Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
J Neuroinflammation ; 21(1): 144, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38822334

RESUMO

Cumulative evidence has established that Interferon (IFN)-γ has both pathogenic and protective roles in Multiple Sclerosis and the animal model, Experimental Autoimmune Encephalomyelitis (EAE). However, the underlying mechanisms to the beneficial effects of IFN-γ are not well understood. In this study, we found that IFN-γ exerts therapeutic effects on chronic, relapsing-remitting, and chronic progressive EAE models. The frequency of regulatory T (Treg) cells in spinal cords from chronic EAE mice treated with IFN-γ was significantly increased with no effect on Th1 and Th17 cells. Consistently, depletion of FOXP3-expressing cells blocked the protective effects of IFN-γ, indicating that the therapeutic effect of IFN-γ depends on the presence of Treg cells. However, IFN-γ did not trigger direct in vitro differentiation of Treg cells. In vivo administration of blocking antibodies against either interleukin (IL)-10, transforming growth factor (TGF)-ß or program death (PD)-1, revealed that the protective effects of IFN-γ in EAE were also dependent on TGF-ß and PD-1, but not on IL-10, suggesting that IFN-γ might have an indirect role on Treg cells acting through antigen-presenting cells. Indeed, IFN-γ treatment increased the frequency of a subset of splenic CD11b+ myeloid cells expressing TGF-ß-Latency Associated Peptide (LAP) and program death ligand 1 (PD-L1) in a signal transducer and activator of transcription (STAT)-1-dependent manner. Furthermore, splenic CD11b+ cells from EAE mice preconditioned in vitro with IFN-γ and myelin oligodendrocyte glycoprotein (MOG) peptide exhibited a tolerogenic phenotype with the capability to induce conversion of naïve CD4+ T cells mediated by secretion of TGF-ß. Remarkably, adoptive transfer of splenic CD11b+ cells from IFN-γ-treated EAE mice into untreated recipient mice ameliorated clinical symptoms of EAE and limited central nervous system infiltration of mononuclear cells and effector helper T cells. These results reveal a novel cellular and molecular mechanism whereby IFN-γ promotes beneficial effects in EAE by endowing splenic CD11b+ myeloid cells with tolerogenic and therapeutic activities.


Assuntos
Antígeno CD11b , Encefalomielite Autoimune Experimental , Interferon gama , Camundongos Endogâmicos C57BL , Células Mieloides , Baço , Animais , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Camundongos , Interferon gama/metabolismo , Células Mieloides/efeitos dos fármacos , Células Mieloides/imunologia , Células Mieloides/metabolismo , Baço/imunologia , Antígeno CD11b/metabolismo , Feminino , Glicoproteína Mielina-Oligodendrócito/toxicidade , Glicoproteína Mielina-Oligodendrócito/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Fragmentos de Peptídeos/toxicidade , Fragmentos de Peptídeos/farmacologia , Fator de Crescimento Transformador beta/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Receptor de Morte Celular Programada 1/imunologia , Fatores de Transcrição Forkhead/metabolismo , Modelos Animais de Doenças
2.
Brain ; 147(4): 1344-1361, 2024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-37931066

RESUMO

Neuromyelitis optica spectrum disorder (NMOSD) is a CNS autoimmune inflammatory disease mediated by T helper 17 (Th17) and antibody responses to the water channel protein, aquaporin 4 (AQP4), and associated with astrocytopathy, demyelination and axonal loss. Knowledge about disease pathogenesis is limited and the search for new therapies impeded by the absence of a reliable animal model. In our work, we determined that NMOSD is characterized by decreased IFN-γ receptor signalling and that IFN-γ depletion in AQP4201-220-immunized C57BL/6 mice results in severe clinical disease resembling human NMOSD. Pathologically, the disease causes autoimmune astrocytic and CNS injury secondary to cellular and humoral inflammation. Immunologically, the absence of IFN-γ allows for increased expression of IL-6 in B cells and activation of Th17 cells, and generation of a robust autoimmune inflammatory response. Consistent with NMOSD, the experimental disease is exacerbated by administration of IFN-ß, whereas repletion of IFN-γ, as well as therapeutic targeting of IL-17A, IL-6R and B cells, ameliorates it. We also demonstrate that immune tolerization with AQP4201-220-coupled poly(lactic-co-glycolic acid) nanoparticles could both prevent and effectively treat the disease. Our findings enhance the understanding of NMOSD pathogenesis and provide a platform for the development of immune tolerance-based therapies, avoiding the limitations of the current immunosuppressive therapies.


Assuntos
Neuromielite Óptica , Humanos , Animais , Camundongos , Neuromielite Óptica/patologia , Aquaporina 4 , Interferon gama/metabolismo , Camundongos Endogâmicos C57BL , Linfócitos B , Autoanticorpos/metabolismo
3.
Front Immunol ; 14: 1191838, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37334380

RESUMO

Compelling evidence has shown that interferon (IFN)-γ has dual effects in multiple sclerosis and in its animal model of experimental autoimmune encephalomyelitis (EAE), with results supporting both a pathogenic and beneficial function. However, the mechanisms whereby IFN-γ may promote neuroprotection in EAE and its effects on central nervous system (CNS)-resident cells have remained an enigma for more than 30 years. In this study, the impact of IFN-γ at the peak of EAE, its effects on CNS infiltrating myeloid cells (MC) and microglia (MG), and the underlying cellular and molecular mechanisms were investigated. IFN-γ administration resulted in disease amelioration and attenuation of neuroinflammation associated with significantly lower frequencies of CNS CD11b+ myeloid cells and less infiltration of inflammatory cells and demyelination. A significant reduction in activated MG and enhanced resting MG was determined by flow cytometry and immunohistrochemistry. Primary MC/MG cultures obtained from the spinal cord of IFN-γ-treated EAE mice that were ex vivo re-stimulated with a low dose (1 ng/ml) of IFN-γ and neuroantigen, promoted a significantly higher induction of CD4+ regulatory T (Treg) cells associated with increased transforming growth factor (TGF)-ß secretion. Additionally, IFN-γ-treated primary MC/MG cultures produced significantly lower nitrite in response to LPS challenge than control MC/MG. IFN-γ-treated EAE mice had a significantly higher frequency of CX3CR1high MC/MG and expressed lower levels of program death ligand 1 (PD-L1) than PBS-treated mice. Most CX3CR1highPD-L1lowCD11b+Ly6G- cells expressed MG markers (Tmem119, Sall2, and P2ry12), indicating that they represented an enriched MG subset (CX3CR1highPD-L1low MG). Amelioration of clinical symptoms and induction of CX3CR1highPD-L1low MG by IFN-γ were dependent on STAT-1. RNA-seq analyses revealed that in vivo treatment with IFN-γ promoted the induction of homeostatic CX3CR1highPD-L1low MG, upregulating the expression of genes associated with tolerogenic and anti-inflammatory roles and down-regulating pro-inflammatory genes. These analyses highlight the master role that IFN-γ plays in regulating microglial activity and provide new insights into the cellular and molecular mechanisms involved in the therapeutic activity of IFN-γ in EAE.


Assuntos
Encefalomielite Autoimune Experimental , Camundongos , Animais , Microglia/metabolismo , Interferon gama/metabolismo , Antígeno B7-H1/metabolismo , Sistema Nervoso Central
4.
Front Neurol ; 12: 783304, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34987468

RESUMO

Neuromyelitis optica (NMO) is a chronic inflammatory disease of the central nervous system that primarily affects the optic nerves and spinal cord of patients, and in some instances their brainstem, diencephalon or cerebrum as spectrum disorders (NMOSD). Clinical and basic science knowledge of NMO has dramatically increased over the last two decades and it has changed the perception of the disease as being inevitably disabling or fatal. Nonetheless, there is still no cure for NMO and all the disease-modifying therapies (DMTs) are only partially effective. Furthermore, DMTs are not disease- or antigen-specific and alter all immune responses including those protective against infections and cancer and are often associated with significant adverse reactions. In this review, we discuss the pathogenic mechanisms of NMO as they pertain to its DMTs and immune tolerance. We also examine novel research therapeutic strategies focused on induction of antigen-specific immune tolerance by administrating tolerogenic immune-modifying nanoparticles (TIMP). Development and implementation of immune tolerance-based therapies in NMO is likely to be an important step toward improving the treatment outcomes of the disease. The antigen-specificity of these therapies will likely ameliorate the disease safely and effectively, and will also eliminate the clinical challenges associated with chronic immunosuppressive therapies.

5.
J Comp Psychol ; 127(2): 179-93, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23088648

RESUMO

This series of experiments evaluates the nature of the representation that mediates human (Homo sapiens) and rat (Rattus norvegicus) movement characteristics on analogous spatial learning tasks. The results of Experiment 1 demonstrated that self-movement cues were sufficient to guide the performance of human participants during place training and matching-to-place testing tasks adapted to tabletop or manipulatory scale. Experiment 2 investigated the effect of manipulating access to environmental cues during place training on the nature of the representation used to guide performance. Blindfolded human participants appeared to encode the absolute location of the goal, whereas participants with access to environmental cues appeared to encode the relative location of the goal. The results of Experiment 3 demonstrated that human participants with access to environmental cues exhibited a similar response tendency (as observed in Experiment 2) after half as many trials of place training. During Experiment 4, rats exhibited movement characteristics in the water maze that were similar to movement characteristics observed in human participants who were provided access to environmental cues. These observations provide evidence that direction and distance estimation processes mediate performance on spatial tasks that are conserved across humans and rats.


Assuntos
Comportamento Animal/fisiologia , Cinestesia/fisiologia , Aprendizagem em Labirinto/fisiologia , Percepção Espacial/fisiologia , Adolescente , Adulto , Animais , Fenômenos Biomecânicos/fisiologia , Sinais (Psicologia) , Percepção de Distância/fisiologia , Feminino , Humanos , Masculino , Ratos , Ratos Long-Evans , Adulto Jovem
6.
J Immunol ; 189(9): 4602-11, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23008451

RESUMO

Neuromyelitis optica (NMO) is a chronic inflammatory disease of the CNS that is mediated, in part, by a self-reactive Ab against the astrocyte aquaporin-4 protein. In the current study, we examined the possibility and the biological significance of cross-immunoreactivity between bacterial aquaporin-Z and human aquaporin-4 proteins. Sequence-alignment analysis of these proteins revealed several regions of significant structural homology. Some of the homologous regions were also found to overlap with important immune and disease-relevant epitopes. Cross-immunoreactivity between aquaporin-Z and aquaporin-4 was investigated and ascertained in multiple immune-based assays using sera from patients with neuromyelitis optica, immune mouse serum, and Abs raised against aquaporin-Z. The biological significance of this phenomenon was established in series of experiments demonstrating that induction of an immune response against aquaporin-Z or its homologous regions can also trigger an autoimmune reaction against aquaporin-4 and inflammation of the CNS. Our study indicates that the autoimmune response against aquaporin-4 in neuromyelitis optica may be triggered by infection-induced cross-immunoreactivity and presents a new perspective on the pathogenesis of this disease.


Assuntos
Aquaporina 4/metabolismo , Aquaporinas/metabolismo , Proteínas de Escherichia coli/metabolismo , Neuromielite Óptica/imunologia , Neuromielite Óptica/microbiologia , Sequência de Aminoácidos , Animais , Aquaporina 4/genética , Aquaporinas/genética , Aquaporinas/imunologia , Células Cultivadas , Reações Cruzadas/imunologia , Testes Imunológicos de Citotoxicidade/métodos , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/imunologia , Feminino , Células HEK293 , Humanos , Imunoglobulina G/administração & dosagem , Imunoglobulina G/toxicidade , Camundongos , Dados de Sequência Molecular , Neuromielite Óptica/metabolismo , Coelhos , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Homologia Estrutural de Proteína
7.
Rev Neurosci ; 23(2): 145-52, 2012 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-22499673

RESUMO

Oligodendrocyte injury and inflammatory demyelination are key pathological abnormalities of multiple sclerosis (MS), and its animal model, i.e., the experimental autoimmune encephalomyelitis (EAE). Traditionally, they are viewed as destructive processes secondary to a dysregulated autoimmune reaction. New evidence emerged over the last decade indicating that oligodendrocytes are not merely immune targets but rather active participants in the neuroimmune network and, in fact, can regulate the events leading to inflammatory demyelination. In this review, we are discussing the role of interferon regulatory factor 1 (IRF-1) as a master transcription factor orchestrating oligodendrocyte injury and inflammatory demyelination in MS and EAE. We are also discussing the significance of IRF-1 signaling in the induction of oligodendrocyte pyroptosis, a Caspase 1-dependent pro-inflammatory cell death, as a disease-enhancing mechanism. Finally, we are drawing attention to IRF-1 as a potential therapeutic target in MS and to the importance of investigating other oligodendrocyte-dependent disease mechanisms.


Assuntos
Encefalomielite Autoimune Experimental/metabolismo , Fator Regulador 1 de Interferon/metabolismo , Bainha de Mielina/metabolismo , Oligodendroglia/metabolismo , Animais , Caspase 1/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Humanos , Fator Regulador 1 de Interferon/deficiência , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Esclerose Múltipla/imunologia , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Bainha de Mielina/patologia , Oligodendroglia/imunologia , Oligodendroglia/patologia , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA