Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Adv Ther ; 41(8): 3059-3075, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38861218

ABSTRACT

INTRODUCTION: Switching disease-modifying therapy (DMT) may be considered for relapsing-remitting multiple sclerosis (RRMS) if a patient's current therapy is no longer optimal. This was particularly important during the recent COVID-19 pandemic because of considerations around immune deficiency and impaired vaccine response associated with B cell-depleting DMTs. This real-world, single-center study aimed to evaluate change or decline in functional ability and overall disease stability in people with RRMS who were switched from B cell-depleting ocrelizumab (OCRE) to diroximel fumarate (DRF) because of safety concern related to the COVID-19 pandemic. METHODS: Adults with RRMS were included if they had been clinically stable for ≥ 1 year on OCRE. Data collected at baseline and 1 year post switch included relapse rate, magnetic resonance imaging (MRI), blood work for assessment of peripheral immune parameters, the Cognitive Assessment Battery (CAB), optical coherence tomography (OCT), and patient-reported outcomes (PROs). RESULTS: Participants (N = 25) had a mean (SD) age of 52 (9) years, and a mean (SD) duration of 26 (8) months' treatment with OCRE before the switch to DRF. Median washout duration since the last OCRE infusion was 7 months (range 4-18 months). No participants relapsed on DRF during follow-up, and all remained persistent on DRF after 1 year. There were no significant changes in peripheral immune parameters, other than an increase in the percentage of CD19+ cells 1 year after switching (p < 0.05). Similarly, there were no significant changes in CAB, OCT, and PROs. CONCLUSION: These preliminary findings suggest that transition to DRF from OCRE may be an effective treatment option for people with RRMS who are clinically stable but may need to switch for reasons unrelated to effectiveness. Longer follow-up times on larger samples are needed to confirm these observations.


Subject(s)
Antibodies, Monoclonal, Humanized , Multiple Sclerosis, Relapsing-Remitting , Humans , Male , Female , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/adverse effects , Adult , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Immunologic Factors/therapeutic use , Immunologic Factors/adverse effects , Dimethyl Fumarate/therapeutic use , Dimethyl Fumarate/adverse effects , Treatment Outcome , COVID-19 , Drug Substitution , SARS-CoV-2
2.
Mult Scler J Exp Transl Clin ; 9(3): 20552173231191170, 2023.
Article in English | MEDLINE | ID: mdl-37692293

ABSTRACT

People with multiple sclerosis (pwMS) have an increased risk of infection. As disease-modifying therapies (DMTs) and other treatments may interact with the immune system, there may be concerns about vaccine efficacy and safety. Therefore, it is important to evaluate possible interactions between DMTs and vaccines. The fumarates, dimethyl fumarate, diroximel fumarate, and monomethyl fumarate, are approved for the treatment of relapsing multiple sclerosis. This review assesses the evidence on vaccine response in pwMS treated with fumarates, with a particular focus on COVID-19 vaccines. Treatment with fumarates does not appear to result in blunting of humoral responses to vaccination; for COVID-19 vaccines, particularly RNA-based vaccines, evidence indicates antibody responses similar to those of healthy recipients. While data on the effect of fumarates on T-cell responses are limited, they do not indicate any significant blunting. COVID-19 vaccines impart a similar degree of protection against severe COVID-19 infection for pwMS on fumarates as in the general population. Adverse reactions following vaccination are generally consistent with those observed in the wider population; no additional safety signals have emerged in those on fumarates. Additionally, no increase in relapse has been observed in pwMS following vaccination. In pwMS receiving fumarates, vaccination is generally safe and elicits protective immune responses.

4.
Adv Ther ; 38(7): 3550-3588, 2021 07.
Article in English | MEDLINE | ID: mdl-34075554

ABSTRACT

People with multiple sclerosis (MS) are at risk for infections that can result in amplification of baseline symptoms and possibly trigger clinical relapses. Vaccination can prevent infection through the activation of humoral and cellular immune responses. This is particularly pertinent in the era of emerging novel vaccines against severe acute respiratory syndrome coronavirus 2, the virus that causes coronavirus disease 2019 (COVID-19). MS disease-modifying therapies (DMTs), which affect the immune system, may impact immune responses to COVID-19 vaccines in people with MS. The objective of this article is to provide information on immune system responses to vaccinations and review previous studies of vaccine responses in people with MS to support the safety and importance of receiving currently available and emerging COVID-19 vaccines. Immunological studies have shown that coordinated interactions between T and B lymphocytes of the adaptive immune system are key to successful generation of immunological memory and production of neutralizing antibodies following recognition of vaccine antigens by innate immune cells. CD4+ T cells are essential to facilitate CD8+ T cell and B cell activation, while B cells drive and sustain T cell memory. Data suggest that some classes of DMT, including type 1 interferons and glatiramer acetate, may not significantly impair the response to vaccination. DMTs-such as sphingosine-1-phosphate receptor modulators, which sequester lymphocytes from circulation; alemtuzumab; and anti-CD20 therapies, which rely on depleting populations of immune cells-have been shown to attenuate responses to conventional vaccines. Currently, three COVID-19 vaccines have been granted emergency use authorization in the USA on the basis of promising interim findings of ongoing trials. Because analyses of these vaccines in people with MS are not available, decisions regarding COVID-19 vaccination and DMT choice should be informed by data and expert consensus, and personalized with considerations for disease burden, risk of infection, and other factors.


Subject(s)
COVID-19 , Multiple Sclerosis , COVID-19 Vaccines , Glatiramer Acetate , Humans , SARS-CoV-2
5.
Neurobiol Dis ; 93: 21-7, 2016 09.
Article in English | MEDLINE | ID: mdl-27080468

ABSTRACT

Hematogenous macrophages remove myelin debris from injured peripheral nerves to provide a micro-environment conducive to axonal regeneration. Previously, we observed that injured peripheral nerves from Beta-site APP Cleaving Enzyme 1 (BACE1) knockout (KO) mice displayed earlier influx of and enhanced phagocytosis by macrophages when compared to wild-type (WT) mice. These observations suggest that BACE1 might regulate macrophage influx into distal stumps of injured nerves. To determine through which pathway BACE1 influences macrophage influx, we used a mouse inflammation antibody array to assay the expression of inflammation-related proteins in injured nerves of BACE1 KO and WT mice. The most significant change was in expression of tumor necrosis factor receptor 1 (TNFR1) in the distal stump of injured BACE1 KO nerves. Western blotting of protein extracts confirmed increased expression of TNFR1 and its downstream transcriptional factor NFκB in the BACE1 KO distal stumps. Additionally, treatment of WT mice with a BACE1 inhibitor resulted in increased TNFR1 expression and signaling in the distal stump of injured nerves. Exogenous TNFα increased nuclear translocation of p65 NFκB in BACE1 KO tissue and cultured fibroblasts compared with control WT. BACE1 regulates TNFR1 expression at the level of gene expression and not through proteolytic processing. The accelerated macrophage influx in injured nerves of BACE1 KO mice correlates with increased expression and signaling via TNFR1, indicating a link between BACE1 activity and TNFR1 expression/signaling that might contribute to repair of the injured nervous system.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Myelin Sheath/metabolism , Peripheral Nerve Injuries/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction/physiology , Animals , Macrophages/metabolism , Mice, Knockout , Nerve Regeneration/physiology , Peripheral Nerves/metabolism , Phagocytosis/physiology
6.
Neurology ; 86(4): 382-90, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26718578

ABSTRACT

OBJECTIVE: To study the safety profile and characterize the immunologic effects of high- vs low-dose cholecalciferol supplementation in patients with multiple sclerosis (MS). METHODS: In this double-blind, single-center randomized pilot study, 40 patients with relapsing-remitting MS were randomized to receive 10,400 IU or 800 IU cholecalciferol daily for 6 months. Assessments were performed at baseline and 3 and 6 months. RESULTS: Mean increase of 25-hydroxyvitamin D levels from baseline to final visit was larger in the high-dose group (34.9 ng/mL; 95% confidence interval [CI] 25.0-44.7 ng/mL) than in the low-dose group (6.9 ng/mL; 95% CI 1.0-13.7 ng/mL). Adverse events were minor and did not differ between the 2 groups. Two relapses occurred, one in each treatment arm. In the high-dose group, we found a reduction in the proportion of interleukin-17(+)CD4(+) T cells (p = 0.016), CD161(+)CD4(+) T cells (p = 0.03), and effector memory CD4(+) T cells (p = 0.021) with a concomitant increase in the proportion of central memory CD4(+) T cells (p = 0.018) and naive CD4(+) T cells (p = 0.04). These effects were not observed in the low-dose group. CONCLUSIONS: Cholecalciferol supplementation with 10,400 IU daily is safe and tolerable in patients with MS and exhibits in vivo pleiotropic immunomodulatory effects in MS, which include reduction of interleukin-17 production by CD4(+) T cells and decreased proportion of effector memory CD4(+) T cells with concomitant increase in central memory CD4(+) T cells and naive CD4(+) T cells. CLASSIFICATION OF EVIDENCE: This study provides Class I evidence that cholecalciferol supplementation with 10,400 IU daily is safe and well-tolerated in patients with MS and exhibits in vivo pleiotropic immunomodulatory effects.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Cholecalciferol , Immunologic Factors , Interleukin-17/metabolism , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/immunology , Vitamin D/analogs & derivatives , Adult , CD4 Lymphocyte Count , Cholecalciferol/administration & dosage , Cholecalciferol/adverse effects , Cholecalciferol/pharmacology , Dietary Supplements , Double-Blind Method , Female , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/adverse effects , Immunologic Factors/pharmacology , Male , Middle Aged , Pilot Projects , Treatment Outcome , Vitamin D/blood
7.
J Immunol ; 195(4): 1399-1407, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26150529

ABSTRACT

Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the CNS that has been linked with defects in regulatory T cell function. Therefore, strategies to selectively target pathogenic cells via enhanced regulatory T cell activity may provide therapeutic benefit. Kv1.3 is a voltage-gated potassium channel expressed on myelin-reactive T cells from MS patients. Kv1.3-knockout (KO) mice are protected from experimental autoimmune encephalomyelitis, an animal model of MS, and Kv1.3-KO Th cells display suppressive capacity associated with increased IL-10. In this article, we demonstrate that myelin oligodendrocyte glycoprotein-specific Kv1.3-KO Th cells exhibit a unique regulatory phenotype characterized by high CD25, CTLA4, pSTAT5, FoxO1, and GATA1 expression without a corresponding increase in Foxp3. These phenotypic changes result from increased signaling through IL-2R. Moreover, myelin oligodendrocyte glycoprotein-specific Kv1.3-KO Th cells can ameliorate experimental autoimmune encephalomyelitis following transfer to wild-type recipients in a manner that is partially dependent on IL-2R and STAT5 signaling. The present study identifies a population of Foxp3(-) T cells with suppressive properties that arises in the absence of Kv1.3 and enhances the understanding of the molecular mechanism by which these cells are generated. This increased understanding could contribute to the development of novel therapies for MS patients that promote heightened immune regulation.


Subject(s)
Antigens/immunology , Forkhead Transcription Factors/metabolism , Kv1.3 Potassium Channel/deficiency , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Animals , CTLA-4 Antigen/genetics , CTLA-4 Antigen/metabolism , Calcium/metabolism , Cytokines/biosynthesis , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Gene Expression , Immunomodulation , Immunophenotyping , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Mice , Mice, Knockout , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Myelin-Oligodendrocyte Glycoprotein/immunology , NFATC Transcription Factors/metabolism , Phenotype , Phosphorylation , STAT5 Transcription Factor/metabolism , Signal Transduction
8.
J Neurosci ; 35(22): 8626-39, 2015 Jun 03.
Article in English | MEDLINE | ID: mdl-26041928

ABSTRACT

Multiple sclerosis (MS) is a demyelinating disease of the CNS characterized by inflammation and neurodegeneration. Animal models that enable the study of remyelination in the context of ongoing inflammation are greatly needed for the development of novel therapies that target the pathological inhibitory cues inherent to the MS plaque microenvironment. We report the development of an innovative animal model combining cuprizone-mediated demyelination with transfer of myelin-reactive CD4(+) T cells. Characterization of this model reveals both Th1 and Th17 CD4(+) T cells infiltrate the CNS of cuprizone-fed mice, with infiltration of Th17 cells being more efficient. Infiltration correlates with impaired spontaneous remyelination as evidenced by myelin protein expression, immunostaining, and ultrastructural analysis. Electron microscopic analysis further reveals that demyelinated axons are preserved but reduced in caliber. Examination of the immune response contributing to impaired remyelination highlights a role for peripheral monocytes with an M1 phenotype. This study demonstrates the development of a novel animal model that recapitulates elements of the microenvironment of the MS plaque and reveals an important role for T cells and peripheral monocytes in impairing endogenous remyelination in vivo. This model could be useful for testing putative MS therapies designed to enhance remyelination in the setting of active inflammation, and may also facilitate modeling the pathophysiology of denuded axons, which has been a challenge in rodents because they typically remyelinate very quickly.


Subject(s)
Central Nervous System/pathology , Cuprizone/toxicity , Demyelinating Diseases/therapy , Monoamine Oxidase Inhibitors/toxicity , Myelin Sheath/metabolism , Th17 Cells/physiology , Adoptive Transfer , Animals , Cell Polarity/drug effects , Cells, Cultured , Central Nervous System/ultrastructure , Demyelinating Diseases/chemically induced , Disease Models, Animal , Freund's Adjuvant/toxicity , Interleukin-17/metabolism , Leukocyte Common Antigens/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/pathology , Monocytes/ultrastructure , Myelin Proteins/metabolism , Myelin-Oligodendrocyte Glycoprotein/toxicity , Neutrophil Infiltration , Peptide Fragments/toxicity , Regeneration/drug effects , Th17 Cells/ultrastructure , Time Factors
9.
J Neuroimmunol ; 279: 20-4, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25669995

ABSTRACT

Vitamin D deficiency is associated with increased susceptibility to multiple sclerosis (MS) and increased disease activity. Vitamin D is a potent immunomodulator but the effects of vitamin D treatment on T cell memory have not been explored. We studied the effects of 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) on T cell memory in MS patients (n = 10) and healthy controls (n = 10). In vitro treatment of PBMC cultures with 1,25(OH)2D3, led to a decrease in the proportion of effector memory T cells with an increase in naïve T cells, compared to vehicle in both groups. Further studies to unravel the mechanism of this effect and to understand its long-term implications are required.


Subject(s)
Calcitriol/deficiency , Multiple Sclerosis/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Vitamin D Deficiency/pathology , Adult , Antigens, CD/metabolism , Calcitriol/pharmacology , Cells, Cultured , Female , Flow Cytometry , Humans , In Vitro Techniques , Male , Multiple Sclerosis/immunology , Receptors, CXCR3/metabolism , Statistics, Nonparametric , T-Lymphocytes/drug effects , Vitamin D Deficiency/immunology
10.
J Neuroimmunol ; 270(1-2): 13-21, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24680062

ABSTRACT

Fingolimod (FTY720) is a multiple sclerosis (MS) therapeutic that upon phosphorylation causes the internalization of sphingosine-1-phosphate receptors (S1PR) and traps CCR7+ T-cells in lymph nodes but relatively spares CCR7-effector T-cells. Nonetheless, FTY720-treated patients are more susceptible to viral infections, indicating a CD8 T-cell defect. Thus, the effects of FTY720 on CD8 T-cells were investigated. To this end, we utilized experimental autoimmune encephalomyelitis (EAE) and a murine influenza model. CD8 T-cell trafficking, IFNγ and Granzyme B (GrB) production were assessed by flow cytometry. CD8 T-cell cytotoxic function was assessed in vitro by an LDH release assay. FTY720 not only ameliorated EAE by sequestering T-cells, but also reduced IFNγ and Granzyme B (GrB) in splenic CD8 T-cells. Murine influenza infection was exacerbated and mortality was increased, as FTY720 inhibited CD8 T-cell GrB production and lung infiltration. Remarkably, only the unphosphorylated compound was able to reduce IFNγ and GrB levels in CD8 T-cells and inhibits their cytotoxic function in vitro. The phosphorylated moiety had no effect in vitro, indicating that CD8 T-cell suppression by FTY720 is independent of S1PR modulation. The addition of arachidonic acid rescued CD8 T-cell function, suggesting that this effect may be mediated via inhibition of cytosolic phospholipase A2. Herein, we demonstrate that FTY720 suppresses CD8 T-cells independently of its trafficking effects and S1PR modulation. This provides a novel explanation not only for the increased rate of viral infections in FTY720-treated patients, but also for its efficacy in MS, as CD8 T-cells have emerged as crucial mediators of MS pathogenesis.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Encephalomyelitis, Autoimmune, Experimental/immunology , Immunosuppressive Agents/pharmacology , Orthomyxoviridae Infections/immunology , Propylene Glycols/pharmacology , Sphingosine/analogs & derivatives , Animals , CD8-Positive T-Lymphocytes/immunology , Chemotaxis, Leukocyte/drug effects , Female , Fingolimod Hydrochloride , Flow Cytometry , Granzymes/biosynthesis , Influenza A Virus, H1N1 Subtype , Interferon-gamma/biosynthesis , Lysophospholipids/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Sphingosine/metabolism , Sphingosine/pharmacology
11.
Proc Natl Acad Sci U S A ; 110(52): 21101-6, 2013 Dec 24.
Article in English | MEDLINE | ID: mdl-24324134

ABSTRACT

Pharmacologic targeting of T helper (TH) cell trafficking poses an attractive opportunity for amelioration of autoimmune diseases such as multiple sclerosis (MS). MS risk is associated with vitamin D deficiency, and its bioactive form, 1,25-dihydroxyvitamin D3 [1,25(OH)2D3], has been shown to prevent experimental autoimmune encephalomyelitis, a mouse model of MS, via an incompletely understood mechanism. Herein, we systematically examined 1,25(OH)2D3 effects on TH cells during their migration from the lymph nodes to the CNS. Our data demonstrate that myelin-reactive TH cells are successfully generated in the presence of 1,25(OH)2D3, secrete proinflammatory cytokines, and do not preferentially differentiate into suppressor T cells. These cells are able to leave the lymph node, enter the peripheral circulation, and migrate to the s.c. immunization sites. However, TH cells from 1,25(OH)2D3-treated mice are unable to enter the CNS parenchyma but are instead maintained in the periphery. Upon treatment cessation, mice rapidly develop experimental autoimmune encephalomyelitis, demonstrating that 1,25(OH)2D3 prevents the disease only temporarily likely by halting TH cell migration into the CNS.


Subject(s)
Calcitriol/pharmacology , Cell Movement/drug effects , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , T-Lymphocytes, Helper-Inducer/drug effects , Animals , Cell Movement/immunology , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Enzyme-Linked Immunosorbent Assay , Luminescent Measurements , Mice , Mice, Inbred C57BL , Statistics, Nonparametric , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism
12.
Cell Immunol ; 284(1-2): 68-74, 2013.
Article in English | MEDLINE | ID: mdl-23933188

ABSTRACT

Double-negative T (DNT) cells are αßTCR(+)CD3(+)CD4(-)CD8(-)NK1.1(-) cells that constitute a small but significant proportion of the αßTCR(+) T cells. Their developmental pathway and pathological significance remain unclear. In the present study, we utilized chronic in vitro stimulation of CD4(+) T cells to mimic immune hyper-activation of autoimmune lymphoproliferative syndrome and systemic lupus erythematosus, conditions characterized by DNT cells accumulation. After approximately 4-5 rounds of stimulation, the CD3(+)CD4(-) population became apparent. These cells did not express CD8, NK1.1, γδTCR, or B220, exhibited a highly proliferative effector phenotype, and were dependent on T cell receptor (TCR) stimulation for survival. Moreover, CD3(+)CD4(-) cells expressed MHC class II-restricted αßTCR, indicative of their origin from a CD4(+) T cell population. The results presented herein illustrate a novel method of DNT cell generation in vitro and suggest that immune hyper-activation could also be implicated in the genesis of the disease-associated DNT cells in vivo.


Subject(s)
Autoimmune Lymphoproliferative Syndrome/immunology , CD4-Positive T-Lymphocytes/immunology , Lupus Erythematosus, Systemic/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cell Proliferation , Down-Regulation , Female , Flow Cytometry , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype
13.
PLoS One ; 8(1): e54267, 2013.
Article in English | MEDLINE | ID: mdl-23382885

ABSTRACT

Increased expression of the voltage-gated potassium channel Kν1.3 on activated effector memory T cells (T(EM)) is associated with pathology in multiple sclerosis (MS). To date, most studies of Kν1.3 channels in MS have focused on CD4+ T(EM) cells. Much less is known about the functional relevance of Kv1.3 on CD8+ T(EM) cells. Herein, we examined the effects of Kν1.3 blockade on CD8+ T cell proliferation, differentiation into cytotoxic effector cells, and release of granzyme B (GrB), a key effector of CD8+ T cell-mediated cytotoxicity. We confirmed the expression of Kv1.3 channels on activated human CD8+ T lymphocytes by immunofluorescent staining. To test the functional relevance of the Kv1.3 channel in CD8+ T cells, we inhibited this channel via pharmacological blockers or a lentiviral-dominant negative (Kv1.xDN) approach and determined the effects of the blockade on critical pathogenic parameters of CD8+ T cells. We found that blockade of Kv1.3 with both lentivirus and pharmacologic agents effectively inhibited cytotoxic effector memory cells' proliferation, secretion of GrB, and their ability to kill neural progenitor cells. Intriguingly, the KvDN transduced T cells exhibited arrested differentiation from central memory (T(CM)) to effector memory (T(EM)) states. Transduction of cells that had already differentiated into T(EM) with KvDN led to their conversion into T(CM). CD8+ T(EM) have a critical role in MS and other autoimmune diseases. Our present results indicate a critical role for Kv1.3 in the conversion of CD8+ T cells into potential pathogenic effector cells with cytotoxic function.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Granzymes/metabolism , Kv1.3 Potassium Channel/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , Cell Differentiation/genetics , Cell Proliferation/drug effects , Granzymes/genetics , Granzymes/immunology , Humans , Immunologic Memory/drug effects , Immunologic Memory/genetics , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/genetics , Lymphocyte Activation/immunology , Potassium Channel Blockers/pharmacology
14.
J Neuroimmunol ; 249(1-2): 86-92, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22633193

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.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Central Nervous System Diseases/cerebrospinal fluid , T-Lymphocyte Subsets/immunology , Adolescent , Adult , Aged , Aged, 80 and over , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Central Nervous System Diseases/immunology , Central Nervous System Diseases/metabolism , Child , Child, Preschool , Female , Humans , Immunoglobulin G/cerebrospinal fluid , Infant , Inflammation/cerebrospinal fluid , Inflammation/immunology , Inflammation/metabolism , Leukocyte Common Antigens/biosynthesis , Leukocyte Common Antigens/cerebrospinal fluid , Leukocyte Common Antigens/immunology , Male , Middle Aged , Receptors, CCR7/analysis , Receptors, CCR7/biosynthesis , Receptors, CCR7/immunology , T-Lymphocyte Subsets/metabolism , Young Adult
15.
J Immunol ; 188(12): 5877-86, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22581856

ABSTRACT

Increasing evidence suggests ion channels have critical functions in the differentiation and plasticity of T cells. Kv1.3, a voltage-gated K(+) channel, is a functional marker and a pharmacological target for activated effector memory T cells. Selective Kv1.3 blockers have been shown to inhibit proliferation and cytokine production by human and rat effector memory T cells. We used Kv1.3 knockout (KO) mice to investigate the mechanism by which Kv1.3 blockade affects CD4(+) T cell differentiation during an inflammatory immune-mediated disease. Kv1.3 KO animals displayed significantly lower incidence and severity of myelin oligodendrocyte glycoprotein (MOG) peptide-induced experimental autoimmune encephalomyelitis. Kv1.3 was the only K(V) channel expressed in MOG 35-55-specific CD4(+) T cell blasts, and no K(V) current was present in MOG-specific CD4(+) T cell-blasts from Kv1.3 KO mice. Fewer CD4(+) T cells migrated to the CNS in Kv1.3 KO mice following disease induction, and Ag-specific proliferation of CD4(+) T cells from these mice was impaired with a corresponding cell-cycle delay. Kv1.3 was required for optimal expression of IFN-γ and IL-17, whereas its absence led to increased IL-10 production. Dendritic cells from Kv1.3 KO mice fully activated wild-type CD4(+) T cells, indicating a T cell-intrinsic defect in Kv1.3 KO mice. The loss of Kv1.3 led to a suppressive phenotype, which may contribute to the mechanism by which deletion of Kv1.3 produces an immunotherapeutic effect. Skewing of CD4(+) T cell differentiation toward Ag-specific regulatory T cells by pharmacological blockade or genetic suppression of Kv1.3 might be beneficial for therapy of immune-mediated diseases such as multiple sclerosis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Kv1.3 Potassium Channel/metabolism , Lymphocyte Activation/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Blotting, Western , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Electrophysiology , Encephalomyelitis, Autoimmune, Experimental/pathology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phenotype , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/cytology
16.
J Biol Chem ; 287(2): 1261-8, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22110135

ABSTRACT

The maintenance of T cell memory is critical for the development of rapid recall responses to pathogens, but may also have the undesired side effect of clonal expansion of T effector memory (T(EM)) cells in chronic autoimmune diseases. The mechanisms by which lineage differentiation of T cells is controlled have been investigated, but are not completely understood. Our previous work demonstrated a role of the voltage-gated potassium channel Kv1.3 in effector T cell function in autoimmune disease. In the present study, we have identified a mechanism by which Kv1.3 regulates the conversion of T central memory cells (T(CM)) into T(EM). Using a lentiviral-dominant negative approach, we show that loss of function of Kv1.3 mediates reversion of T(EM) into T(CM), via a delay in cell cycle progression at the G2/M stage. The inhibition of Kv1.3 signaling caused an up-regulation of SMAD3 phosphorylation and induction of nuclear p21(cip1) with resulting suppression of Cdk1 and cyclin B1. These data highlight a novel role for Kv1.3 in T cell differentiation and memory responses, and provide further support for the therapeutic potential of Kv1.3 specific channel blockers in T(EM)-mediated autoimmune diseases.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cyclin-Dependent Kinase Inhibitor p21/immunology , Immunologic Memory , Kv1.3 Potassium Channel/immunology , Signal Transduction/immunology , Smad3 Protein/immunology , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , CD4-Positive T-Lymphocytes/metabolism , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/immunology , CDC2 Protein Kinase/metabolism , Cell Cycle Checkpoints/genetics , Cell Cycle Checkpoints/immunology , Cell Division/genetics , Cell Division/immunology , Cells, Cultured , Cyclin B1/genetics , Cyclin B1/immunology , Cyclin B1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , G2 Phase/genetics , G2 Phase/immunology , Humans , Kv1.3 Potassium Channel/genetics , Kv1.3 Potassium Channel/metabolism , Phosphorylation/genetics , Phosphorylation/immunology , Signal Transduction/genetics , Smad3 Protein/genetics , Smad3 Protein/metabolism
17.
J Exp Med ; 208(11): 2291-303, 2011 Oct 24.
Article in English | MEDLINE | ID: mdl-21987655

ABSTRACT

Fumarates improve multiple sclerosis (MS) and psoriasis, two diseases in which both IL-12 and IL-23 promote pathogenic T helper (Th) cell differentiation. However, both diseases show opposing responses to most established therapies. First, we show in humans that fumarate treatment induces IL-4-producing Th2 cells in vivo and generates type II dendritic cells (DCs) that produce IL-10 instead of IL-12 and IL-23. In mice, fumarates also generate type II DCs that induce IL-4-producing Th2 cells in vitro and in vivo and protect mice from experimental autoimmune encephalomyelitis. Type II DCs result from fumarate-induced glutathione (GSH) depletion, followed by increased hemoxygenase-1 (HO-1) expression and impaired STAT1 phosphorylation. Induced HO-1 is cleaved, whereupon the N-terminal fragment of HO-1 translocates into the nucleus and interacts with AP-1 and NF-κB sites of the IL-23p19 promoter. This interaction prevents IL-23p19 transcription without affecting IL-12p35, whereas STAT1 inactivation prevents IL-12p35 transcription without affecting IL-23p19. As a consequence, GSH depletion by small molecules such as fumarates induces type II DCs in mice and in humans that ameliorate inflammatory autoimmune diseases. This therapeutic approach improves Th1- and Th17-mediated autoimmune diseases such as psoriasis and MS by interfering with IL-12 and IL-23 production.


Subject(s)
Dendritic Cells/immunology , Fumarates/immunology , Fumarates/therapeutic use , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Psoriasis/drug therapy , Psoriasis/immunology , Animals , Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/drug effects , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Heme Oxygenase-1/metabolism , Humans , Interleukin-12/immunology , Interleukin-23/immunology , Macrophages/immunology , Mice , NIH 3T3 Cells , Promoter Regions, Genetic , Reactive Oxygen Species/metabolism , Signal Transduction/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Transcription Factor RelA/metabolism
18.
Cell ; 144(1): 132-42, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-21215375

ABSTRACT

The adaptive immune system is thought to be a rich source of protein biomarkers, but diagnostically useful antibodies remain unknown for a large number of diseases. This is, in part, because the antigens that trigger an immune response in many diseases remain unknown. We present here a general and unbiased approach to the identification of diagnostically useful antibodies that avoids the requirement for antigen identification. This method involves the comparative screening of combinatorial libraries of unnatural, synthetic molecules against serum samples obtained from cases and controls. Molecules that retain far more IgG antibodies from the case samples than the controls are identified and subsequently tested as capture agents for diagnostically useful antibodies. The utility of this method is demonstrated using a mouse model for multiple sclerosis and via the identification of two candidate IgG biomarkers for Alzheimer's disease.


Subject(s)
Alzheimer Disease/diagnosis , Antibodies , Immunoglobulin G , Peptide Library , Animals , Biomarkers/metabolism , Encephalomyelitis, Autoimmune, Experimental/diagnosis , Humans , Mice
19.
J Immunol ; 185(12): 7161-4, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21076063

ABSTRACT

Th17 cells play a significant role in inflammatory and autoimmune responses. Although a number of molecular pathways that contribute to the lineage differentiation of T cells have been discovered, the mechanisms by which lineage commitment occurs are not fully understood. Transcription factors play a key role in driving T cells toward specific lineages. We have identified a role for the transcription factor Kruppel-like factor (KLF) 4 in the development of IL-17-producing CD4(+) T cells. KLF4 was required for the production of IL-17, and further, chromatin immunoprecipitation analysis demonstrated binding of KLF4 to the IL-17 promoter, indicating a direct effect on the regulation of IL-17. Further, KLF4-deficient T cells upregulated expression of retinoic acid-related orphan receptor γt similar to wild-type during the polarization process toward Th17, suggesting that these two transcription factors are regulated independently.


Subject(s)
Cell Differentiation/immunology , Kruppel-Like Transcription Factors/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Promoter Regions, Genetic/immunology , Th17 Cells/immunology , Up-Regulation/immunology , Animals , Cell Differentiation/genetics , Interleukin-17/genetics , Interleukin-17/immunology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Promoter Regions, Genetic/genetics , Th17 Cells/cytology , Up-Regulation/genetics
20.
Ann Neurol ; 67(4): 498-507, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20437585

ABSTRACT

OBJECTIVE: To determine if suppressing Nogo-A, an axonal inhibitory protein, will promote functional recovery in a murine model of multiple sclerosis (MS). METHODS: A small interfering RNA was developed to specifically suppress Nogo-A (siRNA-NogoA). The siRNA-NogoA silencing effect was evaluated in vitro and in vivo via immunohistochemistry. The siRNA was administered intravenously in 2 models of experimental autoimmune encephalomyelitis (EAE). Axonal repair was measured by upregulation of GAP43. Enzyme-linked immunosorbent assay, flow cytometry, and (3)H-thymidine incorporation were used to determine immunological changes in myelin-specific T cells in mice with EAE. RESULTS: The siRNA-NogoA suppressed Nogo-A expression in vitro and in vivo. Systemic administration of siRNA-NogoA ameliorated EAE and promoted axonal repair, as demonstrated by enhanced GAP43+ axons in the lesions. Myelin-specific T-cell proliferation and cytokine production were unchanged in the siRNA-NogoA-treated mice. INTERPRETATION: Silencing Nogo-A in EAE promotes functional recovery. The therapeutic benefit appears to be mediated by axonal growth and repair, and is not attributable to changes in the encephalitogenic capacity of the myelin-specific T cells. Silencing Nogo-A may be a therapeutic option for MS patients to prevent permanent functional deficits caused by immune-mediated axonal damage.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Myelin Proteins/metabolism , RNA, Small Interfering/therapeutic use , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Enzyme-Linked Immunosorbent Assay/methods , Flow Cytometry/methods , GAP-43 Protein/genetics , GAP-43 Protein/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glycoproteins/adverse effects , Interferon-gamma/metabolism , Interleukin-10/metabolism , Lymphocytes/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Basic Protein/genetics , Myelin Basic Protein/pharmacology , Myelin Proteins/genetics , Myelin-Oligodendrocyte Glycoprotein , Neuroblastoma , Nogo Proteins , Peptide Fragments/adverse effects , Peptide Fragments/genetics , Peptide Fragments/pharmacology , RNA, Small Interfering/genetics , Spinal Cord/metabolism , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/pharmacology , Transfection/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...