de-Arellano
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, College of Medicine, The Ohio State University, Columbus, OH 43210; .,Institute of Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH 43210.,Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210; and.,Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
15
|
Multiple Sclerosis and Obesity: Possible Roles of Adipokines. Mediators Inflamm 2016; 2016:4036232. [PMID: 27721574 PMCID: PMC5046034 DOI: 10.1155/2016/4036232] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/22/2016] [Accepted: 08/08/2016] [Indexed: 12/21/2022] Open
Abstract
Multiple Sclerosis (MS) is an autoimmune disorder of the Central Nervous System that has been associated with several environmental factors, such as diet and obesity. The possible link between MS and obesity has become more interesting in recent years since the discovery of the remarkable properties of adipose tissue. Once MS is initiated, obesity can contribute to increased disease severity by negatively influencing disease progress and treatment response, but, also, obesity in early life is highly relevant as a susceptibility factor and causally related risk for late MS development. The aim of this review was to discuss recent evidence about the link between obesity, as a chronic inflammatory state, and the pathogenesis of MS as a chronic autoimmune and inflammatory disease. First, we describe the main cells involved in MS pathogenesis, both from neural tissue and from the immune system, and including a new participant, the adipocyte, focusing on their roles in MS. Second, we concentrate on the role of several adipokines that are able to participate in the mediation of the immune response in MS and on the possible cross talk between the latter. Finally, we explore recent therapy that involves the transplantation of adipocyte precursor cells for the treatment of MS.
Collapse
|
16
|
Seledtsova GV, Ivanova IP, Shishkov AA, Seledtsov VI. Immune responses to polyclonal T-cell vaccination in patients with progressive multiple sclerosis. J Immunotoxicol 2016; 13:879-884. [PMID: 27602793 DOI: 10.1080/1547691x.2016.1223767] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The overall objective of disease management in autoimmune diseases is to suppress chronic inflammation and prevent organ damage. Therapies often revolve around five drug classes: non-steroidal anti-inflammatory drugs (NSAIDS), anti-malarials, steroids, immunosuppressants, and bio-therapies. However, none of these is a 'cure' and each displays a potential for adverse events. In particular, while all of them suppress harmful autoimmune responses, they also impact on useful protective immune responses. T-Cell receptor (TCR) immunogenicity provides a rationale for T-cell vaccinations to induce anti-idiotypic immune responses with the purpose of down-regulating functionality of idiotype-bearing self-reactive T-cells. To explore this, in this study, 39 patients with progressive (chronic) multiple sclerosis (MS) were multiply immunized with autological polyclonal T-cell vaccines (TCVs). None of the TCV-treated patients experienced any significant side-effects during the entire follow-up period (2 years). T-Cell vaccination had no significant effects on T-cell sub-population contents in the blood of MS patients after 2 years of immunotherapy initiation. However, a substantial reduction in the frequency of CD4+ and CD8+ memory T-cells able to produce interferon (IFN)-γ following activation were noted in the blood of TCV-treated patients. Moreover, significant and sustained reduction in plasma IFNγ levels and concomitant increases in interleukin (IL)-4 levels were documented in these samples. The TCV-treated subjects, however, exhibited no significant changes in plasma IL-17 and IL-18. More importantly was a significant decline in proliferative T-cell responses to myelin antigens in the TCV-treated patients, indicating attenuation of myelin-specific T-cell activity. Collectively, the results suggest that polyclonal T-cell vaccination is safe to use, able to induce measurable, long-lasting, anti-inflammatory immune effects in patients with advanced MS.
Collapse
Affiliation(s)
- Galina V Seledtsova
- a State Research Institute for Fundamental and Clinical Immunology , Novosibirsk , Russia
| | - Irina P Ivanova
- a State Research Institute for Fundamental and Clinical Immunology , Novosibirsk , Russia
| | - Alexey A Shishkov
- a State Research Institute for Fundamental and Clinical Immunology , Novosibirsk , Russia
| | | |
Collapse
|
17
|
Larochelle C, Metz I, Lécuyer MA, Terouz S, Roger M, Arbour N, Brück W, Prat A. Immunological and pathological characterization of fatal rebound MS activity following natalizumab withdrawal. Mult Scler 2016; 23:72-81. [PMID: 27037182 DOI: 10.1177/1352458516641775] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Severe rebound multiple sclerosis (MS) activity is a life-threatening complication of natalizumab (NTZ) withdrawal, for which pathogenesis and treatment are still unclear. We report the immunological and pathological characterization of a case of central nervous system (CNS) inflammatory demyelination after NTZ discontinuation. OBJECTIVE To understand the pathophysiology of this neuroinflammatory condition. METHODS Antemortem blood and cerebrospinal fluid (CSF) analysis was compared with postmortem pathological studies, as well as with novel flow cytometry characterization of immune cells isolated from the CNS parenchyma. RESULTS Pathological analysis of the brain revealed the presence of innumerable active inflammatory demyelinating lesions typical of immunopathological pattern II. Monocytes/macrophages and B cells were enriched in the CNS parenchyma compared to the CSF. Numerous plasma cells were present in the lesions, but CD8 T lymphocytes were predominant in the parenchyma, as opposed to CD4 in the CSF. CNS-infiltrating lymphocytes expressed high levels of adhesion molecules, granzyme B (GzB), interferon-gamma (IFN-γ), and interleukin (IL)-17. CONCLUSIONS Our results underline the differences in immune cell populations between the CSF and the CNS parenchyma, and suggest that aggressive immunosuppressive therapy targeting both T and B lymphocytes is warranted to control the overwhelming CNS inflammation.
Collapse
Affiliation(s)
- Catherine Larochelle
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada/Multiple Sclerosis Clinic, Division of Neurology, CHUM-Notre-Dame Hospital, Montréal, QC, Canada/Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Imke Metz
- Department of Neuropathology, Faculty of Medicine, Universitätsmedizin Göttingen, Göttingen, Germany
| | - Marc-André Lécuyer
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Simone Terouz
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Michel Roger
- Department of Microbiology and Immunology, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Nathalie Arbour
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada/Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Wolfgang Brück
- Department of Neuropathology, Faculty of Medicine, Universitätsmedizin Göttingen, Göttingen, Germany
| | - Alexandre Prat
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada/Multiple Sclerosis Clinic, Division of Neurology, CHUM-Notre-Dame Hospital, Montréal, QC, Canada/Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
18
|
Kozela E, Juknat A, Gao F, Kaushansky N, Coppola G, Vogel Z. Pathways and gene networks mediating the regulatory effects of cannabidiol, a nonpsychoactive cannabinoid, in autoimmune T cells. J Neuroinflammation 2016; 13:136. [PMID: 27256343 PMCID: PMC4891926 DOI: 10.1186/s12974-016-0603-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 05/27/2016] [Indexed: 11/29/2022] Open
Abstract
Background Our previous studies showed that the non-psychoactive cannabinoid, cannabidiol (CBD), ameliorates the clinical symptoms in mouse myelin oligodendrocyte glycoprotein (MOG)35-55-induced experimental autoimmune encephalomyelitis model of multiple sclerosis (MS) as well as decreases the memory MOG35-55-specific T cell (TMOG) proliferation and cytokine secretion including IL-17, a key autoimmune factor. The mechanisms of these activities are currently poorly understood. Methods Herein, using microarray-based gene expression profiling, we describe gene networks and intracellular pathways involved in CBD-induced suppression of these activated memory TMOG cells. Encephalitogenic TMOG cells were stimulated with MOG35-55 in the presence of spleen-derived antigen presenting cells (APC) with or without CBD. mRNA of purified TMOG was then subjected to Illumina microarray analysis followed by ingenuity pathway analysis (IPA), weighted gene co-expression network analysis (WGCNA) and gene ontology (GO) elucidation of gene interactions. Results were validated using qPCR and ELISA assays. Results Gene profiling showed that the CBD treatment suppresses the transcription of a large number of proinflammatory genes in activated TMOG. These include cytokines (Xcl1, Il3, Il12a, Il1b), cytokine receptors (Cxcr1, Ifngr1), transcription factors (Ier3, Atf3, Nr4a3, Crem), and TNF superfamily signaling molecules (Tnfsf11, Tnfsf14, Tnfrsf9, Tnfrsf18). “IL-17 differentiation” and “IL-6 and IL-10-signaling” were identified among the top processes affected by CBD. CBD increases a number of IFN-dependent transcripts (Rgs16, Mx2, Rsad2, Irf4, Ifit2, Ephx1, Ets2) known to execute anti-proliferative activities in T cells. Interestingly, certain MOG35-55 up-regulated transcripts were maintained at high levels in the presence of CBD, including transcription factors (Egr2, Egr1, Tbx21), cytokines (Csf2, Tnf, Ifng), and chemokines (Ccl3, Ccl4, Cxcl10) suggesting that CBD may promote exhaustion of memory TMOG cells. In addition, CBD enhanced the transcription of T cell co-inhibitory molecules (Btla, Lag3, Trat1, and CD69) known to interfere with T/APC interactions. Furthermore, CBD enhanced the transcription of oxidative stress modulators with potent anti-inflammatory activity that are controlled by Nfe2l2/Nrf2 (Mt1, Mt2a, Slc30a1, Hmox1). Conclusions Microarray-based gene expression profiling demonstrated that CBD exerts its immunoregulatory effects in activated memory TMOG cells via (a) suppressing proinflammatory Th17-related transcription, (b) by promoting T cell exhaustion/tolerance, (c) enhancing IFN-dependent anti-proliferative program, (d) hampering antigen presentation, and (d) inducing antioxidant milieu resolving inflammation. These findings put forward mechanism by which CBD exerts its anti-inflammatory effects as well as explain the beneficial role of CBD in pathological memory T cells and in autoimmune diseases. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0603-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ewa Kozela
- The Dr Miriam and Sheldon G. Adelson Center for the Biology of Addictive Diseases, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 6997801, Israel. .,Department of Neurobiology, Weizmann Institute of Science, Rehovot, 76100, Israel.
| | - Ana Juknat
- The Dr Miriam and Sheldon G. Adelson Center for the Biology of Addictive Diseases, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 6997801, Israel.,Department of Neurobiology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Fuying Gao
- Departments of Psychiatry and Neurology, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Nathali Kaushansky
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Giovanni Coppola
- Departments of Psychiatry and Neurology, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Zvi Vogel
- The Dr Miriam and Sheldon G. Adelson Center for the Biology of Addictive Diseases, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 6997801, Israel.,Department of Neurobiology, Weizmann Institute of Science, Rehovot, 76100, Israel
| |
Collapse
|
19
|
Plantone D, De Angelis F, Doshi A, Chataway J. Dimethyl fumarate may still have a role in progressive multiple sclerosis. Ther Adv Neurol Disord 2016; 9:344-5. [PMID: 27366242 DOI: 10.1177/1756285616640396] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Domenico Plantone
- NMR Research Unit, Queen Square MS Centre, UCL Institute of Neurology, London WC1E 6BT, UK
| | - Floriana De Angelis
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, University College London (UCL), London, UK
| | - Anisha Doshi
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, University College London (UCL), London, UK
| | - Jeremy Chataway
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, University College London (UCL), London, UK
| |
Collapse
|
20
|
Ho EL, Ronquillo R, Altmeppen H, Spudich SS, Price RW, Sinclair E. Cellular Composition of Cerebrospinal Fluid in HIV-1 Infected and Uninfected Subjects. PLoS One 2013; 8:e66188. [PMID: 23822975 PMCID: PMC3688831 DOI: 10.1371/journal.pone.0066188] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 05/07/2013] [Indexed: 11/18/2022] Open
Abstract
In order to characterize the cellular composition of cerebrospinal fluid (CSF) in a healthy state and in the setting of chronic pleocytosis associated with HIV-1 (HIV) infection, multi-parameter flow cytometry was used to identify and quantitate cellular phenotypes in CSF derived from HIV-uninfected healthy controls and HIV-infected subjects across a spectrum of disease and treatment. CD4+ T cells were the most frequent CSF population and the CD4:CD8 ratio was significantly increased in the CSF compared to blood (p = 0.0232), suggesting preferential trafficking of CD4+ over CD8+ T cells to this compartment. In contrast, in HIV-infection, CD8+ T cells were the major cellular component of the CSF and were markedly increased compared to HIV-uninfected subjects (p<0.001). As with peripheral blood, the CSF CD4:CD8 ratio was reversed in HIV-infected subjects compared to HIV-uninfected subjects. Monocytes, B cells and NK cells were rare in the CSF in both groups, although absolute counts of CSF NK cells and B cells were significantly increased in HIV-infected subjects (p<0.05). Our studies show that T cells are the major cellular component of the CSF in HIV-infected and uninfected subjects. The CSF pleocytosis characteristic of HIV infection involves all lymphocyte subsets we measured, except for CD4+ T cells, but is comprised primarily of CD8+ T cells. The reduced proportion of CD4+ T cells in the CSF may reflect both HIV-related peripheral loss and changes in trafficking patterns in response to HIV infection in the central nervous system.
Collapse
Affiliation(s)
- Emily L. Ho
- Department of Neurology, Harborview Medical Center, University of Washington, Seattle, Washington, United States of America
| | - Rollie Ronquillo
- Department of Immunology and Microbiology, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Hermann Altmeppen
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Serena S. Spudich
- Department of Neurology, Yale University, New Haven, Connecticut, United States of America
| | - Richard W. Price
- Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
| | - Elizabeth Sinclair
- Division of Experimental Medicine, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
21
|
Zafranskaya MM, Nizheharodova DB, Yurkevich MY, Lamouskaya NV, Motuzova YM, Bagatka SS, Ivanchik HI, Fedulov AS. In vitro assessment of mesenchymal stem cells immunosuppressive potential in multiple sclerosis patients. Immunol Lett 2013; 149:9-18. [PMID: 23089549 DOI: 10.1016/j.imlet.2012.10.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 09/20/2012] [Accepted: 10/12/2012] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cells (MSC) are promising for multiple sclerosis (MS) treatment. However, clinical results remain controversial, and no criteria are available for predicting the efficiency of MSC therapy. Using an in vitro model of lymphocytes and MSC cocultivation we revealed that the Index of MSC Suppression of myelin-induced memory T cells proliferation was stronger than that of PHA-stimulated proliferation and inversely correlated with patients'EDSS score. In vitro expression of CD119 (IFNGR1) in mitogen/myelin-stimulated T cells increased in the presence of MSC being inversely correlated with T-lymphocytes proliferation. The Index of MSC Suppression and CD119 expression in T-lymphocytes may be useful when assessing MSC immunosuppressive potential in MS patients.
Collapse
Affiliation(s)
- Marina M Zafranskaya
- Belarusian Medical Academy of Post-Graduate Education, 3, Brovki str., build. 3, Minsk 220013, Belarus.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Immunoregulatory T cells in multiple sclerosis and the effect of interferon beta and glatiramer acetate treatment on T cell subpopulations. J Neurol Sci 2012; 319:18-23. [DOI: 10.1016/j.jns.2012.05.036] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 05/10/2012] [Accepted: 05/14/2012] [Indexed: 11/20/2022]
|
23
|
Mullen KM, Gocke AR, Allie R, Ntranos A, Grishkan IV, Pardo C, Calabresi PA. Expression of CCR7 and CD45RA in CD4+ and CD8+ subsets in cerebrospinal fluid of 134 patients with inflammatory and non-inflammatory neurological diseases. J Neuroimmunol 2012; 249:86-92. [PMID: 22633193 DOI: 10.1016/j.jneuroim.2012.04.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 04/21/2012] [Accepted: 04/25/2012] [Indexed: 10/28/2022]
Abstract
We investigated CD45RA and CCR7 expression in CD4+ and CD8+ subsets of cerebrospinal fluid (CSF) lymphocytes, both immediately ex vivo and after stimulation, from 134 patients with a variety of inflammatory and non-inflammatory neurological diseases. Most inflammatory diseases had a higher CD4+:CD8+ ratio and higher percentage of effector memory T cells (T(EM)) than non-inflammatory controls, excluding active infection. Moreover, we found that patients with highly elevated cell counts in the CSF tended to have a lower percentage of central memory T cells (T(CM)) than patients with low or absent pleocytosis, with a concomitant increase in T(EM). We also found that samples with elevated IgG index or presence of oligoclonal bands had a significantly higher CD4+:CD8+ ratio than normal samples, consistent with increased CD4+ help for intrathecal IgG synthesis by B cells.
Collapse
Affiliation(s)
- Katherine M Mullen
- Johns Hopkins School of Medicine, Department of Neurology, 600 N. Wolfe St., Baltimore, MD 21287, United States
| | | | | | | | | | | | | |
Collapse
|
24
|
Tsakiri A, Kjærsgaard E, Grigoriadis N, Svane IM, Frederiksen JL. Effector and regulatory T cells in patients with acute optic neuritis. Neuroimmunomodulation 2012; 19:111-20. [PMID: 22248727 DOI: 10.1159/000330242] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 06/17/2011] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVE Optic neuritis (ON) is an autoimmune acute demyelinating disease of the optic nerve and may occur in patients with confirmed multiple sclerosis (MS) or as a clinically isolated syndrome. T lymphocytes play a central role in the pathogenesis of MS. The phenotype of different T cell subsets is usually characterized by the expression of distinct cell surface receptors such as CD45RA, CD45RO, CCR7, CD27 and CD28. The aim of this study was to characterize the phenotype of distinct subsets of CD4 and CD8 T cells in patients with isolated ON. METHODS CD4 and CD8 T cell subsets were characterized by flow cytometry in fresh peripheral blood and cerebrospinal fluid (CSF) samples using the surface markers CD27, CD25, CD45RA, CD45RO and intracellular FOXP3. The T cell subsets expressed in patients with acute ON (n = 64; symptom onset of ON within the preceding 28 days) were compared to those of a gender- and age-matched healthy control (HC) group (n = 32). RESULTS Both CD4+ and CD8+ naïve T cells in the ON group were significantly reduced in the CSF. In contrast, most of the intermediate-stage and late effector CD4+ and CD8+ T cell subsets as well as the CD4+ T regulatory cells were expressed in ON patients, though not at all in the CSF of the HC group. CONCLUSION These results add important evidence for inflammatory and regulatory activity in ON and early MS.
Collapse
Affiliation(s)
- Anna Tsakiri
- Department of Neurology, Glostrup Hospital, University of Copenhagen, Glostrup, Denmark.
| | | | | | | | | |
Collapse
|
25
|
de Graaf MT, de Jongste AHC, Kraan J, Boonstra JG, Smitt PAES, Gratama JW. Flow cytometric characterization of cerebrospinal fluid cells. CYTOMETRY PART B-CLINICAL CYTOMETRY 2011; 80:271-81. [DOI: 10.1002/cyto.b.20603] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Revised: 04/12/2011] [Accepted: 04/16/2011] [Indexed: 12/12/2022]
|
26
|
Mikulkova Z, Praksova P, Stourac P, Bednarik J, Michalek J. Imbalance in T-cell and cytokine profiles in patients with relapsing-remitting multiple sclerosis. J Neurol Sci 2011; 300:135-41. [PMID: 20884014 DOI: 10.1016/j.jns.2010.08.053] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Revised: 08/01/2010] [Accepted: 08/25/2010] [Indexed: 01/31/2023]
Abstract
Multiple sclerosis (MS) is characterized by autoimmune attack leading to demyelination of the white matter in the central nervous system with devastating clinical consequences. Several immune-mediated destruction mechanisms were previously proposed including different T-cell subsets but complex view on immune system function in patients with MS is missing. In the present study, T-lymphocyte populations and pro-inflammatory as well as suppressive cytokine profiles were evaluated in detail in previously untreated patients with relapsing-remitting MS (RRMS). CD4(+) and CD8(+) naïve, central memory (Tcm), effector memory (Tem), terminal effector memory (Ttem), CD4(+) regulatory T-cells (Treg) and CD8(+) T-suppressor cells (Ts) were analysed using flow cytometry, and levels of ten plasma cytokines were determined using fluorescent bead-based immunoassay. We evaluated two groups of RRMS with minor (n=33) and major (n=25) clinical impairment and compared them with healthy controls (n=40) in order to detect any correlation between severity of MS clinical symptoms and immune disturbances. Significant differences were noted in CD4(+)CD45RA(+)CCR7(+) naïve T-cells, CD4(+)CD45RO(+)CCR7(-) and CD8(+)CD45RO(+)CCR7(-) Tem cells, while no differences were recognized in Tcm, Ttem, Treg and Ts cells in RRMS patients. Nine out of ten studied cytokines were disturbed in plasma samples of patients with RRMS. In conclusion, we demonstrate complex immune dysbalances in untreated MS patients.
Collapse
Affiliation(s)
- Z Mikulkova
- University Cell Immunotherapy Center, Babak Research Institute, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00 Brno, Republic
| | | | | | | | | |
Collapse
|
27
|
Ferrandi C, Richard F, Tavano P, Hauben E, Barbié V, Gotteland JP, Greco B, Fortunato M, Mariani MF, Furlan R, Comi G, Martino G, Zaratin PF. Characterization of immune cell subsets during the active phase of multiple sclerosis reveals disease and c-Jun N-terminal kinase pathway biomarkers. Mult Scler 2010; 17:43-56. [PMID: 20855355 DOI: 10.1177/1352458510381258] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Autoimmune activation and deregulated apoptosis of T lymphocytes are involved in multiple sclerosis (MS). c-Jun N-terminal kinase (JNK) plays a role in T-cell survival and apoptosis. OBJECTIVES The aim of this work was to investigate the role of the JNK-dependent apoptosis pathway in relapsing-remitting MS (RRMS). METHODS The immunomodulatory effect of AS602801, a JNK inhibitor, was firstly evaluated on activated peripheral blood mononuclear cells (PBMCs) from healthy volunteers (HVs) and secondly in unstimulated purified CD4+, CD8+ and CD11b+ cells from RRMS patients and HVs. Moreover JNK/inflammation/apoptosis related genes were investigated in RRMS and HV samples. RESULTS In activated PBMCs from HVs, we showed that AS602801 blocked T-lymphocyte proliferation and induced apoptosis. In RRMS CD4+ and CD8+ cells, AS602801 induced apoptosis genes and expression of surface markers, while in RRMS CD11b+ cells it induced expression of innate immunity receptors and co-stimulatory molecules. Untreated cells from RRMS active-phase patients significantly released interleukin-23 (IL-23) and interferon-gamma (IFN-γ) and expressed less apoptosis markers compared to the cells of HVs. Moreover, gene expression was significantly different in cells from RRMS active-phase patients vs. HVs. By comparing RRMS PBMCs in the active and stable phases, a specific genomic signature for RRMS was indentified. Additionally, CASP8AP2, CD36, ITGAL, NUMB, OLR1, PIAS-1, RNASEL, RTN4RL2 and THBS1 were identified for the first time as being associated to the active phase of RRMS. CONCLUSIONS The analysis of the JNK-dependent apoptosis pathway can provide biomarkers for activated lymphocytes in the active phase of RRMS and a gene expression signature for disease status. The reported results might be useful to stratify patients, thereby supporting the development of novel therapies.
Collapse
|
28
|
Numerical defects in CD8+CD28- T-suppressor lymphocyte population in patients with type 1 diabetes mellitus and multiple sclerosis. Cell Immunol 2010; 262:75-9. [PMID: 20219185 DOI: 10.1016/j.cellimm.2010.02.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2009] [Revised: 01/18/2010] [Accepted: 02/01/2010] [Indexed: 01/12/2023]
Abstract
Type 1 diabetes mellitus (T1D) and multiple sclerosis (MS) are organ-specific autoimmune diseases leading to an attack of auto-aggressive lymphocytes against the pancreatic beta-cells and central nervous system, respectively. Using four-colour flow cytometry, T-lymphocyte populations having an important function in autoimmune processes were analyzed. T-regulatory cells (Treg) CD4(+)CD25(+)CD127(low), T-suppressor cells (Ts) CD8(+)CD28(-), activated helper CD4(+)CD25(+)CD127(+) and cytotoxic CD8(+)CD25(+) T-cells and also naive CD4(+)CD45RA(+) and memory T-cells CD4(+)CD45RO(+) were compared in the group of patients with T1D (n=30), MS (n=31) and in the group of healthy controls (n=29). Significant differences in Ts cells, activated helper and cytotoxic cells and also memory T-cells were recognized in the group of T1D patients compared to healthy controls. Ts population was significantly lowered in MS patients as well. However, no significant differences were noticed in Treg population. The observed data demonstrate significant differences among patients with T1D and MS in comparison to healthy individuals.
Collapse
|
29
|
Aristimuño C, de Andrés C, Bartolomé M, de las Heras V, Martínez-Ginés ML, Arroyo R, Fernández-Cruz E, Sánchez-Ramón S. IFNbeta-1a therapy for multiple sclerosis expands regulatory CD8+ T cells and decreases memory CD8+ subset: a longitudinal 1-year study. Clin Immunol 2009; 134:148-57. [PMID: 19900844 DOI: 10.1016/j.clim.2009.09.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2009] [Revised: 09/18/2009] [Accepted: 09/23/2009] [Indexed: 11/28/2022]
Abstract
The beneficial effects of interferon beta-1a (IFNbeta-1a) in multiple sclerosis (MS) remain only partially understood. CD8(+) T cells are key cells in MS pathogenesis that contribute to axonal damage in MS, whereas CD4(+) regulatory T cells (T(Reg)) and CD8(+) regulatory/suppressor T cells (Ts) play an important role in protecting against subsequent MS activity. We analysed ex vivo changes on T(Reg) and on the different subsets of CD4(+) and CD8(+) T lymphocytes, before IFNbeta-1a (Rebif) therapy and at 3, 6, and 12 months after treatment, in 23 MS patients and in 26 healthy controls. IFNbeta-1a significantly increased the proportions of CD4(+) T(Reg) and regulatory CD8(+) T cells (Tr). Memory CD8(+) T cells were significantly decreased after 1 year of treatment, maybe reflecting down-regulation of abnormally persistent systemic activation in MS patients. After 1 year of IFNbeta-1a, a direct correlation was observed between plasmacytoid dendritic cells and effector CD8(+) T cells.
Collapse
Affiliation(s)
- Carol Aristimuño
- Department of Neurology, Gregorio Marañón University General Hospital, Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Immunological and histochemical analyses of cerebrospinal fluid and peripheral blood from patients with neurological and psychiatric disorders. Acta Neuropsychiatr 2009; 21 Suppl 2:51-7. [PMID: 25384871 DOI: 10.1017/s0924270800032737] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Epidemiological, clinical and post mortem studies indicate that inflammatory and immune reactions are involved in the pathomechanisms of affective and schizophrenic spectrum disorders. However, in psychiatric patients, only sporadic investigation on immunochemistry has been performed and information about immunofunction derived by investigation of immunocompetent cells in the CSF is not available to date. Here we present an interdisciplinary work of neurologists, psychiatrists and hemato-immunologists focusing on the immunology of psychiatric and neurological disorders. In a first study including 63 patients with therapy resistant affective and schizophrenic spectrum disorders we applied conventional, validated neurological CSF investigation such as analysis of albumin, IgG, IgA, IgM, oligoclonal IgG and specific antibodies, cell count and interpreted the data by Reibergrams. In a second study, we applied the highly sensitive and specific multicolour flowcytometry of paired samples of CSF and peripheral blood cells to characterize the immunostatus of psychiatric and neurological patients. We demonstrate that flowcytometry technology constitutes an appropriate method to investigate subsets of lymphocytes even with low CSF cell numbers, and therefore as a promising diagnostic tool for routine purposes in the differential diagnosis of psychiatric diseases. Furthermore, knowledge of the frequencies of T cell subsets such as the T regulatory cell type might open new avenues to models of psychiatric and neurological diseases as well as diagnostic and monitoring implications.
Collapse
|
31
|
McCoy L, Tsunoda I, Fujinami RS. Multiple sclerosis and virus induced immune responses: autoimmunity can be primed by molecular mimicry and augmented by bystander activation. Autoimmunity 2008; 39:9-19. [PMID: 16455578 DOI: 10.1080/08916930500484799] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Polymicrobial infections have been associated with plausible immune mediated diseases, including multiple sclerosis (MS). Virus infection can prime autoimmune T cells specific for central nervous system (CNS) antigens, if virus has molecular mimicry with CNS proteins. On the other hand, infection of irrelevant viruses will induce two types of cytokine responses. Infection with a virus such as lymphocytic choriomeningitis virus (LCMV), can induce interferon (IFN)-alpha/beta production and suppress autoimmunity, while infection with a virus, such as murine cytomegalovirus (MCMV), can activate natural killer (NK), NKT and dendritic cells, resulting in interleukin (IL)-12 and IFN-gamma production. These cytokines can cause bystander activation of autoreactive T cells. We established an animal model, where mice infected with vaccinia virus encoding myelin protein can mount autoimmune responses. However, the mice develop clinical disease only after irrelevant immune activation either with complete Freund's adjuvant or MCMV infection. In this review, we propose that a combination of two mechanisms, molecular mimicry and bystander activation, induced by virus infection, can lead to CNS demyelinating diseases, including MS. Viral proteins having molecular mimicry with self-proteins in the CNS can prime genetically susceptible individuals. Once this priming has occurred, an immunologic challenge could result in disease through bystander activation by cytokines.
Collapse
Affiliation(s)
- Lori McCoy
- University of Utah School of Medicine, Department of Neurology, 30 North 1900 East, Room 3R330, Salt Lake City, UT 84132-2305, USA
| | | | | |
Collapse
|
32
|
Phillips SM, Bhopale MK, Constantinescu CS, Ciric B, Hilliard B, Ventura E, Lavi E, Rostami A. Effect of DAB(389)IL-2 immunotoxin on the course of experimental autoimmune encephalomyelitis in Lewis rats. J Neurol Sci 2007; 263:59-69. [PMID: 17603081 DOI: 10.1016/j.jns.2007.05.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2006] [Revised: 04/26/2007] [Accepted: 05/08/2007] [Indexed: 12/11/2022]
Abstract
Activated T cells express the high affinity interleukin 2 receptor (IL-2R also CD25) that binds interleukin 2 (IL-2) and transduces signals important for the proliferation and survival of these cells. We investigated the effect of the genetically engineered immunotoxin DAB(389)IL-2 on experimental autoimmune encephalomyelitis (EAE), an autoimmune disease of the central nervous system (CNS) mediated by activated myelin-reactive T cells. EAE is the most commonly used animal model of the human disease multiple sclerosis (MS). DAB(389)IL-2 is a recombinant fusion product made of a portion of diphtheria toxin, which contains binding and translocation components of the toxin linked to IL-2. The diphtheria toxin targets and kills cells expressing the high affinity IL-2 receptor and has been successfully used in several autoimmune and neoplastic conditions. We observed a significant suppression of guinea-pig spinal cord homogenate (gpSCH)-MBP induced active EAE in Lewis rats at 2 x 1,600 kU of DAB(389)IL-2 given on days 7 and 9 post-immunization and complete suppression with the same dose on days 7, 8 and 9 or 7, 8, 9 and 10 after immunization during the active disease period. There were reduced mononuclear cell infiltrates of CD4(+), CD8(+), CD25(+) and alphabetaTCR(+) T cells in the spinal cord of treated rats. However, treatment at day 11 or 12 post-immunization led to severe, fatal disease. The toxin added to cultures in vitro or injected in vivo suppressed antigen- and mitogen-induced T cell proliferation. DAB(389)IL-2 treatment in vivo or exposure of encephalitogenic T cells in vitro prior to transfer did have a significant inhibitory effect on adoptive transfer EAE. Our data demonstrate that DAB(389)IL-2 immunotoxin can suppress active and passive EAE if applied at specific, early time points, but can have negative consequences at later time points.
Collapse
MESH Headings
- Animals
- Antigens, CD/metabolism
- Cell Proliferation/drug effects
- Cells, Cultured
- Diphtheria Toxin/genetics
- Diphtheria Toxin/therapeutic use
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Female
- Guinea Pigs
- Immunotoxins/therapeutic use
- Interleukin-2/genetics
- Interleukin-2/therapeutic use
- Lymphocyte Activation/drug effects
- Mutation/physiology
- Myelin Basic Protein
- Rats
- Rats, Inbred Lew
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/therapeutic use
- Severity of Illness Index
- Spinal Cord/pathology
- T-Lymphocytes, Helper-Inducer/drug effects
- Time Factors
Collapse
Affiliation(s)
- S Michael Phillips
- Department of Allergy and Immunology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Jilek S, Schluep M, Rossetti AO, Guignard L, Le Goff G, Pantaleo G, Du Pasquier RA. CSF enrichment of highly differentiated CD8+ T cells in early multiple sclerosis. Clin Immunol 2007; 123:105-13. [PMID: 17188575 DOI: 10.1016/j.clim.2006.11.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2006] [Revised: 10/06/2006] [Accepted: 11/08/2006] [Indexed: 01/21/2023]
Abstract
CD8+ T cells may play an important role in multiple sclerosis (MS). Whether these cells would be involved in early stages of MS is unclear. We enrolled 52 patients with suspected MS, determined the recruitment of their highly differentiated (CCR7-/CD45RA+ or -) T cells (T(HD)) in the CSF as compared to peripheral blood and followed them for 12+/-7.3 months. A ROC curve showed that a CD8+/CD4+ T(HD) cells ratio of 0.94 helped to distinguish relapsing-remitting (RR-MS) and possible MS (Po-MS) from primary-progressive MS (PP-MS) and other neurological diseases patients (OND) patients (p=0.039), risk ratio of 2.29 (95% CI: 1.13-4.66; p=0.006). The CSF enrichment in CD8+ T(HD) cells was greater than in CD4+ T(HD) cells in RR/Po-MS patients (p=0.024) and than in CD8+ T(HD) cells in PP-MS/OND patients (p=0.006). These data suggest that CD8+ T(HD) cells play a role in the early stages of RR-MS.
Collapse
Affiliation(s)
- Samantha Jilek
- Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
34
|
Zafranskaya M, Oschmann P, Engel R, Weishaupt A, van Noort JM, Jomaa H, Eberl M. Interferon-beta therapy reduces CD4+ and CD8+ T-cell reactivity in multiple sclerosis. Immunology 2006; 121:29-39. [PMID: 17239199 PMCID: PMC2265917 DOI: 10.1111/j.1365-2567.2006.02518.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Therapy with interferon-beta (IFN-beta) has well-established clinical effects in multiple sclerosis (MS), albeit the immunomodulatory mechanisms are not fully understood. We assessed the prevalence and functional capacity of CD4+ and CD8+ T cells in healthy donors, and in untreated and IFN-beta-treated MS patients, in response to myelin oligodendrocyte glycoprotein (MOG). The proportion of CD45RO+ memory T cells was higher in MS patients than in healthy donors, but returned to normal values upon therapy with IFN-beta. While CD45RO+ CD4+ T cells from all three groups responded to MOG in vitro, untreated patients showed augmented proliferative responses compared to healthy individuals and IFN-beta treatment reduced this elevated reactivity back to the values observed in healthy donors. Similarly, the response of CD45RO+ CD8+ T cells to MOG was strongest in untreated patients and decreased to normal values upon immunotherapy. Overall, the frequency of peripheral CD45RO+ memory T cells ex vivo correlated with the strength of the cellular in vitro response to MOG in untreated patients but not in healthy donors or IFN-beta-treated patients. Compared with healthy individuals, responding CD4+ and CD8+ cells were skewed towards a type 1 cytokine phenotype in untreated patients, but towards a type 2 phenotype under IFN-beta therapy. Our data suggest that the beneficial effect of IFN-beta in MS might be the result of the suppression or depletion of autoreactive, pro-inflammatory memory T cells in the periphery. Assessment of T-cell subsets and their reactivity to MOG may represent an important diagnostic tool for monitoring successful immunotherapy in MS.
Collapse
Affiliation(s)
- Marina Zafranskaya
- Biochemisches Institut, Infektiologie, Justus-Liebig-Universität Giessen, Germany
| | | | | | | | | | | | | |
Collapse
|
35
|
Ndejembi MP, Tang AL, Farber DL. Reshaping the past: Strategies for modulating T-cell memory immune responses. Clin Immunol 2006; 122:1-12. [PMID: 16916619 DOI: 10.1016/j.clim.2006.06.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2006] [Accepted: 06/26/2006] [Indexed: 02/08/2023]
Abstract
Memory T cells are generated following an initial encounter with antigen, persist over the lifetime of an individual, and mediate rapid and robust functional responses upon antigenic recall. While immune memory is generally associated with protective immune response to pathogens, memory T cells can be generated to diverse types of antigens including autoantigens and alloantigens through homologous or crossreactive priming and comprise the majority of circulating T cells during adulthood. Memory T cells can therefore play critical roles in propagating and perpetuating autoimmune disease and in mediating allograft rejection, although the precise pathways for regulation of memory immune responses remain largely undefined. Moreover, evaluating and designing strategies to modulate memory T-cell responses are challenging given the remarkable heterogeneity of memory T cells, with different subsets predominating in lymphoid versus non-lymphoid tissue sites. In this review, we discuss what is presently known regarding the effect of current immunomodulation strategies on the memory T-cell compartment and potential strategies for controlling immunological recall.
Collapse
Affiliation(s)
- Modesta P Ndejembi
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | |
Collapse
|
36
|
Okuda Y, Apatoff BR, Posnett DN. Apoptosis of T cells in peripheral blood and cerebrospinal fluid is associated with disease activity of multiple sclerosis. J Neuroimmunol 2006; 171:163-70. [PMID: 16290072 DOI: 10.1016/j.jneuroim.2005.09.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2005] [Accepted: 09/30/2005] [Indexed: 12/20/2022]
Abstract
Apoptotic elimination of pathogenic T cells is considered to be one of regulatory mechanisms in multiple sclerosis (MS). To explore the potential relationship between Fas-mediated apoptosis and the disease course of MS, we examined apoptosis, defined by annexin V (AV) binding, and Fas (CD95) expression in CD4+ and in CD8+ T cells in MS patients by using five-color flow cytometry. The percentage of AV+CD4+CD3+ cells and CD95+AV+CD4+CD3+ cells in peripheral blood and cerebrospinal fluid (CSF) were significantly decreased in active MS patients compared with inactive MS patients. A significantly lower proportion of CD95+AV+CD8+CD3+ cells in CSF was observed in active MS patients compared with inactive MS patients, but not in peripheral blood. These results indicate that the resistance of T cells to Fas-mediated apoptosis is involved in exacerbation of MS and/or that Fas-mediated apoptosis of T cells is associated with remission of MS.
Collapse
Affiliation(s)
- Yoshinobu Okuda
- Department of Medicine, Division of Hematology-Oncology, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10021, USA.
| | | | | |
Collapse
|