Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.288
Filter
1.
Front Immunol ; 15: 1439393, 2024.
Article in English | MEDLINE | ID: mdl-39238642

ABSTRACT

Objective: To evaluate the safety of COVID-19 vaccines in patients with multiple sclerosis (MS) by assessing their impact on serum neurofilament light chain (sNfL) levels as a marker of neuroaxonal damage. Methods: Single-center observational longitudinal study including patients with MS who consecutively received their initial vaccination against SARS-CoV-2 at Hospital Universitario Ramón y Cajal, following the first national immunization program in Spain. Serum samples were collected at baseline and after receiving the second dose of the vaccine. sNfL levels were quantified using the single molecule array (SIMOA) technique. Adverse events, including clinical or radiological reactivation of the disease, were recorded. Results: Fifty-two patients were included (median age, 39.7 years [range, 22.5-63.3]; 71.2% female). After SARS-CoV-2 vaccination, no increased inflammatory activity, either determined by the presence of relapses and/or new MRI lesions and/or high sNfL levels, was detected. Accordingly, there was no difference between median sNfL levels before and after vaccination (5.39 vs. 5.76 pg/ml, p=0.6). Despite this, when looking at baseline patient characteristics before vaccination, younger age associated with disease activity after vaccination (OR 0.87, 95% CI: 0.77-0.98, p=0.022). Larger studies are needed to validate these results. Conclusion: COVID-19 vaccines did not cause reactivation of disease at a clinical, radiological or molecular level, thus suggesting that they are safe in MS patients.


Subject(s)
COVID-19 Vaccines , COVID-19 , Multiple Sclerosis , Neurofilament Proteins , SARS-CoV-2 , Humans , Female , Male , Adult , Middle Aged , Multiple Sclerosis/immunology , Multiple Sclerosis/blood , COVID-19/immunology , COVID-19/prevention & control , COVID-19/blood , COVID-19 Vaccines/adverse effects , COVID-19 Vaccines/immunology , Neurofilament Proteins/blood , SARS-CoV-2/immunology , Young Adult , Longitudinal Studies , Biomarkers/blood , Axons/pathology , Spain/epidemiology , Vaccination/adverse effects
2.
Neurol Neuroimmunol Neuroinflamm ; 11(6): e200306, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39231385

ABSTRACT

BACKGROUND AND OBJECTIVES: The rs763361 nonsynonymous variant in the CD226 gene, which results in a glycine-to-serine substitution at position 307 of the CD226 protein, has been implicated as a risk factor of various immune-mediated diseases, including multiple sclerosis (MS). Compelling evidence suggests that this allele may play a significant role in predisposing individuals to MS by decreasing the immune-regulatory capacity of Treg cells and increasing the proinflammatory potential of effector CD4 T cells. However, the impact of this CD226 gene variant on CD8 T-cell functions, a population that also plays a key role in MS, remains to be determined. METHODS: To study whether the CD226 risk variant affects human CD8 T-cell functions, we used CD8 T cells isolated from peripheral blood mononuclear cell of 16 age-matched healthy donors homozygous for either the protective or the risk allele of CD226. We characterized these CD8 T cells on T-cell receptor (TCR) stimulation using high-parametric flow cytometry and bulk RNAseq and through characterization of canonical signaling pathways and cytokine production. RESULTS: On TCR engagement, the phenotype of ex vivo CD8 T cells bearing the protective (CD226-307Gly) or the risk (CD226-307Ser) allele of CD226 was largely overlapping. However, the transcriptomic signature of CD8 T cells from the donors carrying the risk allele presented an enrichment in TCR, JAK/STAT, and IFNγ signaling. We next found that the CD226-307Ser risk allele leads to a selective increase in the phosphorylation of the mitogen-activated protein kinases extracellular signal-regulated kinases 1 and 2 (ERK1/2) associated with enhanced phosphorylation of STAT4 and increased production of IFNγ. DISCUSSION: Our data suggest that the CD226-307Ser risk variant imposes immune dysregulation by increasing the pathways related to IFNγ signaling in CD8 T cells, thereby contributing to the risk of developing chronic inflammation.


Subject(s)
Antigens, Differentiation, T-Lymphocyte , CD8-Positive T-Lymphocytes , Multiple Sclerosis , Humans , CD8-Positive T-Lymphocytes/immunology , Antigens, Differentiation, T-Lymphocyte/genetics , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Adult , Female , Male , Middle Aged
3.
Front Immunol ; 15: 1431403, 2024.
Article in English | MEDLINE | ID: mdl-39224589

ABSTRACT

Introduction: There are no reports in LATAM related to longitudinal humoral and cellular response to adenovirus based COVID-19 vaccines in people with Multiple Sclerosis (pwMS) under different disease modifying therapies (DMTs) and neutralization of the Omicron and Wuhan variants of SARS-COV-2. Methods: IgG anti- SARS-COV-2 spike titer were measured in a cohort of 101 pwMS under fingolimod, dimethyl fumarate, cladribine and antiCD20, as well as 28 healthy controls (HC) were measured 6 weeks after vaccination with 2nd dose (Sputnik V or AZD1222) and 3nd dose (homologous or heterologous schedule). Neutralizing capacity was against Omicron (BA.1) and Wuhan (D614G) variants and pseudotyped particles and Cellular response were analyzed. Results: Multivariate regression analysis showed anti-cd20 (ß= -,349, 95% CI: -3655.6 - -369.01, p=0.017) and fingolimod (ß=-,399, 95% CI: -3363.8 - -250.9, p=0.023) treatments as an independent factor associated with low antibody response (r2 adjusted=0.157). After the 2nd dose we found a correlation between total and neutralizing titers against D614G (rho=0.6; p<0.001; slope 0.8, 95%CI:0.4-1.3), with no differences between DMTs. Neutralization capacity was lower for BA.1 (slope 0.3, 95%CI:0.1-0.4). After the 3rd dose, neutralization of BA.1 improved (slope: 0.9 95%CI:0.6-1.2), without differences between DMTs. A fraction of pwMS generated anti-Spike CD4+ and CD8+ T cell response. In contrast, pwMS under antiCD20 generated CD8+TNF+IL2+ response without differences with HC, even in the absence of humoral response. The 3rd dose significantly increased the neutralization against the Omicron, as observed in the immunocompetent population. Discussion: Findings regarding humoral and cellular response are consistent with previous reports.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Immunosuppressive Agents , Multiple Sclerosis , SARS-CoV-2 , Humans , Male , Female , Immunosuppressive Agents/therapeutic use , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , SARS-CoV-2/immunology , Middle Aged , Multiple Sclerosis/immunology , Multiple Sclerosis/drug therapy , COVID-19/immunology , COVID-19/prevention & control , Adult , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Argentina , Adenoviridae/genetics , Adenoviridae/immunology , Immunity, Humoral , Spike Glycoprotein, Coronavirus/immunology
4.
Front Immunol ; 15: 1446748, 2024.
Article in English | MEDLINE | ID: mdl-39224590

ABSTRACT

Multiple sclerosis (MS) is a devastating immune-mediated disorder of the central nervous system resulting in progressive disability accumulation. As there is no cure available yet for MS, the primary therapeutic objective is to reduce relapses and to slow down disability progression as early as possible during the disease to maintain and/or improve health-related quality of life. However, optimizing treatment for people with MS (pwMS) is complex and challenging due to the many factors involved and in particular, the high degree of clinical and sub-clinical heterogeneity in disease progression among pwMS. In this paper, we discuss these many different challenges complicating treatment optimization for pwMS as well as how a shift towards a more pro-active, data-driven and personalized medicine approach could potentially improve patient outcomes for pwMS. We describe how the 'Clinical Impact through AI-assisted MS Care' (CLAIMS) project serves as a recent example of how to realize such a shift towards personalized treatment optimization for pwMS through the development of a platform that offers a holistic view of all relevant patient data and biomarkers, and then using this data to enable AI-supported prognostic modelling.


Subject(s)
Artificial Intelligence , Multiple Sclerosis , Precision Medicine , Humans , Artificial Intelligence/trends , Biomarkers , Disease Progression , Multiple Sclerosis/therapy , Multiple Sclerosis/immunology , Precision Medicine/methods , Precision Medicine/trends , Prognosis , Quality of Life
5.
Front Immunol ; 15: 1334720, 2024.
Article in English | MEDLINE | ID: mdl-39257578

ABSTRACT

Multiple sclerosis (MS) is a prototypical autoimmune disease of the central nervous system (CNS). In addition to CD4+ T cells, memory B cells are now recognized as a critical cell type in the disease. This is underlined by the fact that the best-characterized environmental risk factor for MS is the Epstein-Barr virus (EBV), which can infect and persist in memory B cells throughout life. Several studies have identified changes in anti-EBV immunity in patients with MS. Examples include elevated titers of anti-EBV nuclear antigen 1 (EBNA1) antibodies, interactions of these with the MS-associated HLA-DR15 haplotype, and molecular mimicry with MS autoantigens like myelin basic protein (MBP), anoctamin-2 (ANO2), glial cell adhesion molecule (GlialCAM), and alpha-crystallin B (CRYAB). In this study, we employ a simple in vitro assay to examine the memory B cell antibody repertoire in MS patients and healthy controls. We replicate previous serological data from MS patients demonstrating an increased secretion of anti-EBNA1380-641 IgG in cell culture supernatants, as well as a positive correlation of these levels with autoantibodies against GlialCAM262-416 and ANO21-275. For EBNA1380-641 and ANO21-275, we provide additional evidence suggesting antibody cross-reactivity between the two targets. Further, we show that two efficacious MS treatments - natalizumab (NAT) and autologous hematopoietic stem cell transplantation (aHSCT) - are associated with distinct changes in the EBNA1-directed B cell response and that these alterations can be attributed to the unique mechanisms of action of these therapies. Using an in vitro system, our study confirms MS-associated changes in the anti-EBNA1 memory B cell response, EBNA1380-641 antibody cross-reactivity with ANO21-275, and reveals treatment-associated changes in the immunoglobulin repertoire in MS.


Subject(s)
Cross Reactions , Epstein-Barr Virus Nuclear Antigens , Memory B Cells , Multiple Sclerosis , Humans , Multiple Sclerosis/immunology , Epstein-Barr Virus Nuclear Antigens/immunology , Cross Reactions/immunology , Female , Male , Adult , Memory B Cells/immunology , Herpesvirus 4, Human/immunology , Middle Aged , Antibodies, Viral/immunology , Epstein-Barr Virus Infections/immunology , B-Lymphocytes/immunology , Immunologic Memory
6.
Sci Rep ; 14(1): 21793, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39294186

ABSTRACT

Multiple sclerosis (MS) is an inflammatory demyelinating disease with heterogeneous clinical presentations and variable long-term disability accumulation. There are currently no standard criteria to accurately predict disease outcomes. In this study we investigated the cross-sectional relationship between disease phenotype and immune-modulating cytokines and chemokines in cerebrospinal fluid (CSF). We analyzed CSF from 20 DMT-naïve MS patients using Olink Proteomics' Target 96 Inflammation panel and correlated the resulting analytes with respect to (1) disease subtype, (2) patient age and sex, (3) extent of clinical disability, and (4) MRI segmental brain volumes. We found that intrathecal IL-4 correlated with higher Expanded Disability Status Scale (EDSS) scores and longer 25-foot walk times, and CD8A correlated with decreased thalamic volumes and longer 9-hole peg test times. Male sex was associated with higher FGF-19 expression, and Tumefactive MS with elevated CCL4. Several inflammatory markers were correlated with older age at the time of LP. Finally, higher intrathecal IL-33 correlated with increased MS lesion burden and multi-compartment brain atrophy. This study confirms immune heterogeneity underlying CSF profiles in MS, but also identifies several inflammatory protein biomarkers that may be of use for predicting clinical outcomes in future algorithms.


Subject(s)
Biomarkers , Multiple Sclerosis , Proteomics , Humans , Male , Female , Biomarkers/cerebrospinal fluid , Adult , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/immunology , Proteomics/methods , Middle Aged , Phenotype , Cytokines/cerebrospinal fluid , Magnetic Resonance Imaging , Cross-Sectional Studies
7.
Gut Microbes ; 16(1): 2402547, 2024.
Article in English | MEDLINE | ID: mdl-39287045

ABSTRACT

Multiple sclerosis (MS) is an autoimmune disease characterized by inflammatory demyelination occurring in the central nervous system (CNS). Inulin is a common prebiotic that can improve metabolic disorders by modulating the gut microbiota. However, its capacity to affect CNS autoimmunity is poorly recognized. Experimental autoimmune encephalomyelitis (EAE) is a classical mouse model of MS. Herein, we found that oral administration of inulin ameliorated the severity EAE in mice, accompanied by reductions in inflammatory cell infiltration and demyelination in the CNS. These reductions were associated with decreased proportion and numbers of Th17 cells in brain and spleen. Consistent with the findings, the serum concentrations of IL-17, IL-6, and TNF-α were reduced in inulin treated EAE mice. Moreover, the proliferation of auto-reactive lymphocytes, against MOG35-55 antigen, was attenuated ex vivo. Mechanistically, inulin treatment altered the composition of gut microbiota. It increased Lactobacillus and Dubosiella whereas decreased g_Prevotellaceae_NK3B31_group at the genus level, alongside with elevated concentration of butyric acid in fecal content and serum. In vitro, butyrate, but not inulin, could inhibit the activation of MOG35-55 stimulated lymphocytes. Furthermore, fecal microbiota transplantation assay confirmed that fecal contents of inulin-treated normal mice had an ameliorative effect on EAE mice. In contrast, antibiotic cocktail (ABX) treatment diminished the therapeutic effect of inulin in EAE mice as well as the reduction of Th17 cells, while supplementation with Lactobacillus reuteri restored the amelioration effect. These results confirmed that the attenuation of inulin on Th17 cells and inflammatory demyelination in EAE mice was dependent on its modulation on gut microbiota and metabolites. Our findings provide a potential therapeutic regimen for prebiotic inulin supplementation in patients with multiple sclerosis.


Subject(s)
Autoimmunity , Encephalomyelitis, Autoimmune, Experimental , Fatty Acids, Volatile , Gastrointestinal Microbiome , Inulin , Mice, Inbred C57BL , Multiple Sclerosis , Prebiotics , Th17 Cells , Animals , Gastrointestinal Microbiome/drug effects , Inulin/administration & dosage , Inulin/pharmacology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/microbiology , Th17 Cells/immunology , Mice , Prebiotics/administration & dosage , Female , Fatty Acids, Volatile/metabolism , Autoimmunity/drug effects , Multiple Sclerosis/immunology , Multiple Sclerosis/drug therapy , Multiple Sclerosis/microbiology , Central Nervous System/immunology , Bacteria/classification , Bacteria/isolation & purification
8.
Front Immunol ; 15: 1416074, 2024.
Article in English | MEDLINE | ID: mdl-39086476

ABSTRACT

Introduction: Progressive Multifocal Leukoencephalopathy (PML) is a rare and deadly demyelinating disease caused by JC virus (JCV) replication in the central nervous system. PML occurs exclusively in patients with severe underlying immune deficiencies, including AIDS and hematological malignancies. PML has also emerged as a significant threat to patients on potent new immunosuppressive biologics, including natalizumab in multiple sclerosis. Methods: Here, we developed an IFN-γ release assay (IGRA) that mainly detects JCV-specific effector memory T cells and effectors T cells in the blood. Results: This assay was frequently positive in patients with active PML (with a positive JCV PCR in CSF) of various underlying immunosuppression causes (84% sensitivity). Only 3% of healthy donors had a positive response (97% specificity). The frequency of positivity also increased in multiple sclerosis patients according to the time on natalizumab (up to 36% in patients treated for more than 48 months, who are considered at a higher risk of PML). Discussion: The results show this assay's frequent or increased positivity in patients with PML or an increased risk of PML, respectively. The assay may help to stratify the risk of PML.


Subject(s)
Interferon-gamma , JC Virus , Leukoencephalopathy, Progressive Multifocal , Memory T Cells , Humans , Leukoencephalopathy, Progressive Multifocal/immunology , Leukoencephalopathy, Progressive Multifocal/diagnosis , Leukoencephalopathy, Progressive Multifocal/etiology , Male , JC Virus/immunology , Female , Middle Aged , Adult , Memory T Cells/immunology , Memory T Cells/metabolism , Natalizumab/therapeutic use , Aged , Multiple Sclerosis/immunology , Multiple Sclerosis/drug therapy
9.
Gut Microbes ; 16(1): 2387794, 2024.
Article in English | MEDLINE | ID: mdl-39114974

ABSTRACT

Multiple sclerosis (MS) is a chronic and progressive autoimmune disease of the central nervous system (CNS), with both genetic and environmental factors contributing to the pathobiology of the disease. While human leukocyte antigen (HLA) genes have emerged as the strongest genetic factor, consensus on environmental risk factors are lacking. Recently, trillions of microbes residing in our gut (microbiome) have emerged as a potential environmental factor linked with the pathobiology of MS as PwMS show gut microbial dysbiosis (altered gut microbiome). Thus, there has been a strong emphasis on understanding the factors (host and environmental) regulating the composition of the gut microbiota and the mechanism(s) through which gut microbes contribute to MS disease, especially through immune system modulation. A better understanding of these interactions will help harness the enormous potential of the gut microbiota as a therapeutic approach to treating MS.


Subject(s)
Dysbiosis , Gastrointestinal Microbiome , Multiple Sclerosis , Multiple Sclerosis/microbiology , Multiple Sclerosis/immunology , Humans , Dysbiosis/microbiology , Animals , Bacteria/genetics , Bacteria/classification , Bacteria/metabolism
10.
Front Immunol ; 15: 1412668, 2024.
Article in English | MEDLINE | ID: mdl-39086478

ABSTRACT

Rituximab has been used to treat MS patients in Iceland for over a decade. However, long-term effect of rituximab on leukocyte populations has not yet been elucidated. By retrospective analysis of flow cytometric data from 349 patients visiting the neurological ward at The National University Hospital of Iceland from 2012 to 2023 for rituximab treatment, the long-term effect of rituximab and whether the effect was dose dependent (1000mg vs 500mg) was evaluated. No difference was detected in efficacy of B cell depletion in patients treated with 500mg as an initial dose of rituximab when compared to 1000mg. Long-term use of rituximab led to an increase in T cell count (p=0,0015) in patients receiving 3-8 doses of rituximab (1.5-8 years of treatment). The increase occurred in both CD4+ (p=0,0028) and CD8+ T cells (p=0,0015) and led to a decrease in the CD4/CD8 ratio (p=0,004). The most notable difference lies in reshaping the balance between näive and effector CD8+ T cells. The clinical implications of long-term treatment with rituximab and its effect on the T cell pool needs to be explored further. Since no difference in B cell depletion was detected between the two patient groups, 1000mg as an initial dose might be excessive, suggesting a personalized dosing regimen might have therapeutic and financial advantages.


Subject(s)
Multiple Sclerosis , Rituximab , Humans , Rituximab/administration & dosage , Rituximab/therapeutic use , Rituximab/adverse effects , Male , Female , Adult , Middle Aged , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Retrospective Studies , Lymphocyte Count , Immunologic Factors/administration & dosage , Immunologic Factors/therapeutic use , Aged , CD4-CD8 Ratio , B-Lymphocytes/immunology , B-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/drug effects
11.
Front Immunol ; 15: 1436717, 2024.
Article in English | MEDLINE | ID: mdl-39108272

ABSTRACT

Neurological disorders, including multiple sclerosis (MS) and amyotrophic lateral sclerosis (ALS), may be associated with alterations in blood cell composition and phenotype. Here, we focused our attention on circulating mucosal-associated invariant T (MAIT) cells, a CD8+ T cell memory population expressing the invariant Vα7.2 region in the T cell receptor and high surface levels of the CD161 marker. Transcriptomics data relative to peripheral blood mononuclear cells (PBMC) highlighted downregulation of CD161 and other MAIT-associated markers in progressive MS and not relapsing remitting (RR)-MS when gene expressions relative to each disease course were compared to those from healthy controls. Multiparametric flow cytometry of freshly isolated PBMC samples from untreated RR-MS, primary or secondary progressive MS (PP- or SP-MS), ALS and age- and sex-matched healthy controls revealed specific loss of circulating CD8+ MAIT cells in PP-MS and no other MS courses or another neurological disorder such as ALS. Overall, these observations point to the existence of immunological changes in blood specific for the primary progressive course of MS that may support clinical definition of disease.


Subject(s)
Amyotrophic Lateral Sclerosis , Mucosal-Associated Invariant T Cells , Humans , Amyotrophic Lateral Sclerosis/immunology , Amyotrophic Lateral Sclerosis/blood , Mucosal-Associated Invariant T Cells/immunology , Mucosal-Associated Invariant T Cells/metabolism , Male , Middle Aged , Female , Adult , Aged , Multiple Sclerosis/immunology , Multiple Sclerosis/blood , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Biomarkers , Flow Cytometry
12.
Handb Clin Neurol ; 202: 55-74, 2024.
Article in English | MEDLINE | ID: mdl-39111918

ABSTRACT

Hematopoietic stem cell transplantation (HSCT) is a multistep procedure aimed at eradicating the immune system and replacing it with a new one reconstituted from hematopoietic stem cells which in autologous HSCT (AHSCT) have previously been harvested from the same individual. Over the last two decades, AHSCT has been developed as a treatment option for people affected by aggressive multiple sclerosis (MS), and it exerts a long-standing effect on new inflammation-driven disease activity. The rationale for the use of AHSCT in MS will be discussed, starting from the first observations on experimental models. The mechanisms and kinetics of repopulation (i.e., quantitative recovery) and reconstitution (i.e., qualitative changes) of the immune cell populations will be explored, focusing on immune reconstitution of the T and B cells compartments and briefly covering changes in the innate immune system. Finally, potential immunologic markers of response to treatment will be reviewed. Insights into the supposed mechanism(s) of action of AHSCT will be provided, discussing the leading hypothesis of the "rebuilding" of a newly tolerant immune system, and examining the apparent paradox of the long-standing control of disease activity despite a relatively short-term immunosuppressive effect of the procedure.


Subject(s)
Hematopoietic Stem Cell Transplantation , Immune Reconstitution , Multiple Sclerosis , Transplantation, Autologous , Humans , Hematopoietic Stem Cell Transplantation/methods , Transplantation, Autologous/methods , Multiple Sclerosis/immunology , Multiple Sclerosis/therapy , Animals
13.
Handb Clin Neurol ; 202: 7-21, 2024.
Article in English | MEDLINE | ID: mdl-39111919

ABSTRACT

Multiple sclerosis (MS) is an inflammatory disease of the central nervous system (CNS), which can clinically manifest as attacks of neurologic disability and new lesion formation, and a progression of sustained neurologic disability over time. In MS, activated B and T cells are recruited from outside the CNS, and contribute to inflammation, demyelination, and tissue damage inside the brain parenchyma. In the last decades, the treatment of MS has improved by the introduction of several disease-modifying therapies (DMTs). These drugs target generic mechanisms of lymphocyte activation and recruitment or deplete lymphocyte fractions from the circulation. This contributes to a suppression of relapses and new MS lesion formation on MRI. However, the impact on disability progression without relapses is much more variable. In addition, risk mitigation strategies are warranted to control for unwanted side effects of the attenuated immune competence induced by DMTs. In this chapter, we argue that an understanding of the impact of these DMTs on B and T cells both outside and inside the CNS can help to understand the benefits of these therapies but can also help to identify the challenges and opportunities that lie ahead for future MS therapies.


Subject(s)
B-Lymphocytes , Multiple Sclerosis , T-Lymphocytes , Humans , Multiple Sclerosis/immunology , Multiple Sclerosis/drug therapy , Multiple Sclerosis/pathology , Multiple Sclerosis/therapy , B-Lymphocytes/immunology , B-Lymphocytes/drug effects , T-Lymphocytes/immunology , Immunologic Factors/therapeutic use , Animals
14.
BMC Neurol ; 24(1): 291, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39164636

ABSTRACT

BACKGROUND: To investigate the safety (adverse events [AEs] and post-vaccination multiple sclerosis [MS] activity within 6 weeks), clinical efficacy (protection against coronavirus disease 2019 [COVID-19]), and vaccine-induced humoral immunogenicity (SARS-CoV-2 neutralizing antibody, anti-nucleocapsid IgG, and anti-spike IgG) of the Sinopharm (BBIBP-CorV) vaccine among people with MS (PwMS) receiving different disease-modifying therapies (DMTs). METHODS: This prospective cohort study was conducted between November 2021 and May 2022. PwMS were followed for six months after the 2nd dose of vaccination. Antibody responses were measured 2-16 weeks after the 2nd dose injection. Multivariate logistic regression was employed to assess the impact of each DMT on dichotomous antibody responses, adjusting for age, sex, MS phenotype, expanded disability status scale, disease duration, and vaccination-antibody titration interval. RESULTS: Among the 261 screened PwMS, 209 (aged 38.23 ± 9.73 years, female: 70.8%; relapsing-remitting MS: 80.4%) were included. The frequencies of experiencing non-serious AEs and post-vaccination MS activity were 66.0% and 4.8%, respectively. Breakthrough COVID-19 infection was observed in 14.8% of the PwMS. A subcohort of 125 PwMS was assessed for antibody responses. Positive neutralizing antibodies, anti-nucleocapsid IgG, and anti-spike IgG were detected in 36.8%, 35.2%, and 52.0% of the PwMS, respectively. Multivariate regression indicated a 96% (OR: 0.04 [95% CI: 0.00, 0.51], P = 0.013), 93% (OR: 0.07 [0.01, 0.64], P = 0.019), and 89% (OR: 0.11 [0.01, 0.96], P = 0.045) reduced odds of positive neutralizing antibody, anti-nucleocapsid IgG, and anti-spike IgG, respectively, among fingolimod-receivers. Additionally, anti-CD20s-receivers had 88% (OR: 0.12 [0.02, 0.85], P = 0.034) lower odds of being positive for anti-nucleocapsid IgG. CONCLUSIONS: BBIBP-CorV appeared to be well tolerated in PwMS, with promising clinical efficacy. However, a suboptimal humoral response was observed in PwMS receiving fingolimod and anti-CD20s. Future research should investigate the relationship between humoral responses and the frequency and severity of COVID-19 infection across various DMTs.


Subject(s)
Antibodies, Neutralizing , COVID-19 Vaccines , COVID-19 , Multiple Sclerosis , Vaccines, Inactivated , Adult , Female , Humans , Male , Middle Aged , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Cohort Studies , COVID-19/prevention & control , COVID-19/immunology , COVID-19/epidemiology , COVID-19 Vaccines/adverse effects , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Immunogenicity, Vaccine/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Multiple Sclerosis/immunology , Multiple Sclerosis/drug therapy , Prospective Studies , SARS-CoV-2/immunology , Treatment Outcome , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/adverse effects , Vaccines, Inactivated/immunology
15.
Neurol Neuroimmunol Neuroinflamm ; 11(5): e200293, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39133885

ABSTRACT

OBJECTIVES: In myelin oligodendrocyte glycoprotein IgG-associated disease (MOGAD) and aquaporin-4 IgG+ neuromyelitis optica spectrum disorder (AQP4+NMOSD), the autoantibodies are mainly composed of IgG1, and complement-dependent cytotoxicity is a primary pathomechanism in AQP4+NMOSD. We aimed to evaluate the CSF complement activation in MOGAD. METHODS: CSF-C3a, CSF-C4a, CSF-C5a, and CSF-C5b-9 levels during the acute phase before treatment in patients with MOGAD (n = 12), AQP4+NMOSD (n = 11), multiple sclerosis (MS) (n = 5), and noninflammatory neurologic disease (n = 2) were measured. RESULTS: CSF-C3a and CSF-C5a levels were significantly higher in MOGAD (mean ± SD, 5,629 ± 1,079 pg/mL and 2,930 ± 435.8 pg/mL) and AQP4+NMOSD (6,017 ± 3,937 pg/mL and 2,544 ± 1,231 pg/mL) than in MS (1,507 ± 1,286 pg/mL and 193.8 ± 0.53 pg/mL). CSF-C3a, CSF-C4a, and CSF-C5a did not differ between MOGAD and AQP4+NMOSD while CSF-C5b-9 (membrane attack complex, MAC) levels were significantly lower in MOGAD (17.4 ± 27.9 ng/mL) than in AQP4+NMOSD (62.5 ± 45.1 ng/mL, p = 0.0019). Patients with MOGAD with severer attacks (Expanded Disability Status Scale [EDSS] ≥ 3.5) had higher C5b-9 levels (34.0 ± 38.4 ng/m) than those with milder attacks (EDSS ≤3.0, 0.9 ± 0.7 ng/mL, p = 0.044). DISCUSSION: The complement pathway is activated in both MOGAD and AQP4+NMOSD, but MAC formation is lower in MOGAD, particularly in those with mild attacks, than in AQP4+NMOSD. These findings may have pathogenetic and therapeutic implications in MOGAD.


Subject(s)
Aquaporin 4 , Complement Activation , Immunoglobulin G , Myelin-Oligodendrocyte Glycoprotein , Neuromyelitis Optica , Humans , Neuromyelitis Optica/cerebrospinal fluid , Neuromyelitis Optica/immunology , Neuromyelitis Optica/blood , Aquaporin 4/immunology , Male , Female , Middle Aged , Adult , Myelin-Oligodendrocyte Glycoprotein/immunology , Immunoglobulin G/cerebrospinal fluid , Immunoglobulin G/blood , Immunoglobulin G/immunology , Autoantibodies/cerebrospinal fluid , Autoantibodies/blood , Aged , Complement C5a/cerebrospinal fluid , Complement C5a/metabolism , Complement C5a/immunology , Young Adult , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/immunology , Complement C3a/metabolism , Complement C3a/cerebrospinal fluid , Complement C3a/immunology , Complement Membrane Attack Complex/metabolism , Complement Membrane Attack Complex/cerebrospinal fluid , Complement Membrane Attack Complex/immunology
16.
Int J Mol Sci ; 25(16)2024 Aug 18.
Article in English | MEDLINE | ID: mdl-39201672

ABSTRACT

Multiple sclerosis (MS) is a disease of the central nervous system (CNS) characterized by inflammation and autoimmune responses. This review explores the participation of T cells, particularly certain CD3+CD20+ T cells, in the clinical manifestations of MS and highlights their presence in diagnosed patients. These T cells show aberrant expression of CD20, normally considered a B-cell marker. In this review, relevant journal articles available in PubMed and CINAHL were identified by employing diverse search terms, such as MS, CD3+CD20+ T cells, the incidence and significance of CD3+CD20+ T cells in MS patients, and the impact of rituximab treatment. The search was limited to articles published in the ten-year period from 2014 to 2024. The results of this review suggest that most scholars agree on the presence of CD3+CD20+ T cells in cerebrospinal fluid. Emerging concepts relate to the fundamental role of CD20-expressing T cells in determining the target and efficacy of MS therapeutics and the presence of T cells in the cerebrospinal fluid of MS patients. The results clearly show that CD20+ T cells indicate disease chronicity and high disease activity.


Subject(s)
Antigens, CD20 , CD3 Complex , Multiple Sclerosis , Rituximab , T-Lymphocytes , Humans , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Antigens, CD20/metabolism , Antigens, CD20/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , CD3 Complex/metabolism , CD3 Complex/immunology , Rituximab/therapeutic use
17.
J Neuroimmunol ; 394: 578421, 2024 09 15.
Article in English | MEDLINE | ID: mdl-39088907

ABSTRACT

Niacin was found in the lysolecithin model of multiple sclerosis (MS) to promote the phagocytic clearance of debris and enhance remyelination. Lysolecithin lesions have prominent microglia/macrophages but lack lymphocytes that populate plaques of MS or its experimental autoimmune encephalomyelitis (EAE) model. Thus, the current study assessed the efficacy of niacin in EAE. We found that niacin inconsistently affects EAE clinical score, and largely does not ameliorate neuropathology. In culture, niacin enhances phagocytosis by macrophages, but does not reduce T cell proliferation. We suggest that studies of niacin for potential remyelination in MS should include a therapeutic that targets adaptive immunity.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Mice, Inbred C57BL , Multiple Sclerosis , Niacin , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Animals , Niacin/therapeutic use , Female , Mice , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Phagocytosis/drug effects , Macrophages/drug effects , Macrophages/immunology , Cells, Cultured , Disease Models, Animal , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
18.
J Neuroimmunol ; 394: 578408, 2024 09 15.
Article in English | MEDLINE | ID: mdl-39098102

ABSTRACT

Epstein-Barr virus (EBV) is deemed a necessary, yet insufficient factor in the development of multiple sclerosis (MS). In this study, myelin basic protein-specific transgenic T cell receptor mice were infected with murid gammaherpesvirus 68 virus (MHV68), an EBV-like virus that infects mice, resulting in the onset neurological deficits at a significantly higher frequency than influenza or mock-infected mice. MHV68 infected mice exhibited signs including optic neuritis and ataxia which are frequently observed in MS patients but not in experimental autoimmune encephalomyelitis mice. MHV68-infected mice exhibited increased focal immune cell infiltration in the central nervous system. Single cell RNA sequencing identified the emergence of a population of B cells that express genes associated with antigen presentation and costimulation, indicating that gammaherpesvirus infection drives a distinct, pro-inflammatory transcriptional program in B cells that may promote autoreactive T cell responses in MS.


Subject(s)
B-Lymphocytes , Disease Models, Animal , Mice, Transgenic , Multiple Sclerosis , Animals , Multiple Sclerosis/immunology , Multiple Sclerosis/virology , Mice , B-Lymphocytes/immunology , Mice, Inbred C57BL , Rhadinovirus/immunology , Female , Encephalomyelitis, Autoimmune, Experimental/immunology
19.
Mult Scler Relat Disord ; 90: 105813, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39154595

ABSTRACT

Few data are available regarding vaccine induced SARS-CoV-2 specific T cell responses over time and after booster doses in multiple sclerosis (MS) patients on different disease modifying treatments. We measured SARS-CoV-2 specific CD4+ T cell responses in 72 samples collected from 36 MS patients. The percentage of CD4+ CTVlow CD25+ ICOS+ T cells after stimulation with Spike Recombinant Protein was 29.9 (17.0-43.6) on teriflunomide, 32.4 (11.9-42.5) on ocrelizumab, but much lower (0.6 [0.3-5.9]) on sphingosine-1-phospate receptor modulators (ß = -26.35, p = 0.003). SARS-CoV-2 specific T cells were mainly of Th1 type and stable over time and after booster vaccine doses. mRNA vaccines elicit strong and persistent CD4+ T cell responses against SARS-CoV-2 in MS patients on anti-CD20 and teriflunomide, but not in those on sphingosine-1-phospate receptor modulators.


Subject(s)
CD4-Positive T-Lymphocytes , COVID-19 Vaccines , COVID-19 , Multiple Sclerosis , Nitriles , SARS-CoV-2 , Humans , Male , Female , Adult , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/pharmacology , Middle Aged , COVID-19/prevention & control , COVID-19/immunology , SARS-CoV-2/immunology , Multiple Sclerosis/immunology , Multiple Sclerosis/drug therapy , CD4-Positive T-Lymphocytes/immunology , Nitriles/pharmacology , Toluidines/pharmacology , Toluidines/therapeutic use , Crotonates/pharmacology , Crotonates/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Hydroxybutyrates/pharmacology , mRNA Vaccines , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/therapeutic use , Immunologic Factors/pharmacology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
20.
Mult Scler Relat Disord ; 90: 105829, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39182455

ABSTRACT

BACKGROUND: The previous Japanese clinical practice guidelines for multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD) were published in 2017. Recently, for the first time in 6 years, the MS and NMOSD guideline development committee revised the Japanese guidelines for MS, NMOSD, and myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD). METHODS: The committee utilized the Grading of Recommendations Assessment, Development, and Evaluation system based on the "Minds Handbook for Clinical Practice Guideline Development 2020 Ver. 3.0″ with a focus on clinical questions (CQs). The committee also discussed clinical issues other than CQs, categorizing them as a question-and-answer (Q&A) section, including "issues on which experts' opinions agree to a certain extent" and "issues that are important but not included in the CQ". RESULTS: The committee identified 3, 1, and 1 key CQs related to MS, NMOSD, and MOGAD, respectively, and presented recommendations. A Q&A session regarding disease-modifying therapies and relapse prevention therapies for MS, NMOSD, and MOGAD was conducted. The revised guidelines were published in September 2023. CONCLUSIONS: The Japanese guidelines for clinical practice on MS, NMOSD, and MOGAD were updated. Treatment strategies for MS, NMOSD, and MOGAD are changing, and these updated guidelines may assist with treatment decisions for these diseases in clinical practice.


Subject(s)
Multiple Sclerosis , Myelin-Oligodendrocyte Glycoprotein , Neuromyelitis Optica , Humans , Neuromyelitis Optica/therapy , Neuromyelitis Optica/immunology , Neuromyelitis Optica/diagnosis , Neuromyelitis Optica/drug therapy , Myelin-Oligodendrocyte Glycoprotein/immunology , Multiple Sclerosis/therapy , Multiple Sclerosis/immunology , Multiple Sclerosis/diagnosis , Japan , Practice Guidelines as Topic/standards , Autoantibodies/blood
SELECTION OF CITATIONS
SEARCH DETAIL