Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
2.
Mult Scler J Exp Transl Clin ; 7(2): 20552173211004634, 2021.
Article in English | MEDLINE | ID: mdl-33948221

ABSTRACT

BACKGROUND: Optimizing multiple sclerosis treatment warrants understanding of changes in physical, mental, and social health. OBJECTIVE: To assess the impact of natalizumab on Quality of Life in Neurological Disorders (Neuro-QoL) scores. METHODS: Annualized change in T-scores and likelihood of ≥5-point improvement over baseline were calculated for each Neuro-QoL domain after natalizumab initiation. Comparisons with ocrelizumab-treated patients were conducted after propensity score weighting and adjustment for relevant co-medications, year, and drug-year interaction. RESULTS: Among 164 natalizumab patients analyzed, 8 of 12 Neuro-QoL domains improved significantly, with greater improvement in patients with abnormal baseline Neuro-QoL. In the subgroup comparison of natalizumab-treated (n = 145) and ocrelizumab-treated (n = 520) patients, significant improvement occurred in 9 of 12 and 4 of 12 domains, respectively. The difference between groups was statistically significant for positive affect and well-being (p = 0.02), sleep (p = 0.003), and satisfaction with social roles and activities (SRA) (p = 0.03) in the overall population and for emotional and behavioral dyscontrol (p = 0.01), participation in SRA (p = 0.0001), and satisfaction with SRA (p = 0.02) in patients with abnormal baseline Neuro-QoL. CONCLUSIONS: Natalizumab can produce clinically meaningful improvements in mental and social health. Such improvements are unlikely to be primarily driven by expectation bias, as their magnitude exceeded improvements with another high-efficacy therapy, ocrelizumab.

3.
Adv Ther ; 38(7): 3724-3742, 2021 07.
Article in English | MEDLINE | ID: mdl-34014549

ABSTRACT

INTRODUCTION: STRIVE was a 4-year, multicenter, observational, open-label, single-arm study of natalizumab treatment in anti-JC virus antibody-negative (JCV-negative) relapsing-remitting multiple sclerosis (RRMS) patients with disease duration ≤ 3 years. The objective of STRIVE was to examine no evidence of disease activity (NEDA) status and predictors of NEDA in natalizumab-treated patients with early RRMS. METHODS: Proportions of patients with NEDA were evaluated along with baseline predictors of NEDA, annualized relapse rate, 24-week confirmed disability worsening (CDW), magnetic resonance imaging assessments (T2 and gadolinium-enhancing lesions), and serious adverse events. RESULTS: In years 1 and 2, 56.1% (95% confidence interval [CI] 48.7-63.4%) and 73.6% (95% CI 66.2-80.2%) of patients (intent-to-treat population [N = 222]), respectively, achieved NEDA. In years 3 and 4, 84.6% (95% CI 78.0-89.9%) and 91.9% (95% CI 86.4-95.8%) of patients, respectively, achieved Clinical NEDA (no relapses or 24-week CDW). Baseline predictors of NEDA in year 4 were Expanded Disability Status Scale score ≤ 2.0 (odds ratio [OR] = 3.85 [95% CI 1.54-9.63]; p = 0.004) and T2 lesion volume > 4 cc (OR = 0.39 [95% CI 0.15-0.98]; p = 0.046), with the latter also predicting Clinical NEDA in year 4 (OR = 0.21 [95% CI 0.05-0.92]; p = 0.038). The cumulative probability of CDW at year 4 was 19.3%. Serious adverse events were reported in 11.3% of patients. CONCLUSION: These results support the long-term safety and effectiveness of natalizumab. Baseline predictors of NEDA help to inform benefit-risk assessments of natalizumab treatment in JCV-negative patients with early RRMS. TRIAL REGISTRATION: ClinicalTrials.gov identifier NCT01485003.


Subject(s)
Multiple Sclerosis, Relapsing-Remitting , Multiple Sclerosis , Humans , Immunologic Factors/adverse effects , Magnetic Resonance Imaging , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Natalizumab/adverse effects , Treatment Outcome
4.
Int J MS Care ; 18(5): 230-238, 2016.
Article in English | MEDLINE | ID: mdl-27803638

ABSTRACT

Background: Physicians must weigh the benefits against the risk of progressive multifocal leukoencephalopathy (PML) in patients treated with natalizumab, especially beyond 2 years. However, disability progression associated with switching therapies versus continuing natalizumab therapy after 2 years has not been fully evaluated. Methods: In this retrospective analysis using the NARCOMS Registry, disability progression (Patient-Determined Disease Steps [PDDS] scale) and physical health-related quality of life (HRQOL) worsening (12-item Short Form Health Status Survey Physical Component Score [SF-12 PCS]) were compared between participants switching to fingolimod (n = 50) or interferon beta (IFNß)/glatiramer acetate (GA) (n = 71) therapy and those continuing natalizumab (n = 406) after 2 years or more of treatment (median follow-up: natalizumab, 4 years; fingolimod, 4.5 years; IFNß/GA, 5 years). Results: Participants continuing to take natalizumab had less disability progression (mean PDDS change: natalizumab, 0.3; fingolimod, 0.6; IFNß/GA, 0.7; P = .0036), were less likely to report disability progression (proportion with PDDS increase: natalizumab, 31%; fingolimod, 46%; IFNß/GA, 42%; P = .0296), and had less worsening in physical HRQOL (mean SF-12 PCS change: natalizumab, -1.4; fingolimod, -2.8; IFNß/GA, -4.6; P = .0476) than those switching treatment. Conclusions: Although all medication groups exhibited some level of worsening, switching from natalizumab treatment after 2 years was associated with increased disability progression and worsening physical HRQOL. The risk of disability progression from disease activity and the risk of PML should be considered when making natalizumab treatment decisions.

6.
J Neurol ; 262(2): 326-36, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25381458

ABSTRACT

The objective of this study is to characterize the timing and extent of radiologic MS disease recurrence during the 24-week natalizumab treatment interruption period in RESTORE. RESTORE was a randomized, partially placebo-controlled exploratory study. Natalizumab-treated patients with no gadolinium-enhancing (Gd+) lesions at screening (n = 175) were randomized 1:1:2 to continue natalizumab (n = 45), switch to placebo (n = 42), or switch to other therapies (n = 88) for 24 weeks. MRI assessments were performed every 4 weeks. Predictors of increased numbers of Gd+ lesions during natalizumab treatment interruption were evaluated. The numbers of Gd+ lesions were compared with retrospectively collected pre-natalizumab MRI reports and data from placebo-treated patients from two historical randomized clinical trials. Gd+ lesions were detected in 0 % (0/45) of natalizumab patients, 61 % (25/41) of placebo patients, and 48 % (39/81) of other-therapies patients during the randomized treatment period. Gd+ lesions were detected starting at week 12; most were observed at week 16 or later. Thirteen percent (14/107) of patients had >5 Gd+ lesions on ≥1 (of 6) scans during the randomized treatment period versus 7 % (7/107) of patients pre-natalizumab (based on medical record of a single scan). Younger patients and those with more Gd+ lesions pre-natalizumab were more likely to have increased MRI activity. Distribution of total and persistent Gd+ lesions in RESTORE patients was similar to placebo-treated historical control patients. In most patients, recurring radiological disease activity during natalizumab interruption did not exceed pre-natalizumab levels or levels seen in historical control patients.


Subject(s)
Multiple Sclerosis, Relapsing-Remitting/diagnostic imaging , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Natalizumab/administration & dosage , Adult , Female , Humans , Magnetic Resonance Imaging , Multiple Sclerosis, Relapsing-Remitting/pathology , Radiography , Recurrence
7.
Ann Neurol ; 76(6): 802-12, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25273271

ABSTRACT

OBJECTIVE: The increased risk of progressive multifocal leukoencephalopathy (PML) with natalizumab treatment is associated with the presence of anti-JC virus (JCV) antibodies. We analyzed whether anti-JCV antibody levels, measured as index, may further define PML risk in seropositive patients. METHODS: The association between serum or plasma anti-JCV antibody levels and PML risk was examined in anti-JCV antibody-positive multiple sclerosis (MS) patients from natalizumab clinical studies and postmarketing sources. For PML and non-PML patients, the probabilities of having an index below and above a range of anti-JCV antibody index thresholds were calculated using all available data and applied to the PML risk stratification algorithm. Longitudinal stability of anti-JCV antibody index was also evaluated. RESULTS: Anti-JCV antibody index data were available for serum/plasma samples collected >6 months prior to PML diagnosis from 71 natalizumab-treated PML patients and 2,522 non-PML anti-JCV antibody-positive patients. In patients with no prior immunosuppressant use, anti-JCV antibody index distribution was significantly higher in PML patients than in non-PML patients (p < 0.0001). Among patients who were anti-JCV antibody negative at baseline in the AFFIRM and STRATIFY-1 trials, 97% remained consistently negative or below an index threshold of 1.5 over 18 months. Retrospective analyses of pre-PML samples collected longitudinally from PML patients displayed sustained higher anti-JCV antibody index over time. INTERPRETATION: Anti-JCV antibody levels in serum/plasma, measured as index, may differentiate PML risk in anti-JCV antibody-positive MS patients with no prior immunosuppressant use. Continued evaluation of anti-JCV antibody index and PML risk is warranted.


Subject(s)
Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Viral/blood , JC Virus/metabolism , Leukoencephalopathy, Progressive Multifocal/blood , Leukoencephalopathy, Progressive Multifocal/diagnosis , Biomarkers/blood , Humans , Leukoencephalopathy, Progressive Multifocal/chemically induced , Longitudinal Studies , Natalizumab , Risk Factors
8.
Mult Scler ; 20(5): 610-6, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23999609

ABSTRACT

BACKGROUND: Sexual dysfunction is a prevalent symptom in multiple sclerosis (MS) that may affect patients' health-related quality of life (HrQoL). OBJECTIVE: The objective of this paper is to examine the impact of sexual dysfunction on HrQoL in a large national sample using The Multiple Sclerosis Intimacy and Sexuality Questionnaire-19 (MSISQ-19). METHODS: Participants were recruited from a large MS registry, the North American Research Committee on Multiple Sclerosis (NARCOMS) Registry. Participants self-reported demographic information and completed the Patient Determined Disease Steps (PDDS), MSISQ-19, and the Short Form-12 (SF-12). RESULTS: The study population included 6183 persons (mean age: 50.6, SD = 9.6; 74.7% female, 42.3% currently employed). Using multivariate hierarchical regression analyses, all variables excluding gender predicted both the physical component summary (PCS-12) and the mental component summary (MCS-12) of the SF-12. Scores on the MSISQ-19 uniquely accounted for 3% of the variance in PCS-12 scores while disability level, as measured by PDDS, accounted for 31% of the variance. Conversely, MSISQ-19 scores uniquely accounted for 13% of the variance in MCS-12 scores, whereas disability level accounted for less than 1% of the variance. CONCLUSION: In patients with MS, sexual dysfunction has a much larger detrimental impact on the mental health aspects of HrQoL than severity of physical disability.


Subject(s)
Mental Health , Multiple Sclerosis/epidemiology , Quality of Life , Sexual Dysfunction, Physiological/epidemiology , Sexual Dysfunctions, Psychological/epidemiology , Adult , Disability Evaluation , Female , Health Surveys , Humans , Linear Models , Male , Middle Aged , Multiple Sclerosis/diagnosis , Multiple Sclerosis/physiopathology , Multiple Sclerosis/psychology , Multivariate Analysis , North America , Registries , Risk Factors , Severity of Illness Index , Sexual Dysfunction, Physiological/diagnosis , Sexual Dysfunction, Physiological/physiopathology , Sexual Dysfunction, Physiological/psychology , Sexual Dysfunctions, Psychological/diagnosis , Sexual Dysfunctions, Psychological/physiopathology , Sexual Dysfunctions, Psychological/psychology , Surveys and Questionnaires
9.
Mult Scler ; 19(9): 1197-203, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23369892

ABSTRACT

BACKGROUND: Sexual dysfunction is common in multiple sclerosis (MS) but reliable and valid measurement in this population is needed. OBJECTIVE: The objective of this research is to re-validate the Multiple Sclerosis Intimacy and Sexuality Questionnaire-19 in a large US sample. METHODS: A total of 6300 MS patients from the NARCOMS registry completed the MSISQ-19. Unforced principal component analysis utilizing oblique rotation with Kaiser Normalization validated its construct validity. RESULTS: The scree plot supported a three-component solution, with 63% of total variance explained. The components mirrored the original validation study measuring primary, secondary, and tertiary sexual dysfunction. PCA suggested the scale could be shortened to 15 items, which were found to apply equally well to males and females (with one primary item specific for each sex). The components were moderately intercorrelated (Pearson rs ranged from 0.5 to 0.67). The secondary subscale correlated most highly with self-reported disability (r (6081) = 0.44, p < 0.001), whereas the tertiary subscale correlated most highly with psychological distress (r (5992) = -.37, p < 0.001). Cronbach's alpha for the total scale (0.92) and the subscales (primary, 0.87; secondary, 0.82; tertiary, 0.91) demonstrated good reliability. CONCLUSION: The revised 15-item MSISQ is a reliable and valid measure of sexual dysfunction in men and women with MS.


Subject(s)
Multiple Sclerosis/complications , Sexual Dysfunction, Physiological/etiology , Sexual Dysfunctions, Psychological/etiology , Surveys and Questionnaires , Female , Humans , Male , Middle Aged , Multiple Sclerosis/psychology , Principal Component Analysis , Reproducibility of Results , Sexual Dysfunction, Physiological/epidemiology , Sexual Dysfunctions, Psychological/epidemiology
10.
Int J MS Care ; 15(4): 194-201, 2013.
Article in English | MEDLINE | ID: mdl-24453783

ABSTRACT

Injectable first-line disease-modifying therapies (DMTs) for multiple sclerosis (MS) are generally prescribed for continuous use. Accordingly, the various factors that influence patient persistence with treatment and that can lead some patients to switch medications or discontinue treatment may affect clinical outcomes. Using data from the North American Research Committee on Multiple Sclerosis (NARCOMS) database, this study evaluated participants' reasons for discontinuation of injectable DMTs as well as the relationship between staying on therapy and sustained patient-reported disease progression and annualized relapse rates. Participants selected their reason(s) for discontinuation from among 16 possible options covering the categories of efficacy, safety, tolerability, and burden, with multiple responses permitted. Both unadjusted data and data adjusted for baseline age, disease duration, disability, and sex were evaluated. Discontinuation profiles varied among DMTs. Participants on intramuscular interferon beta-1a (IM IFNß-1a) and glatiramer acetate (GA) reported the fewest discontinuations based on safety concerns, although GA was associated with reports of higher burden and lower efficacy than other therapies. Difficulties with tolerability were more often reported as a reason for discontinuing subcutaneous (SC) IFNß-1a than as a reason for discontinuing IM IFNß-1a, GA, or SC IFNß-1b. In the persistent therapy cohort, less patient-reported disability progression was reported with IM IFNß-1a treatment than with SC IFNß-1a, IFNß-1b, or GA. These findings have relevance to clinical decision making and medication compliance in MS patient care.

11.
Cogn Behav Neurol ; 24(3): 128-33, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21945984

ABSTRACT

BACKGROUND: : In previous studies we and others have demonstrated an association with apolipoprotein (APOE) ε4 genotype and the presence of cognitive deficits in multiple sclerosis (MS). In this follow-up study, we have assessed whether APOE ε4 status exacerbates progression of cognitive deficits in MS. METHODS: : A total of 197 patients with MS were assessed for APOE genotype, and baseline cognitive performance was measured using a standardized battery of tests. One hundred seventy patients (86.3%) were clinically followed up for 1 year and were assessed for progression of cognitive deficits. RESULTS: : The APOE ε4 allele was present in 24.7% of patients. During 1-year follow-up, significant progression of cognitive deficits was found in APOE ε4 carriers (P=0.001) after logistic regression analysis controlling for sex, ethnicity, age, education, disease duration, severity, and subtype. CONCLUSIONS: : APOE ε4 carriers with MS have worsening progression of cognitive deficits than noncarriers. APOE ε4 carrier status predicts cognitive decline in verbal learning and memory.


Subject(s)
Apolipoprotein E4/genetics , Cognition Disorders/genetics , Disease Progression , Genetic Predisposition to Disease/genetics , Multiple Sclerosis/genetics , Adult , Aged , Cognition Disorders/complications , Cognition Disorders/psychology , Female , Follow-Up Studies , Genotype , Heterozygote , Humans , Male , Memory , Middle Aged , Multiple Sclerosis/complications , Multiple Sclerosis/psychology , Neuropsychological Tests , Polymorphism, Genetic , Verbal Learning
12.
Neurologist ; 17(2): 98-104, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21364364

ABSTRACT

BACKGROUND: Under the therapeutic point of view, neuromyelitis optica (NMO) poses major challenges. Patients with NMO manifest severe disability from recurrent demyelinating lesions and the therapies are only partially effective. We performed a retrospective analysis of the records of patients followed at our institution and provide suggestions for management of acute relapses and preventive therapy. METHODS: We searched the electronic database for patients who met criteria for NMO spectrum between January 2003 and June 2009. Patient characteristics, clinical relapses, treatments, neurological status, and medical complications were recorded. RESULTS: In the 18 patients who met the criteria for NMO different regimens of chemotherapies seemed to be modestly effective in preventing clinical relapses. After the year 2006, when rituximab began to be used for NMO patients at our institution, a significant reduction of the relapse rate was observed. After the administration of rituximab, we have systematically been monitoring the percentage of the circulating B cells and we suggest that the clinical relapses occurring while on rituximab therapy correlate with the reconstitution of circulating B cells. CONCLUSIONS: The lack of response to therapies approved for multiple sclerosis demands prompt recognition of NMO patients and the NMO-antibody testing can be critically important for that purpose. We have observed remarkable variability of the disease course with long-lasting relapse-free intervals and clusters of severe, disabling attacks. The best effects in preventing and interrupting the high frequency of relapses is achieved with rituximab whose repeated dosing should be guided by monitoring the circulating B-cell counts.


Subject(s)
Neuromyelitis Optica/pathology , Neuromyelitis Optica/physiopathology , Adult , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Autoantibodies/metabolism , B-Lymphocytes/immunology , Disease Progression , Female , Humans , Immunologic Factors/therapeutic use , Magnetic Resonance Imaging , Male , Middle Aged , Neuromyelitis Optica/drug therapy , Neuromyelitis Optica/immunology , Recurrence , Retrospective Studies , Rituximab , Treatment Outcome
13.
Ann Neurol ; 69(4): 721-34, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21425186

ABSTRACT

OBJECTIVE: The role of natural killer (NK) cells in regulating multiple sclerosis (MS) is not well understood. Additional studies with NK cells might provide insight into the mechanism of action of MS therapies such as daclizumab, an antibody against the interleukin (IL)-2R α-chain, which induces expansion of CD56(bright) NK cells. METHODS: In a relapsing-remitting form of the experimental autoimmune encephalomyelitis (EAE) model of MS induced in SJL mice, we expanded NK cells with IL-2 coupled with an anti-IL-2 monoclonal antibody (mAb) and evaluated the effects of these NK cells on EAE. Further, we investigated the effect of the human version of IL-2/IL-2 mAb on NK cells from MS patients and its effect on central nervous system (CNS) inflammation and pathology in a human-mouse chimera model and assessed the underlying mechanisms. RESULTS: IL-2/IL-2 mAb dramatically expands NK cells both in the peripheral lymphoid organs and in the CNS, and attenuates CNS inflammation and neurological deficits. Disease protection is conferred by CNS-resident NK cells. Importantly, the human version of IL-2/IL-2 mAb restored the defective CD56(+) NK cells from MS patients in a human-mouse chimera model. Both the CD56(bright) and CD56(dim) subpopulations were required to attenuate disease in this model. INTERPRETATION: These findings unveil the immunotherapeutic potential of NK cells, which can act as critical suppressor cells in target organs of autoimmunity. These results also have implications to better understand the mechanism of action of daclizumab in MS.


Subject(s)
Antibodies, Monoclonal/pharmacology , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Immunotherapy/methods , Interleukin-2/immunology , Interleukin-2/pharmacology , Killer Cells, Natural/immunology , Adult , Animals , Antibodies, Monoclonal/therapeutic use , Autoimmunity , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Central Nervous System/pathology , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Flow Cytometry , Granzymes/metabolism , Humans , Inflammation/immunology , Killer Cells, Natural/drug effects , Leukocytes, Mononuclear/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/immunology , Perforin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Severity of Illness Index , T-Lymphocytes, Regulatory/immunology
15.
J Exp Med ; 207(9): 1907-21, 2010 Aug 30.
Article in English | MEDLINE | ID: mdl-20696699

ABSTRACT

Natural killer (NK) cells of the innate immune system can profoundly impact the development of adaptive immune responses. Inflammatory and autoimmune responses in anatomical locations such as the central nervous system (CNS) differ substantially from those found in peripheral organs. We show in a mouse model of multiple sclerosis that NK cell enrichment results in disease amelioration, whereas selective blockade of NK cell homing to the CNS results in disease exacerbation. Importantly, the effects of NK cells on CNS pathology were dependent on the activity of CNS-resident, but not peripheral, NK cells. This activity of CNS-resident NK cells involved interactions with microglia and suppression of myelin-reactive Th17 cells. Our studies suggest an organ-specific activity of NK cells on the magnitude of CNS inflammation, providing potential new targets for therapeutic intervention.


Subject(s)
Autoimmunity , Killer Cells, Natural/immunology , Multiple Sclerosis/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cell Movement , Cells, Cultured , Cytotoxicity, Immunologic , Disease Models, Animal , Female , Immune Tolerance , Interleukin-17/deficiency , Interleukin-17/immunology , Interleukin-2/immunology , Killer Cells, Natural/pathology , Mice , Mice, Knockout , Multiple Sclerosis/pathology
16.
Eur J Immunol ; 40(6): 1577-89, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20352624

ABSTRACT

Human autoimmune diseases are often characterized by a relative deficiency in CD4(+)CD25(+) regulatory T cells (Treg). We therefore hypothesized that expansion of Treg can ameliorate autoimmune pathology. We tested this hypothesis in an experimental model for autoimmune myasthenia gravis (MG), a B-cell-mediated disease characterized by auto-Ab directed against the acetylcholine receptor within neuromuscular junctions. We showed that injection of immune complexes composed of the cytokine IL-2 and anti-IL-2 mAb (JES6-1A12) induced an effective and sustained expansion of Treg, via peripheral proliferation of CD4(+)CD25(+)Foxp3(+) cells and peripheral conversion of CD4(+)CD25(-)Foxp3(-) cells. The expanded Treg potently suppressed autoreactive T- and B-cell responses to acetylcholine receptor and attenuated the muscular weakness that is characteristic of MG. Thus, IL-2/anti-IL-2 mAb complexes can expand functional Treg in vivo, providing a potential clinical application of this modality for treatment of MG and other autoimmune disorders.


Subject(s)
Antibodies, Monoclonal/immunology , Antigen-Antibody Complex/immunology , Interleukin-2/immunology , Myasthenia Gravis, Autoimmune, Experimental/immunology , T-Lymphocytes, Regulatory/immunology , Animals , B-Lymphocytes/immunology , Cell Separation , Cytokines/biosynthesis , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Lymphocyte Activation , Mice , Myasthenia Gravis, Autoimmune, Experimental/pathology
17.
Exp Neurol ; 221(1): 136-45, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19879259

ABSTRACT

Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS) thought to be primarily mediated by T cells. However, emerging evidence supports an important role for B cells in the pathogenesis and inhibition of MS. Glatiramer acetate (GA), a Food and Drug Administration-approved drug for the treatment of MS, has a good safety profile. But GA's mechanism of action in MS is still elusive. In this study, we showed that B cells from GA-treated mice increased production of IL-10 and reduced expression of co-stimulatory molecules viz.: CD80 and CD86. B cells from GA-treated mice also diminished proliferation of myelin oligodendrocyte glycoprotein (MOG(35-55)) specific T cells. Purified B cells transferred from GA-treated mice suppressed experimental autoimmune encephalomyelitis (EAE) in recipient mice compared with B cells transferred from mice treated with PBS or ovalbumin. The treatment effect of GA in EAE was abrogated in B cell-deficient mice. Transfer of B cells from GA-treated mice inhibited the proliferation of autoreactive T cells as well as the development of Th1 and Th17 cells but promoted IL-10 production in recipient mice. The number of peripheral CD11b(+) macrophages in recipient mice also decreased after transfer of B cells from GA-treated mice; however, the number of dendritic cells and regulatory T cells remained unaltered. These results suggest that B cells are important to the protective effects of GA in EAE.


Subject(s)
Adoptive Transfer/methods , B-Lymphocytes/drug effects , B-Lymphocytes/physiology , Encephalomyelitis, Autoimmune, Experimental/therapy , Immunosuppressive Agents/pharmacology , Peptides/pharmacology , Animals , Antigens, CD/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/immunology , Flow Cytometry/methods , Gene Expression Regulation/physiology , Glatiramer Acetate , Glycoproteins , Interleukin-10/metabolism , Interleukin-4/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Proteins , Myelin-Associated Glycoprotein/genetics , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Transforming Growth Factor beta/metabolism
18.
Exp Neurol ; 220(2): 366-73, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19818352

ABSTRACT

T-bet, a tissue-specific transcription factor, controls T helper 1 (Th1) cell differentiation and IFN-production. Given the reciprocal relationship between Th1 and other types of helper T cells, we hypothesized that T-bet impacts multiple helper and regulatory T (Treg) cells, thereby influencing the magnitude of autoimmune disease. We tested this hypothesis in an experimental model of autoimmune myasthenia gravis (EAMG) of mice. Myasthenia gravis (MG) and EAMG are T cell-driven, IgG autoantibody-mediated disorders that destroy muscles by attacking the target antigen acetylcholine receptor (AChR) or other antigens of skeletal muscle at neuromuscular junctions. We show that, compared to wild-type mice, AChR-primed T-bet(-/-) mice are less susceptible to EAMG. This phenotype is associated with a reduction of autoreactive Th1 cells and augmentation of Th2 and Th17 cells as well as an upregulation of Foxp3 expression by T-bet(-/-)CD4(+)CD25(+) Treg cells. Thus, in our model, T-bet not only specifies the Th1 lineage but also has a broad influence on autoreactive Th2, Th17 and Treg cells. These coordinated effects reduce the genesis of pathogenic antibodies and protect against B cell-mediated EAMG.


Subject(s)
Myasthenia Gravis, Autoimmune, Experimental/genetics , T-Box Domain Proteins/genetics , T-Lymphocytes, Helper-Inducer/physiology , Animals , Antimetabolites , Bromodeoxyuridine , CD4 Antigens/genetics , Cell Proliferation/drug effects , Cell Separation , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunoglobulin G/biosynthesis , Immunoglobulin G/genetics , Interleukin-2 Receptor alpha Subunit/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cholinergic/biosynthesis , Receptors, Cholinergic/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
19.
Acta Pharmacol Sin ; 30(6): 715-22, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19448649

ABSTRACT

Cigarette smoke is a major health risk factor which significantly increases the incidence of diseases including lung cancer and respiratory infections. However, there is increasing evidence that smokers have a lower incidence of some inflammatory and neurodegenerative diseases. Nicotine is the main immunosuppressive constituent of cigarette smoke, which inhibits both the innate and adaptive immune responses. Unlike cigarette smoke, nicotine is not yet considered to be a carcinogen and may, in fact, have therapeutic potential as a neuroprotective and anti-inflammatory agent. This review provides a synopsis summarizing the effects of nicotine on the immune system and its (nicotine) influences on various neurological diseases.


Subject(s)
Immune System/drug effects , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Animals , Humans , Immune System/metabolism , Inflammation/drug therapy , Inflammation/immunology , Inflammation/physiopathology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/physiopathology , Nicotine/immunology , Nicotinic Agonists/immunology , Smoking/adverse effects
20.
J Immunol ; 182(3): 1730-9, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19155522

ABSTRACT

The expression of nicotinic acetylcholine receptors by neurons, microglia, and astrocytes suggests possibly diverse mechanisms by which natural nicotinic cholinergic signaling and exposure to nicotine could modulate immune responses within the CNS. In this study, we show that nicotine exposure significantly delays and attenuates inflammatory and autoimmune responses to myelin Ags in the mouse experimental autoimmune encephalomyelitis model. In the periphery, nicotine exposure inhibits the proliferation of autoreactive T cells and alters the cytokine profile of helper T cells. In the CNS, nicotine exposure selectively reduces numbers of CD11c(+) dendritic and CD11b(+) infiltrating monocytes and resident microglial cells and down-regulates the expression of MHC class II, CD80, and CD86 molecules on these cells. The results underscore roles of nicotinic acetylcholine receptors and nicotinic cholinergic signaling in inflammatory and immune responses and suggest novel therapeutic options for the treatment of inflammatory and autoimmune disorders, including those that affect the CNS.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Autoimmune Diseases/immunology , Autoimmune Diseases/prevention & control , Encephalomyelitis, Acute Disseminated/immunology , Encephalomyelitis, Acute Disseminated/prevention & control , Immunosuppressive Agents/therapeutic use , Nicotine/therapeutic use , Amino Acid Sequence , Animals , Autoimmune Diseases/pathology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cells, Cultured , Encephalomyelitis, Acute Disseminated/pathology , Female , Glycoproteins/toxicity , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Myelin Proteolipid Protein/toxicity , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/toxicity , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...