Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Pract Neurol ; 2022 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-35863879

RESUMEN

Infection in people with multiple sclerosis (MS) is of major concern, particularly for those receiving disease-modifying therapies. This article explores the risk of infection in people with MS and provides guidance-developed by Delphi consensus by specialists involved in their management-on how to screen for, prevent and manage infection in this population.

2.
J Neuroinflammation ; 17(1): 349, 2020 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-33222687

RESUMEN

BACKGROUND: Multiple sclerosis (MS) is an immune-mediated disease that damages myelin in the central nervous system (CNS). We investigated the profile of CCN3, a known regulator of immune function and a potential mediator of myelin regeneration, in multiple sclerosis in the context of disease state and disease-modifying treatment. METHODS: CCN3 expression was analysed in plasma, immune cells, CSF and brain tissue of MS patient groups and control subjects by ELISA, western blot, qPCR, histology and in situ hybridization. RESULTS: Plasma CCN3 levels were comparable between collective MS cohorts and controls but were significantly higher in progressive versus relapsing-remitting MS and between patients on interferon-ß versus natalizumab. Higher body mass index was associated with higher CCN3 levels in controls as reported previously, but this correlation was absent in MS patients. A significant positive correlation was found between CCN3 levels in matched plasma and CSF of MS patients which was absent in a comparator group of idiopathic intracranial hypertension patients. PBMCs and CD4+ T cells significantly upregulated CCN3 mRNA in MS patients versus controls. In the CNS, CCN3 was detected in neurons, astrocytes and blood vessels. Although overall levels of area immunoreactivity were comparable between non-affected, demyelinated and remyelinated tissue, the profile of expression varied dramatically. CONCLUSIONS: This investigation provides the first comprehensive profile of CCN3 expression in MS and provides rationale to determine if CCN3 contributes to neuroimmunological functions in the CNS.


Asunto(s)
Interferón beta/uso terapéutico , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Esclerosis Múltiple Recurrente-Remitente/metabolismo , Natalizumab/uso terapéutico , Proteína Hiperexpresada del Nefroblastoma/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Estudios de Cohortes , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteína Hiperexpresada del Nefroblastoma/genética , Resultado del Tratamiento
3.
Brain ; 142(3): 617-632, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30759186

RESUMEN

Elucidation of distinct T-cell subsets involved in multiple sclerosis immune-pathophysiology continues to be of considerable interest since an ultimate goal is to more selectively target the aberrant immune response operating in individual patients. While abnormalities of both effector (Teff) and regulatory (Treg) T cells have been reported in patients with multiple sclerosis, prior studies have mostly assessed average abnormalities in either limb of the immune response, rather than both at the same time, which limits the ability to evaluate the balance between effectors and regulators operating in the same patient. Assessing both phenotypic and functional responses of Teffs and Tregs has also proven important. In studies of adults with multiple sclerosis, in whom biological disease onset likely started many years prior to the immune assessments, an added challenge for any reported abnormality is whether the abnormality indeed contributes to the disease (and hence of interest to target therapeutically) or merely develops consequent to inflammatory injury (in which case efforts to develop targeted therapies are unlikely to be beneficial). Paediatric-onset multiple sclerosis, though rare, offers a unique window into early disease mechanisms. Here, we carried out a comprehensive integrated study, simultaneously assessing phenotype and functional responses of both effector and regulatory T cells in the same children with multiple sclerosis, monophasic inflammatory CNS disorders, and healthy controls, recruited as part of the multicentre prospective Canadian Pediatric Demyelinating Disease Study (CPDDS). Stringent standard operating procedures were developed and uniformly applied to procure, process and subsequently analyse peripheral blood cells using rigorously applied multi-parametric flow cytometry panels and miniaturized functional assays validated for use with cryopreserved cells. We found abnormally increased frequencies and exaggerated pro-inflammatory responses of CD8+CD161highTCR-Vα7.2+ MAIT T cells and CD4+CCR2+CCR5+ Teffs in paediatric-onset multiple sclerosis, compared to both control groups. CD4+CD25hiCD127lowFOXP3+ Tregs of children with multiple sclerosis exhibited deficient suppressive capacity, including diminished capacity to suppress disease-implicated Teffs. In turn, the implicated Teffs of multiple sclerosis patients were relatively resistant to suppression by normal Tregs. An abnormal Teff/Treg ratio at the individual child level best distinguished multiple sclerosis children from controls. We implicate abnormalities in both frequencies and functional responses of distinct pro-inflammatory CD4 and CD8 T cell subsets, as well as Treg function, in paediatric-onset multiple sclerosis, and suggest that mechanisms contributing to early multiple sclerosis development differ across individuals, reflecting an excess abnormality in either Teff or Treg limbs of the T cell response, or a combination of lesser abnormalities in both limbs.


Asunto(s)
Esclerosis Múltiple/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Adolescente , Canadá , Niño , Femenino , Humanos , Activación de Linfocitos/inmunología , Masculino , Fenotipo , Estudios Prospectivos , Subgrupos de Linfocitos T/fisiología , Linfocitos T Reguladores/fisiología
4.
Nat Immunol ; 8(12): 1372-9, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17994023

RESUMEN

Excessive inflammation occurs during infection and autoimmunity in mice lacking the alpha-subunit of the interleukin 27 (IL-27) receptor. The molecular mechanisms underlying this increased inflammation are incompletely understood. Here we report that IL-27 upregulated IL-10 in effector T cells that produced interferon-gamma and expressed the transcription factor T-bet but did not express the transcription factor Foxp3. These IFN-gamma+T-bet+Foxp3- cells resembled effector T cells that have been identified as the main source of host-protective IL-10 during inflammation. IL-27-induced production of IL-10 was associated with less secretion of IL-17, and exogenous IL-27 reduced the severity of adoptively transferred experimental autoimmune encephalomyelitis by a mechanism dependent on IL-10. Our data show that IL-27-induced production of IL-10 by effector T cells contributes to the immunomodulatory function of IL-27.


Asunto(s)
Autoinmunidad/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/inmunología , Interleucina-10/metabolismo , Interleucina-17/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T/metabolismo , Animales , Autoinmunidad/inmunología , Enfermedades del Sistema Nervioso Central/inmunología , Enfermedades del Sistema Nervioso Central/patología , Inflamación/inmunología , Interleucina-10/inmunología , Interleucina-17/farmacología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones , Linfocitos T/inmunología
5.
J Immunol ; 199(2): 435-448, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28592428

RESUMEN

The efficacy of B cell depletion therapy in multiple sclerosis indicates their central pathogenic role in disease pathogenesis. The B lymphotropic EBV is a major risk factor in multiple sclerosis, via as yet unclear mechanisms. We reported in a nonhuman primate experimental autoimmune encephalomyelitis model that an EBV-related lymphocryptovirus enables B cells to protect a proteolysis-sensitive immunodominant myelin oligodendrocyte glycoprotein (MOG) epitope (residues 40-48) against destructive processing. This facilitates its cross-presentation to autoaggressive cytotoxic MHC-E-restricted CD8+CD56+ T cells. The present study extends these observations to intact human B cells and identifies a key role of autophagy. EBV infection upregulated APC-related markers on B cells and activated the cross-presentation machinery. Although human MOG protein was degraded less in EBV-infected than in uninfected B cells, induction of cathepsin G activity by EBV led to total degradation of the immunodominant peptides MOG35-55 and MOG1-20 Inhibition of cathepsin G or citrullination of the arginine residue within an LC3-interacting region motif of immunodominant MOG peptides abrogated their degradation. Internalized MOG colocalized with autophagosomes, which can protect from destructive processing. In conclusion, EBV infection switches MOG processing in B cells from destructive to productive and facilitates cross-presentation of disease-relevant epitopes to CD8+ T cells.


Asunto(s)
Autoinmunidad , Autofagia/inmunología , Linfocitos B/inmunología , Linfocitos B/virología , Esclerosis Múltiple/inmunología , Animales , Autofagosomas/inmunología , Autofagosomas/metabolismo , Linfocitos B/metabolismo , Linfocitos T CD8-positivos/inmunología , Catepsina G/antagonistas & inhibidores , Catepsina G/genética , Catepsina G/inmunología , Catepsina G/metabolismo , Células Cultivadas , Reactividad Cruzada/inmunología , Humanos , Epítopos Inmunodominantes/química , Epítopos Inmunodominantes/inmunología , Ratones , Esclerosis Múltiple/fisiopatología , Esclerosis Múltiple/virología , Glicoproteína Mielina-Oligodendrócito/inmunología , Glicoproteína Mielina-Oligodendrócito/metabolismo
7.
J Immunol ; 197(4): 1074-88, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27412414

RESUMEN

EBV is the major infectious environmental risk factor for multiple sclerosis (MS), but the underlying mechanisms remain obscure. Patient studies do not allow manipulation in vivo. We used the experimental autoimmune encephalomyelitis (EAE) models in the common marmoset and rhesus monkey to model the association of EBV and MS. We report that B cells infected with EBV-related lymphocryptovirus (LCV) are requisite APCs for MHC-E-restricted autoaggressive effector memory CTLs specific for the immunodominant epitope 40-48 of myelin oligodendrocyte glycoprotein (MOG). These T cells drive the EAE pathogenesis to irreversible neurologic deficit. The aim of this study was to determine why LCV infection is important for this pathogenic role of B cells. Transcriptome comparison of LCV-infected B cells and CD20(+) spleen cells from rhesus monkeys shows increased expression of genes encoding elements of the Ag cross-presentation machinery (i.e., of proteasome maturation protein and immunoproteasome subunits) and enhanced expression of MHC-E and of costimulatory molecules (CD70 and CD80, but not CD86). It was also shown that altered expression of endolysosomal proteases (cathepsins) mitigates the fast endolysosomal degradation of the MOG40-48 core epitope. Finally, LCV infection also induced expression of LC3-II(+) cytosolic structures resembling autophagosomes, which seem to form an intracellular compartment where the MOG40-48 epitope is protected against proteolytic degradation by the endolysosomal serine protease cathepsin G. In conclusion, LCV infection induces a variety of changes in B cells that underlies the conversion of destructive processing of the immunodominant MOG40-48 epitope into productive processing and cross-presentation to strongly autoaggressive CTLs.


Asunto(s)
Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Reactividad Cruzada/inmunología , Encefalomielitis Autoinmune Experimental/virología , Infecciones por Herpesviridae/inmunología , Glicoproteína Mielina-Oligodendrócito/inmunología , Animales , Presentación de Antígeno/inmunología , Células Presentadoras de Antígenos/inmunología , Linfocitos B/virología , Western Blotting , Callithrix , Separación Celular , Encefalomielitis Autoinmune Experimental/inmunología , Epítopos de Linfocito T/inmunología , Técnica del Anticuerpo Fluorescente , Lymphocryptovirus , Activación de Linfocitos/inmunología , Macaca mulatta , Reacción en Cadena de la Polimerasa , Infecciones Tumorales por Virus/inmunología
8.
Mol Ther ; 25(2): 401-415, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28153091

RESUMEN

Inflammatory demyelination and axonal damage of the CNS are hallmarks of multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). Fingolimod (FTY720), the first FDA-approved oral medication for MS, suppresses acute disease but is less effective at the chronic stage, and whether it has a direct effect on neuroregeneration in MS and EAE remains unclear. Here we show that FTY720, at nanomolar concentrations, effectively protected survival of neural stem cells (NSCs) and enhanced their development into mature oligodendrocytes (OLGs) in vitro, primarily through the S1P3 and S1P5 receptors. In vivo, treatment with either FTY720 or NSCs alone had no effect on the secondary progressive stage of remitting-relapsing EAE, but a combination therapy with FTY720 and NSCs promoted significant recovery, including ameliorated clinical signs and CNS inflammatory demyelination, enhanced MBP synthesis and remyelination, inhibited axonal degeneration, and reduced astrogliosis. Moreover, FTY720 significantly improved incorporation and survival of transplanted NSCs in the CNS and drove their differentiation into more OLGs but fewer astrocytes, thus promoting remyelination and CNS repair processes in situ. Our data demonstrate a novel effect of FTY720 on NSC differentiation and remyelination, broadening its possible application to NSC-based therapy in the secondary progressive stage of MS.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Clorhidrato de Fingolimod/farmacología , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/terapia , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Ratones , Esclerosis Múltiple , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/metabolismo , Regeneración Nerviosa , Células-Madre Neurales/metabolismo , Oligodendroglía/citología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal , Trasplante de Células Madre , Resultado del Tratamiento
9.
Mult Scler ; 23(8): 1050-1055, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28406354

RESUMEN

From the early days of MS discovery, infections have been proposed as a possible cause of the disease. In the last three decades, an association between human endogenous retrovirus expression and MS has been further investigated and confirmed. Nevertheless, the role of such retroviruses in the disease needs clarification. In this review, we introduce MSRV/HERV-W and describe its association with MS. We then summarize the evidence for the involvement of MSRV/HERV-W in the aetiology and progression of MS and its possible role as biomarker and drug target. Biological mechanisms for HERV effects in MS may involve the activation of innate immune pathways by the envelope protein of MSRV (MSRVEnv). In addition to in vitro and experimental studies, further insight on how HERVs may influence immune-mediated pathology in MS may also come from the use of antiretroviral treatments in patients.


Asunto(s)
Antirretrovirales/uso terapéutico , Retrovirus Endógenos/patogenicidad , Productos del Gen env/uso terapéutico , Esclerosis Múltiple/terapia , Esclerosis Múltiple/virología , Animales , Biomarcadores , Humanos , Inmunidad Innata/inmunología , Esclerosis Múltiple/patología
10.
J Immunol ; 194(12): 5761-74, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25980006

RESUMEN

CD4(+)CD25(hi) FOXP3(+) regulatory T cells (Tregs) maintain tolerance to self-Ags. Their defective function is involved in the pathogenesis of multiple sclerosis (MS), an inflammatory demyelinating disease of the CNS. However, the mechanisms of such defective function are poorly understood. Recently, we reported that stimulation of TLR2, which is preferentially expressed by human Tregs, reduces their suppressive function and skews them into a Th17-like phenotype. In this study, we tested the hypothesis that TLR2 activation is involved in reduced Treg function in MS. We found that Tregs from MS patients expressed higher levels of TLR2 compared with healthy controls, and stimulation with the synthetic lipopeptide Pam3Cys, an agonist of TLR1/2, reduced Treg function and induced Th17 skewing in MS patient samples more than in healthy controls. These data provide a novel mechanism underlying diminished Treg function in MS. Infections that activate TLR2 in vivo (specifically through TLR1/2 heterodimers) could shift the Treg/Th17 balance toward a proinflammatory state in MS, thereby promoting disease activity and progression.


Asunto(s)
Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Receptor Toll-Like 2/metabolismo , Adulto , Estudios de Casos y Controles , Diferenciación Celular/efectos de los fármacos , Citocinas/biosíntesis , Femenino , Humanos , Inmunomodulación , Inmunofenotipificación , Lipoproteínas/farmacología , Masculino , Persona de Mediana Edad , Esclerosis Múltiple Recurrente-Remitente/inmunología , Esclerosis Múltiple Recurrente-Remitente/metabolismo , Factor de Transcripción STAT3/metabolismo , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/citología , Células Th17/citología , Receptor Toll-Like 2/agonistas , Adulto Joven
11.
Mol Ther ; 24(8): 1456-1469, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27203442

RESUMEN

Treatment of chronic neurodegenerative diseases such as multiple sclerosis (MS) remains a major challenge. Here we genetically engineer neural stem cells (NSCs) to produce a triply therapeutic cocktail comprising IL-10, NT-3, and LINGO-1-Fc, thus simultaneously targeting all mechanisms underlie chronicity of MS in the central nervous system (CNS): persistent inflammation, loss of trophic support for oligodendrocytes and neurons, and accumulation of neuroregeneration inhibitors. After transplantation, NSCs migrated into the CNS inflamed foci and delivered these therapeutic molecules in situ. NSCs transduced with one, two, or none of these molecules had no or limited effect when injected at the chronic stage of experimental autoimmune encephalomyelitis; cocktail-producing NSCs, in contrast, mediated the most effective recovery through inducing M2 macrophages/microglia, reducing astrogliosis, and promoting axonal integrity and endogenous oligodendrocyte/neuron differentiation. These engineered NSCs simultaneously target major mechanisms underlying chronicity of multiple sclerosis (MS) and encephalomyelitis (EAE), thus representing a novel and potentially effective therapy for the chronic stage of MS, for which there is currently no treatment available.


Asunto(s)
Autoinmunidad , Ingeniería Celular , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Expresión Génica , Células-Madre Neurales/metabolismo , Transgenes , Animales , Diferenciación Celular , Proliferación Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/diagnóstico , Encefalomielitis Autoinmune Experimental/terapia , Femenino , Vectores Genéticos/genética , Interleucina-10/genética , Lentivirus/genética , Macrófagos/metabolismo , Ratones , Microglía/metabolismo , Esclerosis Múltiple/genética , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Esclerosis Múltiple/terapia , Proteína Básica de Mielina/biosíntesis , Proteínas de la Mielina/metabolismo , Factores de Crecimiento Nervioso/genética , Células-Madre Neurales/citología , Neuronas/metabolismo , Neuronas/patología , Oligodendroglía/citología , Oligodendroglía/metabolismo , Trasplante de Células Madre , Transducción Genética
12.
Mediators Inflamm ; 2017: 1380615, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28356656

RESUMEN

Experimental autoimmune encephalomyelitis (EAE) is an inflammatory, demyelinating disease of the CNS that mimics human multiple sclerosis (MS), and it is thought to be driven by Th1 and Th17 myelin-reactive cells. Although adaptive immunity is clearly pivotal in the pathogenesis of EAE, with an essential role of CD4+ T cells, little is known of early, innate responses in this experimental setting. CpG-rich oligodeoxynucleotides (ODNs), typically found in microbial genomes, are potent activators of TLR9 in plasmacytoid dendritic cells (pDCs). In this study, we compared the effects of two types of CpG, namely, type A and type B, on EAE. We found that treatment with CpG type A ODN (CpG-A), known to induce high amounts of IFN-α in pDCs, significantly reduced disease severity in EAE, relative to controls (12.63 ± 1.86 versus 23.49 ± 1.46, resp.; p = 0.001). Treatment also delayed onset of neurological deficits and reduced spinal cord demyelination, while increasing the percentage of splenic regulatory (Foxp3+ CD4+) T cells. CpG-A likewise reduced the levels of IL-17 and IFN-γ in the CNS. Mechanistic insight into those events showed that CpG-A promoted a regulatory phenotype in pDCs. Moreover, adoptive transfer of pDCs isolated from CpG-A-treated mice inhibited CNS inflammation and induced disease remission in acute-phase EAE. Our data thus identify a link between TLR9 activation by specific ligands and the induction of tolerance via innate immunity mechanisms.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Inmunidad Innata , Oligodesoxirribonucleótidos/metabolismo , Animales , Células Dendríticas/citología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Endotoxinas/metabolismo , Femenino , Inflamación , Ligandos , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple , Fenotipo , Transducción de Señal , Bazo/metabolismo , Linfocitos T Reguladores/citología
13.
Pract Neurol ; 22(3): 253-255, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35027458
15.
J Neuroinflammation ; 11: 130, 2014 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-25069698

RESUMEN

BACKGROUND: Experimental autoimmune encephalomyelitis (EAE) is an animal model of autoimmune inflammatory demyelination that is mediated by Th1 and Th17 cells. The transcription factor interferon regulatory factor 3 (IRF3) is activated by pathogen recognition receptors and induces interferon-ß production. METHODS: To determine the role of IRF3 in autoimmune inflammation, we immunised wild-type (WT) and irf3(-/-) mice to induce EAE. Splenocytes from WT and irf3(-/-) mice were also activated in vitro in Th17-polarising conditions. RESULTS: Clinical signs of disease were significantly lower in mice lacking IRF3, with reduced Th1 and Th17 cells in the central nervous system. Peripheral T-cell responses were also diminished, including impaired proliferation and Th17 development in irf3(-/-) mice. Myelin-reactive CD4+ cells lacking IRF3 completely failed to transfer EAE in Th17-polarised models as did WT cells transferred into irf3(-/-) recipients. Furthermore, IRF3 deficiency in non-CD4+ cells conferred impairment of Th17 development in antigen-activated cultures. CONCLUSION: These data show that IRF3 plays a crucial role in development of Th17 responses and EAE and warrants investigation in human multiple sclerosis.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Factor 3 Regulador del Interferón/deficiencia , Animales , Linfocitos T CD4-Positivos/patología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Femenino , Citometría de Flujo , Factor 3 Regulador del Interferón/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Glicoproteína Mielina-Oligodendrócito/toxicidad , Fragmentos de Péptidos/toxicidad , Ésteres del Forbol/farmacología , Médula Espinal/patología , Linfocitos T/efectos de los fármacos , Células Th17/efectos de los fármacos , Células Th17/metabolismo , Transfección
16.
J Immunol ; 189(5): 2283-9, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22844124

RESUMEN

Short synthetic oligodeoxynucleotides (ODNs) rich in CpG or GpG motifs have been considered as potential modulators of immunity in clinical settings. In this study, we show that a synthetic GpC-ODN conferred highly suppressive activity on mouse splenic plasmacytoid dendritic cells, demonstrable in vivo in a skin test assay. The underlying mechanism involved signaling by noncanonical NF-κB family members and TGF-ß-dependent expression of the immunoregulatory enzyme IDO. Unlike CpG-ODNs, the effects of GpC-ODN required TLR7/TRIF-mediated but not TLR9/MyD88-mediated events, as do sensing of viral ssRNA and the drug imiquimod. Induction of IDO by a GpC-containing ODN could also be demonstrated in human dendritic cells, allowing those cells to assist FOXP3+ T cell generation in vitro. Among potentially therapeutic ODNs, this study identifies GpC-rich sequences as novel activators of TLR7-mediated, IDO-dependent regulatory responses.


Asunto(s)
Células Dendríticas/inmunología , Tolerancia Inmunológica/genética , Oligodesoxirribonucleótidos/farmacología , Animales , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Células Dendríticas/enzimología , Femenino , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Indolamina-Pirrol 2,3,-Dioxigenasa/biosíntesis , Interferón beta/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/genética , Oligodesoxirribonucleótidos/genética , Estructura Terciaria de Proteína , Receptores de Interleucina-1/fisiología , Transducción de Señal/inmunología , Receptor Toll-Like 7/fisiología , Transcripción Genética/inmunología , Factor de Crecimiento Transformador beta/fisiología
17.
Mult Scler Relat Disord ; 86: 105597, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38598954

RESUMEN

BACKGROUND: Epstein barr virus (EBV) infection of B cells is now understood to be one of the triggering events for the development of Multiple Sclerosis (MS), a progressive immune-mediated disease of the central nervous system. EBV infection is also linked to expression of human endogenous retroviruses (HERVs) of the HERV-W group, a further risk factor for the development of MS. Ocrelizumab is a high-potency disease-modifying treatment (DMT) for MS, which depletes B cells by targeting CD20. OBJECTIVES: We studied the effects of ocrelizumab on gene expression in peripheral blood mononuclear cells (PBMC) from paired samples from 20 patients taken prior to and 6 months after beginning ocrelizumab therapy. We hypothesised that EBV and HERV-W loads would be lower in post-treatment samples. METHODS: Samples were collected in Paxgene tubes, subject to RNA extraction and Illumina paired end short read mRNA sequencing with mapping of sequence reads to the human genome using Salmon and differential gene expression compared with DeSeq2. Mapping was also performed separately to the HERV-D database of HERV sequences and the EBV reference sequence. RESULTS: Patient samples were more strongly clustered by individual rather than disease type (relapsing/remitting or primary progressive), treatment (pre and post), age, or sex. Fourteen genes, all clearly linked to B cell function were significantly down regulated in the post treatment samples. Interestingly only one pre-treatment sample had detectable EBV RNA and there were no significant differences in HERV expression (of any group) between pre- and post-treatment samples. CONCLUSIONS: While EBV and HERV expression are clearly linked to triggering MS pathogenesis, it does not appear that high level expression of these viruses is a part of the ongoing disease process or that changes in virus load are associated with ocrelizumab treatment.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Linfocitos B , Retrovirus Endógenos , Leucocitos Mononucleares , Humanos , Retrovirus Endógenos/efectos de los fármacos , Femenino , Masculino , Adulto , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Linfocitos B/efectos de los fármacos , Anticuerpos Monoclonales Humanizados/farmacología , Persona de Mediana Edad , Factores Inmunológicos/farmacología , ARN Viral , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/virología , Esclerosis Múltiple/inmunología , Herpesvirus Humano 4 , Expresión Génica/efectos de los fármacos
18.
Pathogens ; 13(6)2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38921797

RESUMEN

Previous exposure to Epstein-Barr virus (EBV) is strongly associated with the development of multiple sclerosis (MS). By contrast, past cytomegalovirus (CMV) infection may have no association, or be negatively associated with MS. This study aimed to investigate the associations of herpesvirus infections with MS in an Italian population. Serum samples (n = 200) from Italian people with multiple sclerosis (PwMS) classified as the relapsing-and-remitting clinical phenotype and (n = 137) healthy controls (HCs) were obtained from the CRESM Biobank, Orbassano, Italy. Both PwMS and HCs samples were selected according to age group (20-39 years, and 40 or more years) and sex. EBV virus capsid antigen (VCA) IgG, EBV nucleic acid-1 antigen (EBNA-1) IgG, CMV IgG, herpes simplex virus (HSV) IgG, and varicella zoster virus (VZV) IgG testing was undertaken using commercial ELISAs. EBV VCA IgG and EBNA-1 IgG seroprevalences were 100% in PwMS and 93.4% and 92.4%, respectively, in HCs. EBV VCA IgG and EBNA-1 IgG levels were higher (p < 0.001) in PwMS compared with HCs. For PwMS, the EBNA-1 IgG levels decreased with age, particularly in females. The CMV IgG seroprevalence was 58.7% in PwMS and 62.9% in HCs. CMV IgG seroprevalence increased with age. The HSV IgG seroprevalence was 71.2% in PwMS and 70.8% in HCs. HSV IgG levels were lower (p = 0.0005) in PwMS compared with HCs. VZV IgG seroprevalence was 97.5% in PwMS and 98.5% in HCs. In the population studied, several herpesvirus infections markers may have been influenced by the age and sex of the groups studied. The lack of a negative association of MS with CMV infection, and the observation of lower levels of HSV IgG in PwMS compared with HCs are findings worthy of further investigation.

19.
J Immunol ; 187(5): 2278-90, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21775683

RESUMEN

Naturally occurring CD4(+)CD25(+)FOXP3(+) regulatory T cells suppress the activity of pathogenic T cells and prevent development of autoimmune responses. There is growing evidence that TLRs are involved in modulating regulatory T cell (Treg) functions both directly and indirectly. Specifically, TLR2 stimulation has been shown to reduce the suppressive function of Tregs by mechanisms that are incompletely understood. The developmental pathways of Tregs and Th17 cells are considered divergent and mutually inhibitory, and IL-17 secretion has been reported to be associated with reduced Treg function. We hypothesized that TLR2 stimulation may reduce the suppressive function of Tregs by regulating the balance between Treg and Th17 phenotype and function. We examined the effect of different TLR2 ligands on the suppressive functions of Tregs and found that activation of TLR1/2 heterodimers reduces the suppressive activity of CD4(+)CD25(hi)FOXP3(low)CD45RA(+) (naive) and CD4(+)CD25(hi)FOXP3(hi)CD45RA(-) (memory or effector) Treg subpopulations on CD4(+)CD25(-)FOXP3(-)CD45RA(+) responder T cell proliferation while at the same time enhancing the secretion of IL-6 and IL-17, increasing RORC, and decreasing FOXP3 expression. Neutralization of IL-6 or IL-17 abrogated Pam3Cys-mediated reduction of Treg suppressive function. We also found that, in agreement with recent observations in mouse T cells, TLR2 stimulation can promote Th17 differentiation of human T helper precursors. We conclude that TLR2 stimulation, in combination with TCR activation and costimulation, promotes the differentiation of distinct subsets of human naive and memory/effector Tregs into a Th17-like phenotype and their expansion. Such TLR-induced mechanism of regulation of Treg function could enhance microbial clearance and increase the risk of autoimmune reactions.


Asunto(s)
Diferenciación Celular/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Receptor Toll-Like 2/inmunología , Separación Celular , Citometría de Flujo , Humanos , Activación de Linfocitos/inmunología , Fenotipo , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo , Células Th17/citología , Células Th17/metabolismo , Receptor Toll-Like 2/metabolismo
20.
Mol Ther ; 20(7): 1338-48, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22434134

RESUMEN

The interleukin-17 (IL-17) cytokine family is crucial to the progression of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). It has been shown in a neuroectoderm-specific knockout study that astrocyte-restricted ablation of Act1, a key and common transcription factor for signals mediated by IL-17 family members (IL-17A, IL-17F, and IL-17C), ameliorates EAE. However, the effect of Act1 deficiency in astrocytes on ongoing disease, which is of clinical relevance for MS therapy, has not been investigated. Here we report that intracerebroventricular (i.c.v.) injection of a novel lentiviral vector (shAct1) to knockdown Act1 expression in astrocytes effectively inhibited disease progression at EAE induction, clinical onset, and peak of disease (ongoing phases), with significantly reduced numbers of infiltrating inflammatory cells and percentage of Th17 cells in the central nervous system (CNS). This was mainly due to the suppressed expression of Th17-related chemokines in astrocytes, while neurotrophic factors in the CNS and immune responses in the periphery were not affected. These results demonstrate that blocking the IL-17 pathways in astrocytes is a promising therapeutic approach for MS in a CNS-specific manner, which does not interfere with systemic immune responses, a major concern in conventional MS therapy.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Astrocitos/metabolismo , Encefalomielitis Autoinmune Experimental/terapia , Interleucina-17/metabolismo , Animales , Astrocitos/inmunología , Movimiento Celular , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/metabolismo , Quimiocinas/biosíntesis , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Terapia Genética , Vectores Genéticos , Interleucina-17/genética , Interleucina-17/inmunología , Lentivirus/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Esclerosis Múltiple/terapia , Interferencia de ARN , ARN Interferente Pequeño
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA