Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 199(2): 435-448, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28592428

RESUMO

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.


Assuntos
Autoimunidade , Autofagia/imunologia , Linfócitos B/imunologia , Linfócitos B/virologia , Esclerose Múltipla/imunologia , Animais , Autofagossomos/imunologia , Autofagossomos/metabolismo , Linfócitos B/metabolismo , Linfócitos T CD8-Positivos/imunologia , Catepsina G/antagonistas & inibidores , Catepsina G/genética , Catepsina G/imunologia , Catepsina G/metabolismo , Células Cultivadas , Apresentação Cruzada/imunologia , Humanos , Epitopos Imunodominantes/química , Epitopos Imunodominantes/imunologia , Camundongos , Esclerose Múltipla/fisiopatologia , Esclerose Múltipla/virologia , Glicoproteína Mielina-Oligodendrócito/imunologia , Glicoproteína Mielina-Oligodendrócito/metabolismo
2.
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
3.
Drug Discov Today ; 21(8): 1200-5, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27060373

RESUMO

New drugs often fail in the translation from the rodent experimental autoimmune encephalomyelitis (EAE) model to human multiple sclerosis (MS). Here, we present the marmoset EAE model as an indispensable model for translational research into MS. The genetic heterogeneity of this species and lifelong exposure to chronic latent infections and environmental pathogens create a human-like immune system. Unique to this model is the presence of the pathological hallmark of progressive MS, in particular cortical grey matter lesions. Another great possibility of this model is systemic and longitudinal immune profiling, whereas in humans and mice immune profiling is usually performed in a single compartment (i.e. blood or spleen, respectively). Overall, the marmoset model provides unique opportunities for systemic drug-effect profiling.


Assuntos
Callithrix , Encefalomielite Autoimune Experimental/terapia , Imunoterapia , Esclerose Múltipla/terapia , Animais , Linfócitos B/imunologia , Linfócitos B/virologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Substância Cinzenta/patologia , Herpesvirus Humano 4/imunologia , Humanos , Linfonodos/imunologia , Camundongos , Baço/imunologia
4.
Methods Mol Biol ; 1304: 171-86, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-25208751

RESUMO

Experimental autoimmune encephalomyelitis (EAE) in the common marmoset, a small-bodied Neotropical primate, is a well-known and validated animal model for multiple sclerosis (MS). This model can be used for exploratory research, i.e., investigating the pathogenic mechanisms involved in MS, and applied research, testing the efficacy of new potential drugs.In this chapter, we will describe a method to induce EAE in the marmoset. In addition, we will explain the most common immunological techniques involved in the marmoset EAE research, namely isolation of mononuclear cells (MNC) from peripheral blood and lymphoid tissue, assaying T cell proliferation by thymidine incorporation, MNC phenotyping by flow cytometry, antibody measurement by ELISA, generation of B cell lines and antigen-specific T cell lines, and assaying cytotoxic T cells.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Animais , Callithrix , Modelos Animais de Doenças , Imunofenotipagem , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Linfócitos/imunologia , Linfócitos/metabolismo
5.
J Neuroinflammation ; 12: 169, 2015 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-26377397

RESUMO

BACKGROUND: Experimental autoimmune encephalomyelitis (EAE) in the common marmoset monkey (Callithrix jacchus) is a relevant preclinical model for translational research into immunopathogenic mechanisms operating in multiple sclerosis (MS). Prior studies showed a core pathogenic role of T and B cells specific for myelin oligodendrocyte glycoprotein (MOG). However, in those studies, the quality of the response against MOG epitopes was strongly biased by bacterial antigens in the complete Freund's adjuvant (CFA), in which the immunizing recombinant human (rh) MOG protein had been formulated. In response to the need of a more refined EAE model, we have tested whether disease could also be induced with rhMOG in incomplete Freund's adjuvant (IFA). METHOD: Marmosets were immunized with rhMOG emulsified in IFA in the dorsal skin. Monkeys that did not develop neurological deficit were given booster immunizations at 28-day interval with the same antigen preparation. In a second experiment, three marmoset twin pairs were sensitized against MOG peptides in IFA to study a possibility for suppressive activity towards pathogenic T cells directed against the encephalitogenic epitope MOG40-48. RESULTS: Despite the absence of strong danger signals in the rhMOG/IFA inoculum, all monkeys developed clinically evident EAE symptoms. Moreover, in all monkeys, demyelinated lesions were present in the white matter and in two cases also in the cortical grey matter. Immune profiling at height of the disease showed a dominant T cell response against the overlapping peptides 14-36 and 24-46, but reactivity against the pathogenically most relevant peptide 34-56 was conspicuously absent. In the second experiment, there was an indication for a possible suppressive mechanism. CONCLUSIONS: Immunization of marmoset monkeys with rhMOG in IFA elicits clinical EAE in all animals. Moreover, rhMOG contains pathogenic and regulatory epitopes, but the pathogenic hierarchy of rhMOG epitopes is strongly influenced by the adjuvant in which the protein is formulated.


Assuntos
Callithrix , Encefalomielite Autoimune Experimental/imunologia , Adjuvante de Freund/imunologia , Lipídeos/imunologia , Glicoproteína Mielina-Oligodendrócito/imunologia , Animais , Anticorpos/sangue , Encéfalo/metabolismo , Encéfalo/patologia , Proliferação de Células/efeitos dos fármacos , Citocinas/sangue , Citocinas/genética , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/etiologia , Encefalomielite Autoimune Experimental/patologia , Feminino , Adjuvante de Freund/efeitos adversos , Humanos , Imunização/efeitos adversos , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/patologia , Lipídeos/efeitos adversos , Ativação Linfocitária/imunologia , Bainha de Mielina/metabolismo , Bainha de Mielina/patologia , Glicoproteína Mielina-Oligodendrócito/efeitos adversos , Peptídeos/efeitos adversos , Peptídeos/imunologia , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/imunologia , Medula Espinal/metabolismo , Medula Espinal/patologia , Linfócitos T/imunologia
6.
Drug Discov Today ; 19(9): 1394-401, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24704460

RESUMO

The poor translational validity of autoimmune-mediated inflammatory disease (AIMID) models in inbred and specific pathogen-free (SPF) rodents underlies the high attrition of new treatments for the corresponding human disease. Experimental autoimmune encephalomyelitis (EAE) is a frequently used preclinical AIMID model. We discuss here how crucial information needed for the innovation of current preclinical models can be obtained from postclinical analysis of the nonhuman primate EAE model, highlighting the mechanistic reasons why some therapies fail and others succeed. These new insights can also help identify new targets for treatment.


Assuntos
Doenças Autoimunes/fisiopatologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/fisiopatologia , Animais , Desenho de Fármacos , Humanos , Primatas , Roedores , Especificidade da Espécie , Pesquisa Translacional Biomédica/métodos
7.
Exp Anim ; 62(3): 159-71, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23903050

RESUMO

The common marmoset (Callithrix jacchus) is a small-bodied Neotropical primate and a useful preclinical animal model for translational research into autoimmune-mediated inflammatory diseases (AIMID), such as rheumatoid arthritis (RA) and multiple sclerosis (MS). The animal model for MS established in marmosets has proven their value for exploratory research into (etio) pathogenic mechanisms and for the evaluation of new therapies that cannot be tested in lower species because of their specificity for humans. Effective usage of the marmoset in preclinical immunological research has been hampered by the limited availability of blood for immunological studies and of reagents for profiling of cellular and humoral immune reactions. In this paper, we give a concise overview of the procedures and reagents that were developed over the years in our laboratory in marmoset models of the above-mentioned diseases.


Assuntos
Animais de Laboratório , Artrite Reumatoide , Autoimunidade , Callithrix , Modelos Animais de Doenças , Encefalomielite , Esclerose Múltipla , Pesquisa Translacional Biomédica/métodos , Animais , Artrite Reumatoide/imunologia , Reagentes de Ligações Cruzadas , Encefalomielite/imunologia , Feminino , Humanos , Masculino , Esclerose Múltipla/imunologia
8.
J Neuroimmune Pharmacol ; 8(5): 1251-64, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23821341

RESUMO

The experimental autoimmune encephalitis (EAE) model is used for preclinical research into the pathogenesis of multiple sclerosis (MS), mostly in inbred, specific pathogen free (SPF)-raised laboratory mice. However, the naive state of the laboratory mouse immune system is considered a major hurdle in the translation of principles from the EAE model to the MS patient. Non-human primates (NHP) have an immune system harboring T- and B-cell memory against environmental antigens, similar as in humans. We sought to further refine existing NHP EAE models, which may help to bridge the gab between mouse EAE models and MS. We report here on new EAE models in three NHP species: rhesus monkeys (Macaca mulatta), cynomolgus monkeys (Macaca fascicularis) and common marmosets (Callithrix jacchus). EAE was induced with recombinant human myelin oligodendrocyte glycoprotein extracellular domain (1-125) (rhMOG) formulated in incomplete Freund's adjuvant (IFA). IFA lacks the bacterial antigens that are present in complete Freund's adjuvant (CFA), which are notorious for the induction of discomforting side effects. Clinically evident EAE could be induced in two out of five rhesus monkeys, six out of six cynomolgus monkeys and six out of six common marmosets. In each of these species, the presence of an early, high anti-rhMOG IgM response is correlated with EAE with an earlier onset and more severe disease course. Animals without an early high IgM response either did not develop disease (rhesus monkeys) or developed only mild signs of neurological deficit (marmoset and cynomolgus monkeys).


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Adjuvante de Freund/imunologia , Glicoproteína Mielina-Oligodendrócito/imunologia , Adjuvantes Imunológicos/farmacologia , Animais , Encéfalo/patologia , Callithrix , Encefalomielite Autoimune Experimental/patologia , Adjuvante de Freund/farmacologia , Humanos , Imunoglobulina M/imunologia , Imuno-Histoquímica , Macaca fascicularis , Macaca mulatta , Proteínas Recombinantes/imunologia , Medula Espinal/patologia
9.
Front Immunol ; 4: 145, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23781220

RESUMO

The remarkable clinical efficacy of anti-CD20 monoclonal antibodies (mAb) in relapsing-remitting multiple sclerosis points at the critical involvement of B cells in the disease. However, the exact pathogenic contribution of B cells is poorly understood. In this publication we review new data on the role of CD20+ B cells in a unique experimental autoimmune encephalomyelitis (EAE) model in common marmosets (Callithrix jacchus), a small-bodied neotropical primate. We will also discuss the relevance of these data for MS. Different from rodent EAE models, but similar to MS, disease progression in marmosets can develop independent of autoantibodies. Progressive disease is mediated by MHC class Ib (Caja-E) restricted cytotoxic T cells, which are activated by γ-herpesvirus-infected B cells and cause widespread demyelination of cortical gray matter. B-cell directed monoclonal antibody therapies (anti-CD20 versus anti-BLyS and anti-APRIL) have a variable effect on EAE progression, which we found associated with variable depletion of the Epstein Barr virus (EBV)-like γ-herpesvirus CalHV3 from lymphoid organs. These findings support an important pathogenic role of CD20+ B cell in MS, especially of the subset infected with EBV.

10.
J Neuroimmune Pharmacol ; 7(3): 557-70, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22870852

RESUMO

B lymphocyte stimulator (BLyS, also indicated as BAFF (B-cell activating factor) and CD257), and A Proliferation Inducing Ligand (APRIL, CD256) are two members of the TNF superfamily with a central role in B cell survival. Antibodies against these factors have potential therapeutic relevance in autoimmune inflammatory disorders with a proven pathogenic contribution of B cells, such as multiple sclerosis (MS). In the current study we performed a multi-parameter efficacy comparison of monoclonal antibodies against human anti-BLyS and anti-APRIL in a common marmoset (Callithrix jacchus) model of experimental autoimmune encephalomyelitis (EAE). A MS-like disease was induced by immunization with recombinant human myelin/oligodendrocyte glycoprotein (rhMOG) in complete Freund's adjuvant. The results show that the anti-BLyS and anti-APRIL antibody cause significant depletion of circulating CD20+ B cells, but a small subset of CD20 + CD40(high) B cells was not depleted. Induction of CD20+ B cell depletion from lymph nodes was only observed in the anti-BLyS treated monkeys. Both antibodies had a significant inhibitory effect on disease development, but all monkeys developed clinically evident EAE. Anti-BLyS treated monkeys were sacrificed with the same clinical signs as saline-treated monkeys, but nevertheless displayed significantly reduced spinal cord demyelination. This effect was not observed in the anti-APRIL treated monkeys. The two antibodies had a different effect on T cell subset activation and the profiles of ex vivo released cytokines. In conclusion, treatment with anti-BLyS and anti-APRIL delays the development of neurological disease in a relevant preclinical model of MS. The two mAbs achieve this effect via different mechanisms.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Fator Ativador de Células B/imunologia , Encefalomielite Autoimune Experimental/prevenção & controle , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Animais , Anticorpos Monoclonais/fisiologia , Fator Ativador de Células B/antagonistas & inibidores , Callithrix , Células Cultivadas , Encefalomielite Autoimune Experimental/imunologia , Humanos , Masculino , Distribuição Aleatória , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/antagonistas & inibidores
11.
J Neuropathol Exp Neurol ; 71(8): 716-28, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22805775

RESUMO

CD20-positive B-cell depletion is a highly promising treatment for multiple sclerosis (MS), but the mechanisms underlying therapeutic effects are poorly understood. B cells are thought to contribute to MS pathogenesis by producing autoantibodies that amplify demyelination via opsonization of myelin. To analyze autoantibody-nondependent functions of B cells in an animal model of MS, we used a novel T cell-driven experimental autoimmune encephalomyelitis (EAE) model in marmoset monkeys (Callithrix jacchus). In this model, demyelination of brain and spinal cord white and gray matter and the ensuing neurological deficits are induced by immunization with peptide 34 to 56 of myelin/oligodendrocyte glycoprotein (MOG34-56) in incomplete Freund's adjuvant. Although autoantibodies do not have a detectable pathogeniccontribution in the model, depletion of B cells with monoclonal antibody 7D8, a human IgG1κ monoclonal antibody against human CD20, suppressed clinical and pathological EAE. In B cell-depleted monkeys, the activation of peptide-specific Th17-producing and cytotoxic T cells, which in previous studies were found to play an essential role in disease induction, was impaired. Thus, we demonstrate a critical antibody-nondependent role for B cells in EAE, that is, the activation of pathogenic T cells.


Assuntos
Linfócitos B/fisiologia , Callithrix/fisiologia , Doenças Desmielinizantes/patologia , Encefalomielite Autoimune Experimental/patologia , Depleção Linfocítica/efeitos adversos , Linfócitos T/fisiologia , Animais , Anticorpos/análise , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Antígenos CD20/imunologia , Encéfalo/patologia , Linfócitos T CD4-Positivos/fisiologia , Antígeno CD56/imunologia , Linfócitos T CD8-Positivos/fisiologia , Proliferação de Células , Citocinas/biossíntese , Encefalomielite Autoimune Experimental/tratamento farmacológico , Feminino , Imunização , Proteínas da Mielina/imunologia , Glicoproteína Mielina-Oligodendrócito , Fenótipo , Complexo Receptor-CD3 de Antígeno de Linfócitos T/imunologia , Proteínas Recombinantes/imunologia
12.
J Neuroimmune Pharmacol ; 7(1): 253-65, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22012268

RESUMO

The core pathogenic process in the common marmoset model of multiple sclerosis (MS) is the activation of memory-like T cells specific for peptide 34 to 56 derived from the extracellular domain of myelin/oligodendrocyte glycoprotein (MOG(34-56)). Immunization with MOG(34-56) in incomplete Freund's adjuvant is a sufficient stimulus for in vivo activation of these T cells, together with the induction of MS-like disease and CNS pathology. Ex vivo functional characteristics of MOG(34-56) specific T cells are specific cytolysis of peptide pulsed target cells and high IL-17A production. To indentify possible functions in this new model of T helper 1 cells, which play a central pathogenic role in MS models induced with complete Freund's adjuvant, we tested the effect of human interferon-γ (IFNγ) administration during disease initiation of the disease (day 0-25) and around the time of disease expression (psd 56-81). The results show a clear modulatory effect of early IFNγ treatment on humoral and cellular autoimmune parameters, but no generalized mitigating effect on the disease course. These results argue against a prominent pathogenic role of T helper 1 cells in this new marmoset EAE model.


Assuntos
Encéfalo/imunologia , Encefalomielite Autoimune Experimental/imunologia , Interferon gama/imunologia , Células Th1/imunologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Callithrix , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/patologia , Feminino , Citometria de Fluxo , Humanos , Interferon gama/farmacologia , Masculino , Esclerose Múltipla/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Th1/efeitos dos fármacos
13.
Eur J Immunol ; 42(1): 217-27, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21928277

RESUMO

Induction of experimental autoimmune encephalomyelitis (EAE) has been documented in common marmosets using peptide 34-56 from human myelin/oligodendrocyte glycoprotein (MOG(34-56) ) in incomplete Freund's adjuvant (IFA). Here, we report that this EAE model is associated with widespread demyelination of grey and white matter. We performed an in-depth analysis of the specificity, MHC restriction and functions of the activated T cells in the model, which likely cause EAE in an autoantibody-independent manner. T-cell lines isolated from blood and lymphoid organs of animals immunized with MOG(34-56) displayed high production of IL-17A and specific lysis of MOG(34-56) -pulsed EBV B-lymphoblastoid cells as typical hallmarks. Cytotoxicity was directed at the epitope MOG(40-48) presented by the non-classical MHC class Ib allele Caja-E, which is orthologue to HLA-E and is expressed in non-inflamed brain. In vivo activated T cells identified by flow cytometry in cultures with MOG(34-56,) comprised CD4(+) CD56(+) and CD4(+) CD8(+) CD56(+) T cells. Furthermore, phenotypical analysis showed that CD4(+) CD8(+) CD56(+) T cells also expressed CD27, but CD16, CD45RO, CD28 and CCR7 were absent. These results show that, in the MOG34-56/IFA marmoset EAE model, a Caja-E-restricted population of autoreactive cytotoxic T cells plays a key role in the process of demyelination in the grey and white matter.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Esclerose Múltipla/imunologia , Bainha de Mielina/imunologia , Linfócitos T/imunologia , Adjuvantes Imunológicos , Animais , Callithrix , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Interleucina-17/imunologia , Células K562 , Proteínas da Mielina , Glicoproteína Mielina-Oligodendrócito , RNA/química , RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
J Neuroimmune Pharmacol ; 6(3): 341-53, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20700661

RESUMO

We report on the effect of antibody-mediated neutralization of interleukin (IL)-17A in a non-human primate experimental autoimmune encephalomyelitis (EAE) model induced with recombinant human myelin oligodendrocyte glycoprotein (rhMOG). We tested a human-anti-human IL-17A-antibody in two doses (3 and 30 mg/kg) against placebo (PBS). The treatment was started 1 day before EAE induction and continued throughout the experiment. Although all monkeys developed clinically evident EAE, the onset of neurological signs was delayed in some monkeys from both treatment groups. Total CNS lesion volumes, demyelination, or inflammation did not differ between the different groups. Immune profiling revealed an altered distribution of IL-17A producing cells in the lymphoid organs of antibody-treated monkeys. Comparable numbers of IL-17A producing cells were observed in the brain. RhMOG-induced T cell proliferation in the lymph nodes was slightly reduced after anti-IL-17A antibody treatment. To summarize, we found that anti-IL-17A antibody as a single treatment from disease induction effects a trend towards delayed neurological disease progression in the marmoset EAE model, although the effect did not reach statistical significance. This suggests a role of IL-17A in late stage disease in the marmoset EAE model, but IL-17A may not be the dominant pathogenic cytokine.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Interleucina-17/antagonistas & inibidores , Animais , Anticorpos Monoclonais/farmacologia , Encéfalo/imunologia , Encéfalo/patologia , Callithrix , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Imuno-Histoquímica , Interleucina-17/imunologia , Masculino , Proteínas da Mielina , Glicoproteína Associada a Mielina/imunologia , Glicoproteína Mielina-Oligodendrócito , Linfócitos T/imunologia
15.
J Neuropathol Exp Neurol ; 69(4): 372-85, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20448482

RESUMO

Experimental autoimmune encephalomyelitis in the neotropical primate common marmoset (Callithrix jacchus) is a relevant autoimmune animal model of multiple sclerosis. T cells specific for peptide 34 to 56 of myelin/oligodendrocyte glycoprotein (MOG34-56) have a central pathogenic role in this model. The aim of this study was to assess the requirement for innate immune stimulation for activation of this core pathogenic autoimmune mechanism. Marmoset monkeys were sensitized against synthetic MOG34-56 peptide alone or in combination with the nonencephalitogenic peptide MOG74-96 formulated in incomplete Freund adjuvant, which lacks microbial components. Experimental autoimmune encephalomyelitis development was recorded by monitoring neurological signs, brain magnetic resonance imaging, and longitudinal profiling of cellular and humoral immune parameters. All monkeys developed autoimmune inflammatory/demyelinating central nervous system disease characterized by massive brain and spinal cord demyelinating white matter lesions with activated macrophages and CD3+ T cells. Immune profiling ex vivo demonstrated the activation of mainly CD3+CD4+/8+CD56+ T cells against MOG34-56. Upon ex vivo stimulation, these T cells produced more interleukin 17A compared with TH1 cytokines (e.g. interferon-gamma) and displayed peptide-specific cytolytic activity. These results indicate that the full spectrum of marmoset experimental autoimmune encephalomyelitis can be induced by sensitization against a single MOG peptide in incomplete Freund adjuvant lacking microbial compounds for innate immune activation and by eliciting antigen-specific T-cell cytolytic activity.


Assuntos
Doenças Autoimunes Desmielinizantes do Sistema Nervoso Central/induzido quimicamente , Modelos Animais de Doenças , Adjuvante de Freund/química , Glicoproteínas/farmacologia , Fragmentos de Peptídeos/farmacologia , Animais , Antígenos CD/metabolismo , Autoanticorpos/sangue , Autoanticorpos/imunologia , Encéfalo/imunologia , Encéfalo/patologia , Calgranulina B/metabolismo , Callithrix , Linhagem Celular Transformada , Citocinas/metabolismo , Doenças Autoimunes Desmielinizantes do Sistema Nervoso Central/imunologia , Doenças Autoimunes Desmielinizantes do Sistema Nervoso Central/patologia , Doenças Autoimunes Desmielinizantes do Sistema Nervoso Central/fisiopatologia , Citometria de Fluxo/métodos , Humanos , Imunidade Inata , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Imageamento por Ressonância Magnética/métodos , Proteína Proteolipídica de Mielina/metabolismo , Glicoproteína Mielina-Oligodendrócito , Medula Espinal/patologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo
16.
J Neuropathol Exp Neurol ; 67(4): 326-40, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18379435

RESUMO

To elucidate the pathogenetic significance of myelin/oligodendrocyte glycoprotein (MOG)-specific autoreactivity in a genetically and immunologically heterogeneous nonhuman primate model of multiple sclerosis, we analyzed experimental autoimmune encephalomyelitis (EAE) in the outbred common marmoset (Callithrix jacchus). One sibling each of 5 bone marrow chimeric marmoset twins was immunized with myelin derived from wild-type (WT) C57BL/6 mice (WT myelin); the other sibling was immunized with myelin from MOG-deficient C57BL/6 mice (MOG -/- myelin). One twin pair developed acute EAE simultaneously; the 4 remaining twin siblings immunized with WT myelin developed chronic progressive EAE, whereas siblings of these 4 monkeys remained free of clinical disease signs. Many EAE-related abnormalities were identified in the CNS of both groups by magnetic resonance imaging and histologic analysis, but mean percentages of spinal cord demyelination were lower in monkeys immunized with MOG -/- myelin (8.2%) than in WT myelin-immunized animals (40.5%). There was a strong correlation between the development of overt clinical EAE and seropositivity for anti-MOG antibodies, but blood and lymph node T-cell proliferative responses showed no relationship to disease. These results indicate that the initiation of CNS inflammation and demyelination can take place in the absence of detectable autoimmunity against MOG, but the clinical progression and histopathologic severity depends on the presence of antibodies against MOG in this multiple sclerosis model.


Assuntos
Autoimunidade , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/imunologia , Glicoproteína Associada a Mielina/imunologia , Animais , Peso Corporal , Callithrix , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Citocinas/metabolismo , Progressão da Doença , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/fisiopatologia , Ensaio de Imunoadsorção Enzimática/métodos , Haplorrinos , Ativação Linfocitária , Imageamento por Ressonância Magnética/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas da Mielina , Bainha de Mielina/imunologia , Glicoproteína Associada a Mielina/deficiência , Glicoproteína Associada a Mielina/metabolismo , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Fatores de Tempo
17.
Eur J Immunol ; 38(5): 1452-64, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18412169

RESUMO

Oligodendrocyte-specific protein (OSP) is a candidate autoantigen in the development of multiple sclerosis (MS). We evaluated the potential of OSP to induce EAE in rhesus monkeys, an out bred animal model for MS that is immunologically close to humans. Since OSP is a four-membrane spanning protein with highly hydrophobic regions, we synthesized recombinant proteins encompassing only the hydrophilic regions of human OSP (soluble (s)hOSP). Immunization with shOSP proteins induced clinical signs and histological features of optic neuritis in four out of ten rhesus monkeys. The development of clinical disease was associated with the presence of a strong cellular proliferative response to the immunizing shOSP protein. Analysis of the cellular responses in combination with neuropathological observations also indicates an important role for neutrophils in the disease process. Interestingly, all immunized monkeys developed antibody responses to OSP peptide 103-123, a B cell epitope previously identified in MS patients. These responses did not correlate with the development of clinical disease, but may have relevance as a biomarker for immunoreactivity towards OSP in myelin disorders. Our data demonstrate that in rhesus monkeys immune responses directed at OSP are encephalitogenic, leading to inflammatory responses throughout the central nervous system and to selective demyelination of the optic nerve.


Assuntos
Doenças Desmielinizantes/patologia , Encefalomielite Autoimune Experimental/imunologia , Proteínas do Tecido Nervoso/imunologia , Nervo Óptico/patologia , Sequência de Aminoácidos , Animais , Formação de Anticorpos/imunologia , Cegueira/etiologia , Cegueira/patologia , Sistema Nervoso Central/patologia , Claudinas , Citocinas/metabolismo , Encefalomielite Autoimune Experimental/complicações , Encefalomielite Autoimune Experimental/patologia , Epitopos de Linfócito B/imunologia , Epitopos de Linfócito T/imunologia , Feminino , Humanos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Ativação Linfocitária/imunologia , Macaca mulatta , Dados de Sequência Molecular , Esclerose Múltipla/imunologia , Proteínas do Tecido Nervoso/genética , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Proteínas Recombinantes/imunologia , Homologia de Sequência de Aminoácidos , Baço/citologia , Baço/imunologia , Baço/metabolismo , Linfócitos T/imunologia , Vacinação
18.
J Immunol ; 180(3): 1326-37, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18209026

RESUMO

The recombinant human (rh) myelin/oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE) model in the common marmoset is characterized by 100% disease incidence, a chronic disease course, and a variable time interval between immunization and neurological impairment. We investigated whether monkeys with fast and slow disease progression display different anti-MOG T or B cell responses and analyzed the underlying pathogenic mechanism(s). The results show that fast progressor monkeys display a significantly wider specificity diversification of anti-MOG T cells at necropsy than slow progressors, especially against MOG(34-56) and MOG(74-96). MOG(34-56) emerged as a critical encephalitogenic peptide, inducing severe neurological disease and multiple lesions with inflammation, demyelination, and axonal injury in the CNS. Although EAE was not observed in MOG(74-96)-immunized monkeys, weak T cell responses against MOG(34-56) and low grade CNS pathology were detected. When these cases received a booster immunization with MOG(34-56) in IFA, full-blown EAE developed. MOG(34-56)-reactive T cells expressed CD3, CD4, or CD8 and CD56, but not CD16. Moreover, MOG(34-56)-specific T cell lines displayed specific cytotoxic activity against peptide-pulsed B cell lines. The phenotype and cytotoxic activity suggest that these cells are NK-CTL. These results support the concept that cytotoxic cells may play a role in the pathogenesis of multiple sclerosis.


Assuntos
Autoimunidade , Encefalomielite Autoimune Experimental/imunologia , Glicoproteínas/imunologia , Ativação Linfocitária , Glicoproteína Associada a Mielina/imunologia , Fragmentos de Peptídeos/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Autoanticorpos/sangue , Autoanticorpos/imunologia , Encéfalo/imunologia , Encéfalo/patologia , Callithrix , Progressão da Doença , Feminino , Glicoproteínas/toxicidade , Humanos , Masculino , Proteínas da Mielina , Glicoproteína Associada a Mielina/toxicidade , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Peptídeos/toxicidade , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/toxicidade , Linfócitos T Citotóxicos/efeitos dos fármacos
19.
Expert Rev Clin Immunol ; 3(5): 749-61, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20477025

RESUMO

Biotechnology has enabled the development of specifically acting therapies for immune-mediated inflammatory disorders (IMIDs) based on biological molecules. The high species specificity precludes safety and effectivity testing in lower species (mice and rats), thus creating a need for valid experimental models in nonhuman primates (NHPs). Here, we review the creation of relevant NHP model(s) for multiple sclerosis (MS), an IMID of the human CNS. We will also discuss how the model(s) can help in the translation of a scientific principle developed in lower species into a therapy for MS.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...