Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Front Immunol ; 12: 635257, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33679788

RESUMO

Multiple sclerosis (MS) is an increasingly prevalent progressive autoimmune and debilitating chronic disease that involves the detrimental recognition of central nervous system (CNS) antigens by the immune system. Although significant progress has been made in the last decades on the biology of MS and the identification of novel therapies to treat its symptoms, the etiology of this disease remains unknown. However, recent studies have suggested that viral infections may contribute to disease onset. Interestingly, a potential association between herpes simplex virus type 1 (HSV-1) infection and MS has been reported, yet a direct relationship among both has not been conclusively demonstrated. Experimental autoimmune encephalomyelitis (EAE) recapitulates several aspects of MS in humans and is widely used to study this disease. Here, we evaluated the effect of asymptomatic brain infection by HSV-1 on the onset and severity of EAE in C57BL/6 mice. We also evaluated the effect of infection with an HSV-1-mutant that is attenuated in neurovirulence and does not cause encephalitis. Importantly, we observed more severe EAE in mice previously infected either, with the wild-type (WT) or the mutant HSV-1, as compared to uninfected control mice. Also, earlier EAE onset was seen after WT virus inoculation. These findings support the notion that a previous exposure to HSV-1 can accelerate and enhance EAE, which suggests a potential contribution of asymptomatic HSV-1 to the onset and severity of MS.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Herpes Simples/imunologia , Herpesvirus Humano 1/imunologia , Animais , Anticorpos Antivirais/sangue , Doenças Assintomáticas , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/virologia , Permeabilidade Capilar , Citocinas/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/virologia , Feminino , Herpes Simples/genética , Herpes Simples/metabolismo , Herpes Simples/virologia , Herpesvirus Humano 1/patogenicidade , Mediadores da Inflamação/metabolismo , Camundongos Endogâmicos C57BL , Mutação , Índice de Gravidade de Doença , Fatores de Tempo , Virulência
2.
Front Immunol ; 11: 584297, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33329556

RESUMO

Multiple sclerosis (MS) is caused by a combination of genetic and environmental factors. It is believed that previous infection with Epstein Barr Virus (EBV) plays an important role in the development of MS. Previously, we developed a murine model where latent infection with gamma herpesvirus 68 (γHV-68), a murine homolog to EBV, enhanced the symptoms of experimental autoimmune encephalomyelitis (EAE), resulting in disease that more closely resembles MS in humans. Here, we explored the conditions that were necessary for EAE enhancement. We showed that latently infected CD19+IgD- B cells were capable of enhancing EAE symptoms when transferred from mice previously infected with γHV-68 into uninfected mice. We also observed a prevention of enhancement when B cells were depleted before infection. However, depletion after the establishment of latency only partially reduced EAE. This indicated the existence of a mechanism where B cells play an important role as antigen presenting cells (APCs) prior to EAE induction for the priming of Th1 cells. It is possible that these signals persist even after B cell depletion, strongly suggesting a paracrine signaling modulation of non-B cell APCs. These results strongly support the concept that EBV contributes to the development of autoimmunity and highlights the need for a vaccine against EBV that could limit or prevent multiple sclerosis development.


Assuntos
Linfócitos B/imunologia , Infecções por Vírus Epstein-Barr/imunologia , Herpesvirus Humano 4/imunologia , Esclerose Múltipla/imunologia , Esclerose Múltipla/virologia , Animais , Células Apresentadoras de Antígenos/imunologia , Autoimunidade/imunologia , Encéfalo/imunologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/virologia , Camundongos , Camundongos Endogâmicos C57BL , Comunicação Parácrina/imunologia , Transdução de Sinais/imunologia , Células Th1/imunologia
3.
Immunobiology ; 225(2): 151883, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31818507

RESUMO

Prior work demonstrated that epithelial V-like antigen (EVA), a cell surface adhesion molecule, is expressed in B lymphocytes and is necessary for the efficacy of anti-alpha4 integrin treatment of experimental autoimmune encephalomyelitis (EAE), the mouse model of human multiple sclerosis. EVA deficiency is associated with a severe clinical phenotype of EAE in the presence or absence of treatment. Histological analysis revealed enhanced B cell-mediated autoimmunity and deposition of antibody and complement within the brain and spinal cord. Here our goal was to determine the molecular mechanism of EVA regulation of B lymphocyte function. Analysis of bone marrow from MOG-immunized mice revealed increased expansion of CD11c+ B cells in EVA-deficient mice as compared to wild type controls. In vitro studies of mouse bone marrow B lymphocytes revealed enhanced proliferation of the CD11c+ population in response to the Tlr7/8 agonist R848. An increase in R848-induced proliferation of CD11c+ B cells was also seen in vitro in Daudi cells, a human B cell line, following knockdown of the mpzl2 gene that encodes EVA. These mechanisms were characterized further by global expression analysis of bone marrow from immunized EVA-deficient and wild type control mice. These data revealed increased expression of B cell associated genes and decreased expression of the anti-viral oligoadenylate synthase genes, Oas1 and Oas2, in the knockout condition. In Daudi cells, R848 treatment induced an increase in Oas2 expression in control cells that was not observed in EVA-deficient cells. EVA deficiency also was associated with increased transcription of an Epstein-Barr virus gene during lytic replication. These results suggest EVA expression and signaling prevent expansion of CD11c+ B lymphocytes, a cellular phenotype associated with autoimmunity, increase expression of anti-viral oligoadenylate synthase genes, and reduce replication of a DNA virus.


Assuntos
Antivirais/imunologia , Linfócitos B/imunologia , Antígeno CD11c/imunologia , Animais , Autoimunidade/imunologia , Linfócitos B/virologia , Medula Óssea/imunologia , Medula Óssea/virologia , Encéfalo/imunologia , Encéfalo/virologia , Moléculas de Adesão Celular/imunologia , Proliferação de Células/fisiologia , Células Cultivadas , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/virologia , Herpesvirus Humano 4/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Esclerose Múltipla/imunologia , Esclerose Múltipla/virologia , Transdução de Sinais/imunologia , Medula Espinal/imunologia , Medula Espinal/virologia
4.
Sci Rep ; 9(1): 10869, 2019 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-31350464

RESUMO

Patients with multiple sclerosis (MS) develop a variety of lower urinary tract symptoms (LUTS). We previously characterized a murine model of neurogenic bladder dysfunction induced by a neurotropic strain of a coronavirus. In the present study, we further study the role of long-lasting neurodegeneration on the development of neurogenic bladder dysfunction in mice with corona-virus induced encephalitis (CIE). Long-term follow up study revealed three phenotypes of neurodegenerative symptom development: recovery (REC group), chronic progression (C-PRO group) and chronic disease with relapsing-remitting episodes (C-RELAP group). The levels of IL-1ß in REC group, IL-10 in C-RELAP group, and IL-1ß, IL-6, IL-10 and TNF-α in C-PRO group were diminished in the brain. The levels of TNF-α in REC group and INF-γ, IL-2, TGF-ß and TNF-α in the C-PRO group were also diminished in the urinary bladder. Mice in C-RELAP group showed a delayed recovery of voiding function. In vitro contractility studies determined a decreased basal detrusor tone and reduced amplitude of nerve-mediated contractions in C-RELAP group, whereas C-PRO group had elevated muscle-mediated contractions. In conclusion, mice with CIE developed three phenotypes of neurologic impairment mimicking different types of MS progression in humans and showed differential mechanisms driving neurogenic bladder dysfunction.


Assuntos
Infecções por Coronavirus/fisiopatologia , Encefalomielite Autoimune Experimental/fisiopatologia , Sintomas do Trato Urinário Inferior/etiologia , Esclerose Múltipla/complicações , Vírus da Hepatite Murina , Fenótipo , Bexiga Urinaria Neurogênica/etiologia , Animais , Infecções por Coronavirus/virologia , Citocinas/análise , Citocinas/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/virologia , Ensaio de Imunoadsorção Enzimática , Seguimentos , Peptídeos e Proteínas de Sinalização Intercelular/análise , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Sintomas do Trato Urinário Inferior/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/fisiopatologia , Bexiga Urinaria Neurogênica/metabolismo
5.
Proc Natl Acad Sci U S A ; 115(44): 11292-11297, 2018 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-30322946

RESUMO

Pathogens, particularly human herpesviruses (HHVs), are implicated as triggers of disease onset/progression in multiple sclerosis (MS) and other neuroinflammatory disorders. However, the time between viral acquisition in childhood and disease onset in adulthood complicates the study of this association. Using nonhuman primates, we demonstrate that intranasal inoculations with HHV-6A and HHV-6B accelerate an MS-like neuroinflammatory disease, experimental autoimmune encephalomyelitis (EAE). Although animals inoculated intranasally with HHV-6 (virus/EAE marmosets) were asymptomatic, they exhibited significantly accelerated clinical EAE compared with control animals. Expansion of a proinflammatory CD8 subset correlated with post-EAE survival in virus/EAE marmosets, suggesting that a peripheral (viral?) antigen-driven expansion may have occurred post-EAE induction. HHV-6 viral antigen in virus/EAE marmosets was markedly elevated and concentrated in brain lesions, similar to previously reported localizations of HHV-6 in MS brain lesions. Collectively, we demonstrate that asymptomatic intranasal viral acquisition accelerates subsequent neuroinflammation in a nonhuman primate model of MS.


Assuntos
Herpesvirus Humano 6/patogenicidade , Inflamação/virologia , Esclerose Múltipla/virologia , Primatas/virologia , Animais , Encéfalo/virologia , Callithrix , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/virologia , Feminino , Masculino , Infecções por Roseolovirus/virologia
6.
Brain ; 141(6): 1637-1649, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29688408

RESUMO

Multiple sclerosis is an inflammatory demyelinating disease of the central nervous system. Although it has been extensively studied, the proximate trigger of the immune response remains uncertain. Experimental autoimmune encephalomyelitis in the common marmoset recapitulates many radiological and pathological features of focal multiple sclerosis lesions in the cerebral white matter, unlike traditional experimental autoimmune encephalomyelitis in rodents. This provides an opportunity to investigate how lesions form as well as the relative timing of factors involved in lesion pathogenesis, especially during early stages of the disease. We used MRI to track experimental autoimmune encephalomyelitis lesions in vivo to determine their age, stage of development, and location, and we assessed the corresponding histopathology post-mortem. We focused on the plasma protein fibrinogen-a marker for blood-brain barrier leakage that has also been linked to a pathogenic role in inflammatory demyelinating lesion development. We show that fibrinogen has a specific spatiotemporal deposition pattern, apparently deriving from the central vein in early experimental autoimmune encephalomyelitis lesions <6 weeks old, and preceding both demyelination and visible gadolinium enhancement on MRI. Thus, fibrinogen leakage is one of the earliest detectable events in lesion pathogenesis. In slightly older lesions, fibrinogen is found inside microglia/macrophages, suggesting rapid phagocytosis. Quantification demonstrates positive correlation of fibrinogen deposition with accumulation of inflammatory cells, including microglia/macrophages and T cells. The peak of fibrinogen deposition coincides with the onset of demyelination and axonal loss. In samples from chronic multiple sclerosis cases, fibrinogen was found at the edge of chronic active lesions, which have ongoing demyelination and inflammation, but not in inactive lesions, suggesting that fibrinogen may play a role in sustained inflammation even in the chronic setting. In summary, our data support the notion that fibrinogen is a key player in the early pathogenesis, as well as sustained inflammation, of inflammatory demyelinating lesions.


Assuntos
Encéfalo/metabolismo , Encefalomielite Autoimune Experimental/patologia , Fibrinogênio/metabolismo , Esclerose Múltipla/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Axônios/metabolismo , Axônios/patologia , Encéfalo/diagnóstico por imagem , Proteínas de Ligação ao Cálcio , Callithrix , Citocinas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Encefalomielite Autoimune Experimental/diagnóstico por imagem , Encefalomielite Autoimune Experimental/virologia , Feminino , Regulação da Expressão Gênica/fisiologia , Herpesviridae , Humanos , Filamentos Intermediários/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Proteínas dos Microfilamentos , Esclerose Múltipla/diagnóstico por imagem , Esclerose Múltipla/virologia , Bainha de Mielina/metabolismo , Bainha de Mielina/patologia , Fator de Transcrição 2 de Oligodendrócitos/metabolismo , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Fatores de Transcrição/metabolismo
7.
PLoS Pathog ; 13(1): e1006120, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28046066

RESUMO

Human T-cell leukemia virus type 1 (HTLV-1) causes adult T-cell leukemia-lymphoma (ATL) and inflammatory diseases. To enhance cell-to-cell transmission of HTLV-1, the virus increases the number of infected cells in vivo. HTLV-1 bZIP factor (HBZ) is constitutively expressed in HTLV-1 infected cells and ATL cells and promotes T-cell proliferation. However, the detailed mechanism by which it does so remains unknown. Here, we show that HBZ enhances the proliferation of expressing T cells after stimulation via the T-cell receptor. HBZ promotes this proliferation by influencing the expression and function of multiple co-inhibitory receptors. HBZ suppresses the expression of BTLA and LAIR-1 in HBZ expressing T cells and ATL cells. Expression of T cell immunoglobulin and ITIM domain (TIGIT) and Programmed cell death 1 (PD-1) was enhanced, but their suppressive effect on T-cell proliferation was functionally impaired. HBZ inhibits the co-localization of SHP-2 and PD-1 in T cells, thereby leading to impaired inhibition of T-cell proliferation and suppressed dephosphorylation of ZAP-70 and CD3ζ. HBZ does this by interacting with THEMIS, which associates with Grb2 and SHP-2. Thus, HBZ interacts with the SHP containing complex, impedes the suppressive signal from PD-1 and TIGIT, and enhances the proliferation of T cells. Although HBZ was present in both the nucleus and the cytoplasm of T cells, HBZ was localized largely in the nucleus by suppressed expression of THEMIS by shRNA. This indicates that THEMIS is responsible for cytoplasmic localization of HBZ in T cells. Since THEMIS is expressed only in T-lineage cells, HBZ mediated inhibition of the suppressive effects of co-inhibitory receptors accounts for how HTLV-1 induces proliferation only of T cells in vivo. This study reveals that HBZ targets co-inhibitory receptors to cause the proliferation of infected cells.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Proliferação de Células/fisiologia , Infecções por HTLV-I/transmissão , Vírus Linfotrópico T Tipo 1 Humano/patogenicidade , Proteínas/metabolismo , Proteínas dos Retroviridae/metabolismo , Linfócitos T/imunologia , Animais , Fatores de Transcrição de Zíper de Leucina Básica/genética , Complexo CD3/metabolismo , Linhagem Celular Tumoral , Encefalomielite Autoimune Experimental/virologia , Proteína Adaptadora GRB2/metabolismo , Infecções por HTLV-I/virologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Células Jurkat , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptor de Morte Celular Programada 1/biossíntese , Receptor de Morte Celular Programada 1/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Receptores Imunológicos/biossíntese , Receptores Imunológicos/metabolismo , Proteínas dos Retroviridae/genética , Proteína-Tirosina Quinase ZAP-70/metabolismo
8.
Trends Mol Med ; 22(12): 1012-1024, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27836419

RESUMO

Multiple sclerosis (MS) is thought to be initiated by the interaction of genetic and environmental factors, eliciting an autoimmune attack on the central nervous system. Epstein-Barr virus (EBV) is the strongest infectious risk factor, but an explanation for the paradox between high infection prevalence and low MS incidence remains elusive. We discuss new data using marmosets with experimental autoimmune encephalomyelitis (EAE) - a valid primate model of MS. The findings may help to explain how a common infection can contribute to the pathogenesis of MS. We propose that EBV infection induces citrullination of peptides in conjunction with autophagy during antigen processing, endowing B cells with the capacity to cross-present autoantigen to CD8+CD56+ T cells, thereby leading to MS progression.


Assuntos
Encefalomielite Autoimune Experimental/virologia , Infecções por Vírus Epstein-Barr/complicações , Herpesvirus Humano 4/imunologia , Esclerose Múltipla/virologia , Animais , Apresentação de Antígeno , Linfócitos B/imunologia , Linfócitos B/patologia , Linfócitos B/virologia , Citrulina/imunologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Infecções por Vírus Epstein-Barr/imunologia , Infecções por Vírus Epstein-Barr/patologia , Infecções por Vírus Epstein-Barr/virologia , Humanos , Imunidade Celular , Complexo Principal de Histocompatibilidade , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia , Linfócitos T/imunologia , Linfócitos T/patologia , Linfócitos T/virologia
9.
J Immunol ; 197(4): 1074-88, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27412414

RESUMO

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.


Assuntos
Linfócitos B/imunologia , Linfócitos T CD8-Positivos/imunologia , Apresentação Cruzada/imunologia , Encefalomielite Autoimune Experimental/virologia , Infecções por Herpesviridae/imunologia , Glicoproteína Mielina-Oligodendrócito/imunologia , Animais , Apresentação de Antígeno/imunologia , Células Apresentadoras de Antígenos/imunologia , Linfócitos B/virologia , Western Blotting , Callithrix , Separação Celular , Encefalomielite Autoimune Experimental/imunologia , Epitopos de Linfócito T/imunologia , Imunofluorescência , Lymphocryptovirus , Ativação Linfocitária/imunologia , Macaca mulatta , Reação em Cadeia da Polimerase , Infecções Tumorais por Vírus/imunologia
10.
J Immunol ; 194(4): 1454-66, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25589073

RESUMO

Costimulatory and coinhibitory receptor-ligand pairs on T cells and APC control the immune response. We have investigated whether selective blockade of CD28-CD80/86 costimulatory interactions, which preserves the coinhibitory CTLA4-CD80/86 interactions and the function of regulatory T (Treg) cells, abrogates the induction of experimental autoimmune encephalomyelitis (EAE) in rhesus monkeys. EAE was induced by intracutaneous immunization with recombinant human myelin oligodendrocyte glycoprotein (rhMOG) in CFA on day 0. FR104 is a monovalent, PEGylated-humanized Fab' Ab fragment against human CD28, cross-reactive with rhesus monkey CD28. FR104 or placebo was administered on days 0, 7, 14, and 21. FR104 levels remained high until the end of the study (day 42). Placebo-treated animals all developed clinical EAE between days 12 and 27. FR104-treated animals did not develop clinical EAE and were sacrificed at the end of the study resulting in a significantly prolonged survival. FR104 treatment diminished T and B cell responses against rhMOG, significantly reduced CNS inflammation and prevented demyelination. The inflammatory profile in the cerebrospinal fluid and brain material was also strongly reduced. Recrudescence of latent virus was investigated in blood, spleen, and brain. No differences between groups were observed for the ß-herpesvirus CMV and the polyomaviruses SV40 and SA12. Cross-sectional measurement of lymphocryptovirus, the rhesus monkey EBV, demonstrated elevated levels in the blood of FR104-treated animals. Blocking rhesus monkey CD28 with FR104 mitigated autoreactive T and B cell activation and prevented CNS pathology in the rhMOG/CFA EAE model in rhesus monkeys.


Assuntos
Antígenos CD28/antagonistas & inibidores , Encefalomielite Autoimune Experimental/imunologia , Ativação Linfocitária/imunologia , Linfócitos T/imunologia , Animais , Linfócitos B , Encefalomielite Autoimune Experimental/virologia , Humanos , Macaca mulatta , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Recombinantes/imunologia , Viroses/complicações , Latência Viral
11.
Biomed Res Int ; 2013: 589048, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24083230

RESUMO

Multiple sclerosis (MS) is an inflammatory demyelinating disease of the human central nervous system (CNS). Neurotropic demyelinating strain of MHV (MHV-A59 or its isogenic recombinant strain RSA59) induces MS-like disease in mice mediated by microglia, along with a small population of T cells. The mechanism of demyelination is at least in part due to microglia-mediated myelin stripping, with some direct axonal injury. Immunization with myelin oligodendrocyte glycoprotein (MOG) induces experimental autoimmune encephalomyelitis (EAE), a mainly CD4(+) T-cell-mediated disease, although CD8(+) T cells may play a significant role in demyelination. It is possible that both autoimmune and nonimmune mechanisms such as direct viral toxicity may induce MS. Our study directly compares CNS pathology in autoimmune and viral-induced MS models. Mice with viral-induced and EAE demyelinating diseases demonstrated similar patterns and distributions of demyelination that accumulated over the course of the disease. However, significant differences in acute inflammation were noted. Inflammation was restricted mainly to white matter at all times in EAE, whereas inflammation initially largely involved gray matter in acute MHV-induced disease and then is subsequently localized only in white matter in the chronic disease phase. The presence of dual mechanisms of demyelination may be responsible for the failure of immunosuppression to promote long-term remission in many MS patients.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/virologia , Inflamação/patologia , Esclerose Múltipla/imunologia , Esclerose Múltipla/virologia , Vírus da Hepatite Murina/fisiologia , Animais , Encéfalo/patologia , Encéfalo/virologia , Doenças Desmielinizantes/complicações , Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/patologia , Feminino , Humanos , Inflamação/complicações , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/complicações , Esclerose Múltipla/patologia , Vírus da Hepatite Murina/patogenicidade , Medula Espinal/patologia , Virulência , Replicação Viral
12.
Am J Pathol ; 183(5): 1390-1396, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24091251

RESUMO

The polyphenol compound resveratrol is reported to have multiple functions, including neuroprotection, and no major adverse effects have been reported. Although the neuroprotective effects have been associated with sirtuin 1 activation by resveratrol, the mechanisms by which resveratrol exerts such functions are a matter of controversy. We examined whether resveratrol can be neuroprotective in two models of multiple sclerosis: experimental autoimmune encephalomyelitis (EAE) and Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD). EAE was induced in C57BL/6 mice, which were fed a control diet or a diet containing resveratrol during either the induction or effector phase or through the whole course of EAE. SJL/J mice were infected with TMEV and fed a control diet or a diet containing resveratrol during the chronic phase of TMEV-IDD. In EAE, all groups of mice treated with resveratrol had more severe clinical signs than the control group. In particular, resveratrol treatment during the induction phase resulted in the most severe EAE, both clinically and histologically. Similarly, in the viral model, the mice treated with resveratrol developed significantly more severe TMEV-IDD than the control group. Thus, surprisingly, the resveratrol treatment significantly exacerbated demyelination and inflammation without neuroprotection in the central nervous system in both models. Our findings indicate that caution should be exercised in potential therapeutic applications of resveratrol in human inflammatory demyelinating diseases, including multiple sclerosis.


Assuntos
Autoimunidade/efeitos dos fármacos , Progressão da Doença , Esclerose Múltipla/patologia , Esclerose Múltipla/virologia , Estilbenos/efeitos adversos , Theilovirus/fisiologia , Animais , Axônios/efeitos dos fármacos , Axônios/patologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/complicações , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/virologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/complicações , Esclerose Múltipla/imunologia , Glicoproteína Mielina-Oligodendrócito/imunologia , Degeneração Neural/complicações , Degeneração Neural/imunologia , Degeneração Neural/patologia , Degeneração Neural/virologia , Fármacos Neuroprotetores/efeitos adversos , Resveratrol , Theilovirus/patogenicidade , Virulência
13.
J Neuroimmunol ; 259(1-2): 47-54, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23602715

RESUMO

Viral infections can exacerbate multiple sclerosis (MS) through poorly defined mechanisms. We developed an experimental system whereby infection with an asymptomatic neurotropic alphavirus caused a transient acceleration of experimental autoimmune encephalomyelitis (EAE) without altering the expansion or differentiation of autoreactive CD4+ T cells. Instead, this effect on the clinical course of EAE depended on CD8+ T cells that neither participate in viral clearance nor induce neuropathology in infected mice without EAE. Our system should be useful to further unravel how certain viral infections trigger MS exacerbations and to understand how CD8+ T cells can exert pathogenic effects within active demyelinating lesions.


Assuntos
Infecções por Alphavirus/complicações , Infecções por Alphavirus/imunologia , Linfócitos T CD8-Positivos/virologia , Encefalomielite Autoimune Experimental/virologia , Esclerose Múltipla/virologia , Vírus Sindbis/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Doenças Desmielinizantes/imunologia , Doenças Desmielinizantes/virologia , Células Dendríticas/imunologia , Células Dendríticas/virologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/imunologia , Feminino , Citometria de Fluxo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Esclerose Múltipla/imunologia , Bainha de Mielina/imunologia , Células Th1/imunologia , Células Th1/virologia , Células Th17/imunologia , Células Th17/virologia
14.
Prog Neurobiol ; 101-102: 46-64, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23201558

RESUMO

Multiple sclerosis (MS) is a complex inflammatory disease of unknown etiology that affects the central nervous system (CNS) white matter, and for which no effective cure exists. Indeed, whether the primary event in MS pathology affects myelin or axons of the CNS remains unclear. Animal models are necessary to identify the immunopathological mechanisms involved in MS and to develop novel therapeutic and reparative approaches. Specifically, viral models of chronic demyelination and axonal damage have been used to study the contribution of viruses in human MS, and they have led to important breakthroughs in our understanding of MS pathology. The Theiler's murine encephalomyelitis virus (TMEV) model is one of the most commonly used MS models, although other viral models are also used, including neurotropic strains of mouse hepatitis virus (MHV) that induce chronic inflammatory demyelination with similar histological features to those observed in MS. This review will discuss the immunopathological mechanisms involved in TMEV-induced demyelinating disease (TMEV-IDD). The TMEV model reproduces a chronic progressive disease due to the persistence of the virus for the entire lifespan in susceptible mice. The evolution and significance of the axonal damage and neuroinflammation, the importance of epitope spread from viral to myelin epitopes, the presence of abortive remyelination and the existence of a brain pathology in addition to the classical spinal cord demyelination, are some of the findings that will be discussed in the context of this TMEV-IDD model. Despite their limitations, viral models remain an important tool to study the etiology of MS, and to understand the clinical and pathological variability associated with this disease.


Assuntos
Doenças Desmielinizantes/etiologia , Encefalomielite Autoimune Experimental , Esclerose Múltipla , Degeneração Neural/etiologia , Theilovirus/patogenicidade , Animais , Doenças Desmielinizantes/virologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/virologia , Humanos , Camundongos , Esclerose Múltipla/complicações , Esclerose Múltipla/patologia , Esclerose Múltipla/virologia , Degeneração Neural/virologia
15.
Methods Mol Biol ; 900: 381-401, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22933080

RESUMO

Experimental autoimmune encephalomyelitis (EAE) and Theiler's Murine Encephalitis Virus-Induced Demyelinating Disease (TMEV-IDD) are two clinically relevant murine models of multiple sclerosis (MS). Like MS, both are characterized by mononuclear cell infiltration into the CNS and demyelination. EAE is induced by either the administration of myelin protein or peptide in adjuvant or by the adoptive transfer of encephalitogenic T cell blasts into naïve recipients. The relative merits of each of these protocols are compared. Depending on the type of question being asked, different mouse strains and peptides are used. Different disease courses are observed with different strains and different peptides in active EAE. These variations are also addressed. Additionally, issues relevant to clinical grading of EAE in mice are discussed. In addition to EAE induction, useful references for other disease indicators such as DTH, in vitro proliferation, and immunohistochemistry are provided. TMEV-IDD is a useful model for understanding the possible viral etiology of MS. This section provides detailed information on the preparation of viral stocks and subsequent intracerebral infection of mice. Additionally, virus plaque assay and clinical disease assessment are discussed. Recently, recombinant TMEV strains have been created for the study of molecular mimicry which incorporate various 30 amino acid myelin epitopes within the leader region of TMEV.


Assuntos
Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/virologia , Técnicas Imunológicas/métodos , Esclerose Múltipla/patologia , Theilovirus/fisiologia , Transferência Adotiva , Sequência de Aminoácidos , Animais , Progressão da Doença , Encefalomielite Autoimune Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Esclerose Múltipla/imunologia , Peptídeos/química , Vacinação
16.
J Neuroinflammation ; 9: 104, 2012 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-22642802

RESUMO

BACKGROUND: The interplay between IFN-γ, IL-17 and neutrophils during CNS inflammatory disease is complex due to cross-regulatory factors affecting both positive and negative feedback loops. These interactions have hindered the ability to distinguish the relative contributions of neutrophils, Th1 and Th17 cell-derived effector molecules from secondary mediators to tissue damage and morbidity. METHODS: Encephalitis induced by a gliatropic murine coronavirus was used as a model to assess the direct contributions of neutrophils, IFN-γ and IL-17 to virus-induced mortality. CNS inflammatory conditions were selectively manipulated by adoptive transfer of virus-primed wild-type (WT) or IFN-γ deficient (GKO) memory CD4+ T cells into infected SCID mice, coupled with antibody-mediated neutrophil depletion and cytokine blockade. RESULTS: Transfer of GKO memory CD4+ T cells into infected SCID mice induced rapid mortality compared to recipients of WT memory CD4+ T cells, despite similar virus control and demyelination. In contrast to recipients of WT CD4+ T cells, extensive neutrophil infiltration and IL-17 expression within the CNS in recipients of GKO CD4+ T cells provided a model to directly assess their contribution(s) to disease. Recipients of WT CD4+ T cells depleted of IFN-γ did not express IL-17 and were spared from mortality despite abundant CNS neutrophil infiltration, indicating that mortality was not mediated by excessive CNS neutrophil accumulation. By contrast, IL-17 depletion rescued recipients of GKO CD4+ T cells from rapid mortality without diminishing neutrophils or reducing GM-CSF, associated with pathogenic Th17 cells in CNS autoimmune models. Furthermore, co-transfer of WT and GKO CD4+ T cells prolonged survival in an IFN-γ dependent manner, although IL-17 transcription was not reduced. CONCLUSIONS: These data demonstrate that IL-17 mediates detrimental clinical consequences in an IFN-γ-deprived environment, independent of extensive neutrophil accumulation or GM-CSF upregulation. The results also suggest that IFN-γ overrides the detrimental IL-17 effector responses via a mechanism downstream of transcriptional regulation.


Assuntos
Encefalomielite Autoimune Experimental/prevenção & controle , Interferon gama/fisiologia , Interleucina-17/fisiologia , Vírus da Hepatite Murina/imunologia , Neutrófilos/imunologia , Animais , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/virologia , Hepatite Viral Animal/imunologia , Hepatite Viral Animal/patologia , Hepatite Viral Animal/prevenção & controle , Interferon gama/deficiência , Interleucina-17/deficiência , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos SCID , Neutrófilos/patologia , Neutrófilos/virologia , Transcrição Gênica/imunologia
17.
PLoS Pathog ; 8(5): e1002715, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22615572

RESUMO

Epstein-Barr virus (EBV) has been identified as a putative environmental trigger of multiple sclerosis (MS), yet EBV's role in MS remains elusive. We utilized murine gamma herpesvirus 68 (γHV-68), the murine homolog to EBV, to examine how infection by a virus like EBV could enhance CNS autoimmunity. Mice latently infected with γHV-68 developed more severe EAE including heightened paralysis and mortality. Similar to MS, γHV-68EAE mice developed lesions composed of CD4 and CD8 T cells, macrophages and loss of myelin in the brain and spinal cord. Further, T cells from the CNS of γHV-68 EAE mice were primarily Th1, producing heightened levels of IFN-γ and T-bet accompanied by IL-17 suppression, whereas a Th17 response was observed in uninfected EAE mice. Clearly, γHV-68 latency polarizes the adaptive immune response, directs a heightened CNS pathology following EAE induction reminiscent of human MS and portrays a novel mechanism by which EBV likely influences MS and other autoimmune diseases.


Assuntos
Encéfalo/imunologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/virologia , Rhadinovirus/patogenicidade , Medula Espinal/imunologia , Latência Viral , Animais , Encéfalo/patologia , Encéfalo/virologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Células Cultivadas , Encefalomielite Autoimune Experimental/patologia , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/patologia , Herpesvirus Humano 4/patogenicidade , Interferon gama/biossíntese , Interleucina-17/biossíntese , Macrófagos/imunologia , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/imunologia , Esclerose Múltipla/virologia , Medula Espinal/patologia , Medula Espinal/virologia , Proteínas com Domínio T/biossíntese , Células Th1/imunologia , Células Th17/imunologia
18.
J Neuroinflammation ; 9: 58, 2012 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-22436404

RESUMO

BACKGROUND: Prions, composed of a misfolded protein designated PrP(Sc), are infectious agents causing fatal neurodegenerative diseases. We have shown previously that, following induction of experimental autoimmune encephalomyelitis, prion-infected mice succumb to disease significantly earlier than controls, concomitant with the deposition of PrP(Sc) aggregates in inflamed white matter areas. In the present work, we asked whether prion disease acceleration by experimental autoimmune encephalomyelitis results from infiltration of viable prion-infected immune cells into the central nervous system. METHODS: C57Bl/6 J mice underwent intraperitoneal inoculation with scrapie brain homogenates and were later induced with experimental autoimmune encephalomyelitis by inoculation of MOG(35-55) in complete Freund's adjuvant supplemented with pertussis toxin. Spleen and lymph node cells from the co-induced animals were reactivated and subsequently injected into naïve mice as viable cells or as cell homogenates. Control groups were infected with viable and homogenized scrapie immune cells only with complete Freund's adjuvant. Prion disease incubation times as well as levels and sites of PrP(Sc) deposition were next evaluated. RESULTS: We first show that acceleration of prion disease by experimental autoimmune encephalomyelitis requires the presence of high levels of spleen PrP(Sc). Next, we present evidence that mice infected with activated prion-experimental autoimmune encephalomyelitis viable cells succumb to prion disease considerably faster than do mice infected with equivalent cell extracts or other controls, concomitant with the deposition of PrP(Sc) aggregates in white matter areas in brains and spinal cords. CONCLUSIONS: Our results indicate that inflammatory targeting of viable prion-infected immune cells to the central nervous system accelerates prion disease propagation. We also show that in the absence of such targeting it is the load of PrP(Sc) in the inoculum that determines the infectivity titers for subsequent transmissions. Both of these conclusions have important clinical implications as related to the risk of prion disease contamination of blood products.


Assuntos
Sistema Nervoso Central , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Linfócitos/patologia , Doenças Priônicas/patologia , Príons/metabolismo , Animais , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/virologia , Glicoproteínas/efeitos adversos , Humanos , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Peptídeos/efeitos adversos , Doenças Priônicas/complicações , Príons/patogenicidade
19.
J Neurovirol ; 18(1): 45-54, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22281874

RESUMO

Major histocompatibility complex class I-restricted CD8(+) cytotoxic T lymphocytes are involved in the pathogenesis of multiple sclerosis (MS) and both autoimmune, experimental autoimmune encephalomyelitis, and viral, Theiler's murine encephalomyelitis virus (TMEV) infection, animal models of MS. Following TMEV infection, certain T cell hybridomas, generated from cloned TMEV-induced CD8(+) T cells, were able to produce clinical signs of disease (flaccid hind limb paralysis) upon adoptive transfer into naive mice. Dual T cell receptors (TCR) are present on the surface of these cells as both Vß3 and Vß6 were detected by polymerase chain reaction (PCR) screening and flow cytometry and multiple Vα mRNAs were detected by PCR screening. This is the first demonstration of antiviral CD8(+) T cells having more than one TCR initiating an autoimmune disease in the natural host of the virus. We hypothesize that this is a potential mechanism for virus-induced autoimmune disease initiated by CD8(+) T cells.


Assuntos
Infecções por Cardiovirus/imunologia , Encefalomielite Autoimune Experimental/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/biossíntese , Linfócitos T Citotóxicos/imunologia , Theilovirus , Transferência Adotiva , Animais , Infecções por Cardiovirus/patologia , Infecções por Cardiovirus/virologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/virologia , Feminino , Citometria de Fluxo , Hibridomas/imunologia , Hibridomas/transplante , Hibridomas/virologia , Injeções Intravenosas , Camundongos , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Linfócitos T Citotóxicos/transplante , Linfócitos T Citotóxicos/virologia
20.
Biochim Biophys Acta ; 1812(2): 132-40, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20600868

RESUMO

Genetic and environmental factors jointly determine the susceptibility to develop Multiple Sclerosis (MS). Collaborative efforts during the past years achieved substantial progress in defining the genetic architecture, underlying susceptibility to MS. Similar to other autoimmune diseases, HLA-DR and HLA-DQ alleles within the HLA class II region on chromosome 6p21 are the highest-risk-conferring genes. Less-robust susceptibility effects have been identified for MHC class I alleles and for non-MHC regions. The role of environmental risk factors and their interaction with genetic susceptibility alleles are much less well defined, despite the fact that infections have long been associated with MS development. Current data suggest that infectious triggers are most likely ubiquitous, i.e., highly prevalent in the general population, and that they require a permissive genetic trait which predisposes for MS development. In this review article, we illustrate mechanisms of infection-induced immunopathologies in experimental animal models of autoimmune CNS inflammation, discuss challenges for the translation of these experimental data into human immunology research, and provide future perspectives on how novel model systems could be utilized to better define the role of viral pathogens in MS.


Assuntos
Esclerose Múltipla/virologia , Animais , Encefalomielite Autoimune Experimental/etiologia , Encefalomielite Autoimune Experimental/virologia , Retrovirus Endógenos/patogenicidade , Predisposição Genética para Doença , Antígenos HLA-DQ/genética , Antígenos HLA-DR/genética , Herpesviridae/patogenicidade , Herpesvirus Humano 3/patogenicidade , Herpesvirus Humano 4/patogenicidade , Herpesvirus Humano 6/patogenicidade , Humanos , Modelos Biológicos , Mimetismo Molecular/imunologia , Esclerose Múltipla/etiologia , Esclerose Múltipla/genética , Esclerose Múltipla/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Torque teno virus/patogenicidade , Viroses/complicações
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...