Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Autoimmunity ; 50(6): 346-353, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28850269

RESUMO

Experimental autoimmune myasthenia gravis (EAMG), an animal model of myasthenia gravis (MG), can be induced in C57BL/6 (B6, H-2 b) mice by 2-3 injections with Torpedo californica AChR (tAChR) in complete Freund's adjuvant. Some EAMG mice exhibit weight loss with muscle weakness. The loss in body weight, which is closely associated with bone structure, is particularly evident in EAMG mice with severe muscle weakness. However, the relationship between muscle weakness and bone loss in EAMG has not been studied before. Recent investigations on bone have shed light on association of bone health and immunological states. It is possible that muscle weakness in EAMG developed by anti-tAChR immune responses might accompany bone loss. We determined whether reduced muscle strength associates with decreased bone mineral density (BMD) in EAMG mice. EAMG was induced by two injections at 4-week interval of tAChR and adjuvants in two different age groups. The first tAChR injection was either at age 8 weeks or at 15 weeks. We measured BMD at three skeletal sites, including femur, tibia, and lumbar vertebrae, using dual energy X-ray absorptiometry. Among these bone areas, femur of EAMG mice in both age groups showed a significant decrease in BMD compared to control adjuvant-injected and to non-immunized mice. Reduction in BMD in induced EAMG at a later-age appears to parallel the severity of the disease. The results indicate that anti-tAChR autoimmune response alone can reduce bone density in EAMG mice. BMD reduction was also observed in adjuvant-injected mice in comparison to normal un-injected mice, suggesting that BMD decrease can occur even when muscle activity is normal. Decreased BMD observed in both tAChR-injected and adjuvant-injected mice groups were discussed in relation to innate immunity and bone-related immunology involving activated T cells and tumour necrosis factor-related cytokines that trigger osteoclastogenesis and bone loss.


Assuntos
Densidade Óssea/imunologia , Reabsorção Óssea/patologia , Debilidade Muscular/patologia , Miastenia Gravis Autoimune Experimental/patologia , Absorciometria de Fóton , Fatores Etários , Animais , Reabsorção Óssea/induzido quimicamente , Reabsorção Óssea/diagnóstico por imagem , Reabsorção Óssea/imunologia , Fêmur/diagnóstico por imagem , Fêmur/imunologia , Fêmur/patologia , Proteínas de Peixes/administração & dosagem , Adjuvante de Freund/administração & dosagem , Vértebras Lombares/diagnóstico por imagem , Vértebras Lombares/imunologia , Vértebras Lombares/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Debilidade Muscular/induzido quimicamente , Debilidade Muscular/diagnóstico por imagem , Debilidade Muscular/imunologia , Miastenia Gravis Autoimune Experimental/induzido quimicamente , Miastenia Gravis Autoimune Experimental/diagnóstico por imagem , Miastenia Gravis Autoimune Experimental/metabolismo , Receptores Colinérgicos/administração & dosagem , Índice de Gravidade de Doença , Tíbia/diagnóstico por imagem , Tíbia/imunologia , Tíbia/patologia , Fatores de Tempo , Torpedo/metabolismo
2.
Immunol Invest ; 46(5): 490-499, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28375749

RESUMO

BACKGROUND: Myasthenia gravis (MG) is an autoimmune disease of the neuromuscular junction (NMJ), characterized with muscle weakness. While MG develops due to acetylcholine receptor (AChR) antibodies in most patients, antibodies to muscle-specific receptor tyrosine kinase (MuSK) or low-density lipoprotein receptor-related protein 4 (LRP4) may also be identified. Experimental autoimmune myasthenia gravis (EAMG) has been previously induced by both LRP4 immunization and passive transfer of LRP4 antibodies. OBJECTIVE: Our aim was to confirm previous results and to test the pathogenic effects of LRP4 immunization in a commonly used mouse strain C57BL/6 (B6) using a recombinantly expressed human LRP4 protein. METHODS: B6 mice were immunized with human LRP4 in CFA, Torpedo Californica AChR in CFA or only CFA. Clinical and pathogenic aspects of EAMG were compared among groups. RESULTS: LRP4- and AChR-immunized mice showed comparable EAMG clinical severity. LRP4-immunized mice displayed serum antibodies to LRP4 and NMJ IgG and complement factor C3 deposits. IgG2 was the dominant anti-LRP4 isotype. Cultured lymph node cells of LRP4- and AChR-immunized mice gave identical pro-inflammatory cytokine (IL-6, IFN-γ and IL-17) responses to LRP4 and AChR stimulation, respectively. CONCLUSION: Our results confirm the EAMG-inducing action of LRP4 immunization and identify B6 as a LRP4-EAMG-susceptible mouse strain. Demonstration of complement fixing anti-LRP4 antibodies in sera and complement/IgG deposits at the NMJ of LRP4-immunized mice indicates complement activation as a putative pathogenic mechanism. We have thus developed a practical LRP4-induced EAMG model using a non-conformational protein and a widely available mouse strain for future investigation of LRP4-related MG.


Assuntos
Ativação do Complemento/efeitos dos fármacos , Imunização/métodos , Imunoglobulina G/biossíntese , Proteínas Relacionadas a Receptor de LDL/administração & dosagem , Miastenia Gravis Autoimune Experimental/imunologia , Receptores Colinérgicos/administração & dosagem , Animais , Complemento C3/metabolismo , Adjuvante de Freund/administração & dosagem , Humanos , Isotipos de Imunoglobulinas/biossíntese , Interferon gama/biossíntese , Interferon gama/imunologia , Interleucina-17/biossíntese , Interleucina-17/imunologia , Interleucina-6/biossíntese , Interleucina-6/imunologia , Proteínas Relacionadas a Receptor de LDL/imunologia , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Linfonodos/patologia , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Linfócitos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miastenia Gravis Autoimune Experimental/induzido quimicamente , Miastenia Gravis Autoimune Experimental/patologia , Cultura Primária de Células , Receptores Colinérgicos/imunologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia , Índice de Gravidade de Doença , Torpedo/metabolismo
3.
J Neuroimmunol ; 303: 13-21, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28038891

RESUMO

Myasthenia gravis (MG) is usually caused by antibodies against the muscle acetylcholine receptor (AChR). Experimental autoimmune MG (EAMG) is the animal model of MG, typically induced by immunization of rodents with AChR isolated from the electric organ of Torpedo californica. We have successfully induced EAMG in Lewis rats by immunization with the extracellular domains (ECDs) of the human AChR subunits (α, ß, γ, δ and ε) expressed in yeast. Analysis of the antibody titers revealed a robust antigenic response against all the peptides, but a marked difference in their pathogenicity; the α subunit ECD was the most pathogenic, resulting in the highest percentage of affected animals. Measurements of antibody titers, electromyographic tests and quantitation of the muscle AChR content, offered further support to these findings. The EAMG models presented herein, could be used for studying subunit-specific pathogenic mechanisms, and, more importantly, as tools for the evaluation of antigen-specific therapeutic approaches, which rely on the human AChR.


Assuntos
Modelos Animais de Doenças , Miastenia Gravis Autoimune Experimental/induzido quimicamente , Miastenia Gravis Autoimune Experimental/imunologia , Receptores Colinérgicos/administração & dosagem , Receptores Colinérgicos/imunologia , Animais , Feminino , Humanos , Miastenia Gravis Autoimune Experimental/metabolismo , Domínios Proteicos/fisiologia , Subunidades Proteicas/imunologia , Subunidades Proteicas/toxicidade , Ratos , Ratos Endogâmicos Lew
4.
J Neuroimmunol ; 295-296: 84-92, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27235354

RESUMO

Sera of myasthenia gravis (MG) patients with muscle-specific receptor kinase-antibody (MuSK-Ab) predominantly display the non-complement fixing IgG4 isotype. Similarly, mouse IgG1, which is the analog of human IgG4, is the predominant isotype in mice with experimental autoimmune myasthenia gravis (EAMG) induced by MuSK immunization. The present study was performed to determine whether IgG1 anti-MuSK antibody is required for immunized mice to develop EAMG. Results demonstrated a significant correlation between clinical severity of EAMG and levels of MuSK-binding IgG1+, IgG2+ and IgG3+ peripheral blood B cells in MuSK-immunized wild-type (WT) mice. Moreover, MuSK-immunized IgG1 knockout (KO) and WT mice showed similar EAMG severity, serum MuSK-Ab levels, muscle acetylcholine receptor concentrations, neuromuscular junction immunoglobulin and complement deposit ratios. IgG1 and IgG3 were the predominant anti-MuSK isotypes in WT and IgG1 KO mice, respectively. These observations demonstrate that non-IgG1 isotypes can mediate MuSK-EAMG pathogenesis.


Assuntos
Imunoglobulina G/imunologia , Miastenia Gravis Autoimune Experimental/induzido quimicamente , Miastenia Gravis Autoimune Experimental/imunologia , Receptores Proteína Tirosina Quinases/toxicidade , Animais , Antígenos CD/imunologia , Antígenos CD/metabolismo , Autoanticorpos/sangue , Linfócitos B/metabolismo , Linfócitos B/patologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Adjuvante de Freund/toxicidade , Imunização , Imunoglobulina G/genética , Imunoglobulina G/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miastenia Gravis Autoimune Experimental/patologia , Junção Neuromuscular/imunologia , Junção Neuromuscular/metabolismo , Junção Neuromuscular/patologia , Receptores Colinérgicos , Índice de Gravidade de Doença , Estatísticas não Paramétricas
5.
J Neuroimmunol ; 289: 130-8, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26616882

RESUMO

IL-10-competent subset within CD1d(hi)CD5(+) B cells, also known as B10 cells, has been shown to regulate autoimmune diseases. In our previous study, adoptive transfer of CD1d(hi)CD5(+) B cells expanded in vivo by GM-CSF prevented and suppressed experimental autoimmune myasthenia gravis (EAMG). The goal of this study was to further examine the role and mechanism of IL-10 in the regulatory function of B10 cells in EAMG. We found that only IL-10 competent CD1d(hi)CD5(+) B cells sorted from WT mice, but not IL-10 deficient CD1d(hi)CD5(+) B cells exhibited regulatory function in vitro and in vivo. Adoptive transfer of IL-10 competent CD1d(hi)CD5(+) B cells led to higher frequency of Tregs and B10 cells, and low levels of proinflammatory cytokines and autoantibody production. We conclude that IL-10 production within CD1d(hi)CD5(+) B cells plays an important role in immune regulation of EAMG.


Assuntos
Antígenos CD1d/metabolismo , Subpopulações de Linfócitos B/química , Antígenos CD5/metabolismo , Interleucina-10/metabolismo , Miastenia Gravis Autoimune Experimental/imunologia , Miastenia Gravis Autoimune Experimental/patologia , Transferência Adotiva/métodos , Animais , Proliferação de Células/fisiologia , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Interleucina-10/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/metabolismo , Miastenia Gravis Autoimune Experimental/induzido quimicamente , Miastenia Gravis Autoimune Experimental/fisiopatologia , Fragmentos de Peptídeos/imunologia , Receptores Nicotínicos/imunologia
6.
Immunol Lett ; 168(2): 306-12, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26493475

RESUMO

Extraocular muscles (EOM) are preferentially involved in myasthenia gravis (MG) and acetylcholine receptor (AChR) antibody positive MG patients may occasionally present with isolated ocular symptoms. Although experimental autoimmune myasthenia gravis (EAMG) induced by whole AChR immunization closely mimics clinical and immunopathological aspects of MG, EOM are usually not affected. We have previously developed an EAMG model, which imitates EOM symptoms of MG by immunization of human leukocyte antigen (HLA) transgenic mice with α or γ-subunits of human AChR (H-AChR). To investigate the significance of the ϵ-subunit in ocular MG, we immunized HLA-DR3 and HLA-DQ8 transgenic mice with recombinant H-AChR ϵ-subunit expressed in Escherichia coli. HLA-DR3 transgenic mice showed significantly higher clinical ocular and generalized MG severity scores and lower grip strength values than HLA-DQ8 mice. H-AChR ϵ-subunit-immunized HLA-DR3 transgenic mice had higher serum anti-AChR antibody (IgG, IgG1, IgG2b, IgG2c and IgM) levels, neuromuscular junction IgG and complement deposit percentages than ϵ-subunit-immunized HLA-DQ8 transgenic mice. Control mice immunized with E. coli extract or complete Freund adjuvant (CFA) did not show clinical and immunopathological features of ocular and generalized EAMG. Lymph node cells of ϵ-subunit-immunized HLA-DR3 mice showed significantly higher proliferative responses than those of ϵ-subunit-immunized HLA-DQ8 mice, crude E. coli extract-immunized and CFA-immunized transgenic mice. Our results indicate that the human AChR ϵ-subunit is capable of inducing myasthenic muscle weakness. Diversity of the autoimmune responses displayed by mice expressing different HLA class II molecules suggests that the interplay between HLA class II alleles and AChR subunits might have a profound impact on the clinical course of MG.


Assuntos
Antígenos HLA-DQ/imunologia , Antígeno HLA-DR3/imunologia , Miastenia Gravis Autoimune Experimental/imunologia , Músculos Oculomotores/imunologia , Animais , Proliferação de Células , Ensaio de Imunoadsorção Enzimática , Adjuvante de Freund/imunologia , Antígenos HLA-DQ/genética , Antígeno HLA-DR3/genética , Humanos , Imunização/métodos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Imunoglobulina M/sangue , Imunoglobulina M/imunologia , Linfócitos/imunologia , Linfócitos/patologia , Camundongos Transgênicos , Microscopia de Fluorescência , Miastenia Gravis Autoimune Experimental/induzido quimicamente , Miastenia Gravis Autoimune Experimental/genética , Músculos Oculomotores/metabolismo , Músculos Oculomotores/patologia , Subunidades Proteicas/imunologia , Receptores Colinérgicos/imunologia
7.
J Immunol ; 193(10): 5044-55, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25288571

RESUMO

Myasthenia gravis (MG) and experimental autoimmune myasthenia gravis (EAMG) are caused by Ab-mediated autoimmune responses to muscle nicotinic acetylcholine receptors (AChRs) that impair neuromuscular transmission, thereby causing muscle weakness. Previously, we discovered that i.p. injection of a therapeutic vaccine consisting of bacterially expressed cytoplasmic domains of human AChR subunits reduced the development of chronic EAMG in rats. In this article, we show that immunization with the therapeutic vaccine in adjuvants does not induce EAMG and, thus, is safe. The potency and efficacy of the therapeutic vaccine were greatly increased by s.c. administration of repeated low doses in IFA. Onset of chronic EAMG could be prevented. Established chronic EAMG could be rapidly reversed, modeling therapy of chronic MG. Therapy reduced pathological Abs assayed by immune precipitation of a main immunogenic region chimera. Successfully treated rats exhibited long-term resistance to reinduction of EAMG, suggesting a lasting cure of MG. A long-term effect of therapy was to change the isotype of the pathogenic Ab response from IgG2b, which fixes complement, to IgG1, which does not. Prevention and reversal of chronic EAMG was not caused by the isotype switch, but the isotype switch may contribute to resistance to reinduction of EAMG. Immunization with AChR cytoplasmic domains in adjuvant is promising as a safe, Ag-specific, potent, effective, rapidly acting, and long-lasting therapeutic approach to MG.


Assuntos
Miastenia Gravis Autoimune Experimental/tratamento farmacológico , Subunidades Proteicas/imunologia , Receptores Colinérgicos/imunologia , Vacinas/imunologia , Animais , Autoanticorpos/biossíntese , Feminino , Humanos , Switching de Imunoglobulina/efeitos dos fármacos , Imunoglobulina G/biossíntese , Injeções Subcutâneas , Músculos/efeitos dos fármacos , Músculos/imunologia , Músculos/patologia , Miastenia Gravis Autoimune Experimental/induzido quimicamente , Miastenia Gravis Autoimune Experimental/imunologia , Miastenia Gravis Autoimune Experimental/patologia , Subunidades Proteicas/administração & dosagem , Subunidades Proteicas/química , Ratos , Ratos Endogâmicos Lew , Receptores Colinérgicos/administração & dosagem , Receptores Colinérgicos/química , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/química , Proteínas Recombinantes/imunologia , Torpedo , Vacinas/administração & dosagem , Vacinas/química
8.
PLoS One ; 9(2): e87840, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24505322

RESUMO

The ß2-adrenergic receptor agonist, albuterol, has been reported beneficial in treating several forms of congenital myasthenia. Here, for the first time, we examined the potential benefit of albuterol in a mouse model of anti-Muscle Specific Kinase (MuSK) myasthenia gravis. Mice received 15 daily injections of IgG from anti-MuSK positive patients, which resulted in whole-body weakness. At neuromuscular junctions in the tibialis anterior and diaphragm muscles the autoantibodies caused loss of postsynaptic acetylcholine receptors, and reduced the amplitudes of the endplate potential and spontaneous miniature endplate potential in the diaphragm muscle. Treatment with albuterol (8 mg/kg/day) during the two-week anti-MuSK injection series reduced the degree of weakness and weight loss, compared to vehicle-treated mice. However, the compound muscle action potential recorded from the gastrocnemius muscle displayed a decremental response in anti-MuSK-injected mice whether treated with albuterol or vehicle. Ongoing albuterol treatment did not increase endplate potential amplitudes compared to vehicle-treated mice nor did it prevent the loss of acetylcholine receptors from motor endplates. On the other hand, albuterol treatment significantly reduced the degree of fragmentation of endplate acetylcholine receptor clusters and increased the extent to which the remaining receptor clusters were covered by synaptophysin-stained nerve terminals. The results provide the first evidence that short-term albuterol treatment can ameliorate weakness in a robust mouse model of anti-MuSK myasthenia gravis. The results also demonstrate that it is possible for albuterol treatment to reduce whole-body weakness without necessarily reversing myasthenic impairment to the structure and function of the neuromuscular junction.


Assuntos
Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Albuterol/farmacologia , Autoanticorpos/toxicidade , Miastenia Gravis Autoimune Experimental/tratamento farmacológico , Animais , Autoanticorpos/imunologia , Feminino , Humanos , Camundongos , Músculo Esquelético/imunologia , Músculo Esquelético/patologia , Miastenia Gravis Autoimune Experimental/induzido quimicamente , Miastenia Gravis Autoimune Experimental/imunologia , Miastenia Gravis Autoimune Experimental/patologia , Junção Neuromuscular/imunologia , Junção Neuromuscular/patologia , Receptores Proteína Tirosina Quinases/imunologia
9.
Neurology ; 77(20): 1819-26, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22013178

RESUMO

OBJECTIVE: Muscle-specific receptor tyrosine kinase (MuSK) antibody-positive myasthenia gravis (MG) accounts for 5%-15% of autoimmune MG. MuSK mediates the agrin-signaling pathway and also anchors the collagenic tail subunit (ColQ) of acetylcholinesterase (AChE). The exact molecular target of MuSK-immunoglobulin G (IgG), however, remains elusive. As acetylcholine receptor (AChR) deficiency is typically mild and as cholinesterase inhibitors are generally ineffective, we asked if MuSK-IgG interferes with binding of ColQ to MuSK. METHODS: We used 3 assays: in vitro overlay of the human ColQ-tailed AChE to muscle sections of Colq-/- mice; in vitro plate-binding assay to quantitate binding of MuSK to ColQ and to LRP4; and passive transfer of MuSK-IgG to mice. RESULTS: The in vitro overlay assay revealed that MuSK-IgG blocks binding of ColQ to the neuromuscular junction. The in vitro plate-binding assay showed that MuSK-IgG exerts a dose-dependent block of MuSK binding to ColQ by but not to LRP4. Passive transfer of MuSK-IgG to mice reduced the size and density of ColQ to ∼10% of controls and had a lesser effect on the size and density of AChR and MuSK. CONCLUSIONS: As lack of ColQ compromises agrin-mediated AChR clustering in Colq-/- mice, a similar mechanism may lead to AChR deficiency in MuSK-MG patients. Our experiments also predict partial AChE deficiency in MuSK-MG patients, but AChE is not reduced in biopsied NMJs. In humans, binding of ColQ to MuSK may be dispensable for clustering ColQ, but is required for facilitating AChR clustering. Further studies will be required to elucidate the basis of this paradox.


Assuntos
Acetilcolinesterase/metabolismo , Anticorpos Bloqueadores/farmacologia , Autoanticorpos/farmacologia , Sítios de Ligação de Anticorpos , Colágeno/metabolismo , Proteínas Musculares/metabolismo , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/metabolismo , Receptores Colinérgicos/metabolismo , Animais , Colágeno/antagonistas & inibidores , Camundongos , Camundongos Knockout , Proteínas Musculares/antagonistas & inibidores , Miastenia Gravis Autoimune Experimental/induzido quimicamente , Miastenia Gravis Autoimune Experimental/imunologia , Miastenia Gravis Autoimune Experimental/metabolismo , Receptores Proteína Tirosina Quinases/imunologia , Receptores Colinérgicos/imunologia
10.
Immunology ; 128(1 Suppl): e826-36, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19740344

RESUMO

Experimental autoimmune myasthenia gravis (EAMG), an animal model of myasthenia gravis (MG), is a rare organ-specific autoimmune disease targeting the autoantigen nicotinic acetylcholine receptor (AChR). We show here that the balance of T helper type 1 (Th1), Th2, Th17 and regulatory T (Treg) subsets of CD4(+) helper T cells were redistributed during the development of EAMG and that the interleukin-17 (IL-17) cytokine is involved in this disease. The ratio of Th17 cells changed most notably with disease progression accompanied by an up-regulated level of IL-17. Moreover, the proliferative ability of AChR peptide-specific T cells and the anti-AChR antibody-secreting cells increased when stimulated by IL-17 in vitro. These findings suggested that the disequilibrium of the CD4(+) helper T-cell subsets could promote the development of EAMG, and the pathogenic mechanism by which Th17 cells drives autoimmune responses by secreting cytokine IL-17 provides a new target for myasthenia gravis therapy.


Assuntos
Interleucina-17/imunologia , Miastenia Gravis Autoimune Experimental/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Células Th2/imunologia , Animais , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Proliferação de Células/efeitos dos fármacos , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Miastenia Gravis Autoimune Experimental/induzido quimicamente , Ratos , Ratos Endogâmicos Lew , Receptores Nicotínicos/metabolismo , Baço/imunologia , Baço/patologia , Subpopulações de Linfócitos T/efeitos dos fármacos , Células Th1/metabolismo , Células Th2/metabolismo
11.
Muscle Nerve ; 40(2): 279-86, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19609914

RESUMO

Previous studies have shown increased expression of acetylcholine receptor-alpha (AChR-alpha) subunit transcripts in myasthenia gravis (MG) and experimental MG (EAMG), but none examined the functional properties of this overexpression. In this study we examined the mRNA and protein expression of AChR-alpha as well as the pattern of alpha-bungarotoxin labeling in muscle tissue from EAMG mice with varying disease severity. AChR-alpha expression was increased considerably in endplates from mice with severe EAMG, but it was distinct and greatly in excess of alpha-bungarotoxin labeling. This "aberrant expression" occurred in mice with morphologic endplate damage, and the pattern of complement and immunoglobulin deposition in muscle from these mice appeared to mirror the pattern of AChR-alpha expression. The loss of functional AChR in severe MG increases transcription of AChR-alpha mRNA, but the expressed protein is "functionally inert," failing to compensate for loss of AChR. This enhanced expression of AChR may play a role in driving the ongoing autoimmune response. Muscle Nerve 40: 279-286, 2009.


Assuntos
Regulação da Expressão Gênica/fisiologia , Miastenia Gravis Autoimune Experimental/metabolismo , Miastenia Gravis Autoimune Experimental/patologia , Receptores Colinérgicos/genética , Receptores Colinérgicos/metabolismo , Animais , Anticorpos/farmacologia , Bungarotoxinas/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Ensaio de Imunoadsorção Enzimática/métodos , Feminino , Adjuvante de Freund , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Músculo Esquelético/ultraestrutura , Miastenia Gravis Autoimune Experimental/induzido quimicamente , Ligação Proteica , RNA Mensageiro/metabolismo , Receptores Colinérgicos/imunologia , Receptores Colinérgicos/isolamento & purificação , Índice de Gravidade de Doença , Torpedo
12.
Eur J Immunol ; 39(3): 800-9, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19283707

RESUMO

In addition to their capacity to differentiate, BM stromal cells (BMSC) have immunosuppressive qualities that make them strong candidates for use in cell therapy against human autoimmune diseases. We studied the immunoregulatory activities of BMSC on experimental autoimmune myasthenia gravis (EAMG) in vitro and in vivo. Intravenous administration of syngenic BMSC to EAMG-model rats on the day of their second immunization was effective in ameliorating the pathological features of the disease. In vitro, the proliferative ability of T cells or B cells from EAMG rats was inhibited when they were cocultured with BMSC at proper ratios. This inhibitory effect was at least partially dependent on the secretion of IDO. We also determined that the development of EAMG is accompanied by an imbalance among the Th1, Th2, Th17, and Treg cell subsets, and that this can be corrected by the administration of BMSC, which leads to an increase of Th2 (IL-4) and Treg (Foxp3) cells, and a reduction of Th1 (IFN-gamma) and Th17 (IL-17) cells, through an IDO-dependent mechanism. These results provide further insights into the pathogenesis of MG, EAMG, and other immune-mediated diseases, and support a potential role for BMSC in their treatment.


Assuntos
Células da Medula Óssea/imunologia , Citocinas/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Miastenia Gravis Autoimune Experimental/imunologia , Células Estromais/imunologia , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Células da Medula Óssea/metabolismo , Citocinas/biossíntese , Feminino , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Miastenia Gravis Autoimune Experimental/induzido quimicamente , Miastenia Gravis Autoimune Experimental/metabolismo , Peptídeos/farmacologia , Ratos , Ratos Endogâmicos Lew , Células Estromais/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
13.
Ann Neurol ; 63(6): 782-9, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18384168

RESUMO

OBJECTIVE: A subset of myasthenia gravis patients that are seronegative for anti-acetylcholine receptor (anti-AChR) antibodies are instead seropositive for antibodies against the muscle-specific kinase (anti-MuSK-positive). Here, we test whether transfer of IgG from anti-MuSK-positive patients to mice confers impairment of the neuromuscular junction and muscle weakness. METHODS: IgG from anti-MuSK-positive myasthenia gravis patients or control IgG (seronegative for AChR and MuSK) was injected intraperitoneally (45 mg daily for 14 days) into 6-week-old female FVB/NJ and C57BL/6J mice. Changes at neuromuscular junctions in the tibialis anterior and diaphragm muscles were assessed by confocal fluorescent imaging of AChRs stained with fluorescent-alpha-bungarotoxin. Loss of function was assessed by electromyography. RESULTS: In experimental mice injected with anti-MuSK-positive patient IgG, postsynaptic AChR staining was reduced to as little as 22% of that seen in control mice. Experimental mice showed reduced apposition of the nerve terminal (labeled with antibodies against synaptophysin and neurofilament) and the postsynaptic AChR cluster (labeled with fluorescent-alpha-bungarotoxin). Mice injected with IgG from two of three anti-MuSK-positive patients lost weight and developed muscle weakness associated with a decremental electromyographic trace on repetitive nerve stimulation. INTERPRETATION: IgG from anti-MuSK-positive patients can cause myasthenia gravis when injected into mice. This may be explained by a progressive reduction in the density of postsynaptic AChR combined with changes in the nerve terminal and its relation to the postsynaptic structure.


Assuntos
Autoanticorpos/imunologia , Imunoglobulina G/imunologia , Miastenia Gravis/imunologia , Junção Neuromuscular/imunologia , Junção Neuromuscular/fisiopatologia , Receptores Proteína Tirosina Quinases/imunologia , Receptores Colinérgicos/imunologia , Acetilcolina/metabolismo , Animais , Autoanticorpos/toxicidade , Humanos , Imunoglobulina G/toxicidade , Camundongos , Debilidade Muscular/induzido quimicamente , Debilidade Muscular/imunologia , Debilidade Muscular/fisiopatologia , Miastenia Gravis/fisiopatologia , Miastenia Gravis Autoimune Experimental/induzido quimicamente , Miastenia Gravis Autoimune Experimental/imunologia , Miastenia Gravis Autoimune Experimental/fisiopatologia , Junção Neuromuscular/efeitos dos fármacos , Membranas Sinápticas/efeitos dos fármacos , Membranas Sinápticas/imunologia , Membranas Sinápticas/patologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/imunologia
14.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 23(8): 737-40, 2007 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-17618568

RESUMO

AIM: To investigate the effect and immuoregulative mechanisms of NK1.1(+) cells on the development of experimental autoimmune myasthenia gravis (EAMG). METHODS: The NK1.1(+) cells were depleted by intraperitoneal (i.p.) administration of anti-mouse NK1.1 mAb to C57BL/6 mice. Mice were immunized subcutaneously with AChR in CFA. The incidence and severity of EAMG was determined according to the Lennon disease symptoms grading. IFN-gamma and IL-4 in MNCs culture medium were measured by ELISA. AChR IgG of serum was measured by radioimmunoassay. In some experiments, the anti-IFN-gamma mAb was injected (i.p.) to neutralize IFN-gamma. RESULTS: The onset of EAMG was delayed and the severity was decreased obviously in NK1.1(+) cell-depleted mice. However, depletion of NK1.1(+) cells after immunization had no impact on the development of EAMG. Depletion of NK1.1(+) cells could significantly reduce the expression of AChR-specific antibody as well as the production of IFN-gamma. The development of EAMG and production of AChR specific Ab in NK1.1(+) cell-depleted mice were decreased obviously after treating with anti-IFN-gamma antibody. CONCLUSION: NK1.1(+) cells are involved in the early EAMG, and NK1.1(+) cells could enhance the production of IFN-gamma released by AChR-specific T cells as well as the AChR-specific antibodies, which may enhance the outcome of EAMG.


Assuntos
Células Matadoras Naturais/imunologia , Miastenia Gravis Autoimune Experimental/imunologia , Animais , Anticorpos/imunologia , Anticorpos/farmacologia , Ensaio de Imunoadsorção Enzimática , Feminino , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-4/metabolismo , Células Matadoras Naturais/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Miastenia Gravis Autoimune Experimental/induzido quimicamente , Miastenia Gravis Autoimune Experimental/patologia , Radioimunoensaio , Distribuição Aleatória , Receptores Nicotínicos/imunologia
15.
Cell Immunol ; 241(2): 95-101, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17005165

RESUMO

Dendritic cells (DC) are highly specialized antigen presenting cells that play critical roles as instigators and regulators of immune responses including B cell function, antibody synthesis and isotype switch. In this study, we compared immunotherapeutic effect of IL-10-treated DC (IL-10-DC) via both intraperitoneal (i.p.) and subcutaneous (s.c.) delivery in rats with incipient experimental autoimmune myasthenia gravis (EAMG). Spleen DC were isolated from onset of EAMG on day 39 post-immunization, exposed in vitro to IL-10, and then injected into incipient EAMG at dose of 1 x 10(6) cells/rat on day 5 after immunization. Intraperitoneal administration of IL-10-DC suppressed clinical scores, anti-acetylcholine receptors (AChR) antibody secreting cells, antigen-specific IL-10/IFN-gamma production and T cell proliferation compared to control EAMG rats. Importantly, IL-10-DC, if given by s.c. route, failed to ameliorate clinical sign of EAMG. Simultaneously, T cell proliferation, anti-AChR antibody secreting cells and IL-10/IFN-gamma production had no alteration, as compared to control EAMG rats. Both in vitro and in vivo experiments showed that treatment of IL-10 inhibited the migration of DC toward MIP-3beta and lymph node, indicating that in vitro manipulation of DC with IL-10 alters the migration of DC that influences the therapeutic effect in the treatment of autoimmune diseases. In MG patients, neither the improvement of clinical symptom nor the alteration of immunological parameter was observed through s.c. delivery of IL-10-DC, suggesting the limitation of IL-10-DC in the treatment of MG patients.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/transplante , Imunoterapia , Interleucina-10/farmacologia , Miastenia Gravis Autoimune Experimental/terapia , Miastenia Gravis/terapia , Animais , Linfócitos B/imunologia , Movimento Celular , Proliferação de Células , Células Dendríticas/citologia , Células Dendríticas/imunologia , Feminino , Humanos , Imunoglobulina G/imunologia , Injeções Intraperitoneais , Injeções Subcutâneas , Interferon gama/biossíntese , Interleucina-10/administração & dosagem , Interleucina-10/biossíntese , Linfonodos/imunologia , Miastenia Gravis/imunologia , Miastenia Gravis Autoimune Experimental/induzido quimicamente , Miastenia Gravis Autoimune Experimental/imunologia , Miastenia Gravis Autoimune Experimental/patologia , Ratos , Ratos Endogâmicos Lew , Receptores Colinérgicos/imunologia , Linfócitos T/imunologia
16.
Exp Neurol ; 202(2): 287-93, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16859686

RESUMO

In myasthenia gravis (MG), neuromuscular transmission is disrupted due to the production of autoantibodies against acetylcholine receptors (AChR). In previous work, we showed that decay accelerating factor (DAF or CD55), an intrinsic cell surface complement regulator that disables C3/C5 amplification convertases, protects against receptor loss and muscle weakness. In this study, we examined whether, and if so, to what extent CD59, a downstream intrinsic cell surface regulator that prevents assembly of membrane attack complexes (MACs), contributes to this protection. Twenty-four hours after anti-AChR injection, we found that CD59a-/- mice did not significantly differ from WTs, all Daf1-/- CD59a-/- mice either died or required euthanasia. At 48h, Daf1-/- were significantly weaker than CD59a-/- and WT mice, and for these mice immunohistochemistry revealed marked C9 deposition at postsynaptic junctions, radioimmunoassays showed reductions in AChR levels, and electron microscopy demonstrated massive junctional damage. These data indicate that DAF serves as the initial shield that protects the neuromuscular junction whereas CD59 is a further barrier. They argue that complement inhibitor, particularly if targeted to the receptor, could then have therapeutic value in human MG.


Assuntos
Antígenos CD55/fisiologia , Antígenos CD59/fisiologia , Miastenia Gravis Autoimune Experimental/genética , Miastenia Gravis Autoimune Experimental/metabolismo , Miastenia Gravis Autoimune Experimental/fisiopatologia , Análise de Variância , Animais , Anticorpos Monoclonais , Antígenos CD55/genética , Antígenos CD59/genética , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Modelos Animais de Doenças , Imuno-Histoquímica/métodos , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão/métodos , Debilidade Muscular/etiologia , Músculo Esquelético/metabolismo , Miastenia Gravis Autoimune Experimental/induzido quimicamente , Junção Neuromuscular/patologia , Junção Neuromuscular/ultraestrutura , Receptores Colinérgicos/imunologia , Receptores Colinérgicos/metabolismo
17.
Neurosci Res ; 55(1): 40-4, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16504322

RESUMO

The neuromuscular weakness associated with myasthenia gravis (MG) can be transiently relieved by pharmacological inhibitors of acetylcholinesterase (AChE). Here, we expand the anticholinesterase repertoire to include 2'-O-methyl-protected antisense oligonucleotides targeted to AChE mRNA (EN101). Using stimulated-single fiber electromyography, we show that EN101 treatment of rats with experimental autoimmune myasthenia gravis (EAMG), improved the mean consecutive difference (MCD) and blocking for 24h. This treatment was more efficient than pyridostigmine and was accompanied by marked improvement in stamina and clinical profile.


Assuntos
Eletromiografia , Músculos/fisiopatologia , Miastenia Gravis Autoimune Experimental/tratamento farmacológico , Miastenia Gravis Autoimune Experimental/fisiopatologia , Oligonucleotídeos Antissenso/uso terapêutico , Acetilcolinesterase/genética , Animais , Estimulação Elétrica/métodos , Teste de Esforço/métodos , Feminino , Monitorização Fisiológica , Músculos/efeitos dos fármacos , Músculos/efeitos da radiação , Miastenia Gravis Autoimune Experimental/induzido quimicamente , Junção Neuromuscular/efeitos dos fármacos , Junção Neuromuscular/fisiopatologia , Junção Neuromuscular/efeitos da radiação , Oligodesoxirribonucleotídeos , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos Lew , Receptores Colinérgicos/imunologia , Fatores de Tempo
18.
J Neuroimmunol ; 174(1-2): 157-67, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16527362

RESUMO

Immune complexes and classical complement pathway play vital roles in experimental autoimmune myasthenia gravis (EAMG). To analyze the role of immune complex receptors in EAMG, FcgammaRIII knockout (KO) mice were immunized with AChR and were found out to be resistant to EAMG induction. This was associated with reduced neuromuscular junction deposits, lymph node cell (LNC) IL-6 production and serum complement levels. EAMG resistance of anti-C1q Ab-administered mice was also associated with reduced LNC IL-6 production and neuromuscular junction deposits, indicating C1q involvement in EAMG resistance. The data provide the first direct genetic evidence for Fcgamma receptor involvement in EAMG pathogenesis.


Assuntos
Miastenia Gravis Autoimune Experimental/genética , Miastenia Gravis Autoimune Experimental/patologia , Receptores de IgG/fisiologia , Animais , Anticorpos/administração & dosagem , Antígenos CD/metabolismo , Complemento C1q/imunologia , Complemento C1q/metabolismo , Convertases de Complemento C3-C5/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática/métodos , Citometria de Fluxo/métodos , Imunoglobulina G/metabolismo , Imuno-Histoquímica/métodos , Interleucina-6/metabolismo , Linfonodos/efeitos dos fármacos , Linfonodos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/fisiopatologia , Miastenia Gravis Autoimune Experimental/induzido quimicamente , Miastenia Gravis Autoimune Experimental/fisiopatologia , Junção Neuromuscular/metabolismo , Junção Neuromuscular/patologia , Radioimunoensaio/métodos , Receptores de IgG/deficiência , Receptores de IgG/genética , Receptores Nicotínicos/genética , Estatísticas não Paramétricas , Fatores de Tempo
19.
Artigo em Inglês | MEDLINE | ID: mdl-15974179

RESUMO

Whole blood immunoadsorption (WBIA) system, using an adsorbent to remove pathogenic antibodies of myasthenia gravis (MG), was studied. Cellulose-tryptophan adsorbent was synthesized and its adsorption capacity of binding with acetylcholine receptor in the plasma of MG patient was evaluated. Experimental autoimmune myasthenia gravis (EAMG) rabbits were induced by Ta183-200 peptide. The rabbits underwent extracorporeal whole blood adsorption for 2 h. Results showed no significant damages on blood cells and no changes in the concentration of electrolytes. Total protein decreased by 12.0% (P < 0.05), and globulin protein decreased 23.9 +/- 5.6% (P < 0.05). The mean overall removal of antibodies against Ta183-200 was 41.12%. The percentage of decrement of compound muscle action potential in 3, 5, 10Hz of EAMG rabbits all dropped down after the treatment. In conclusion, the adsorbent is biocompatible, was safe for whole blood immunoadsorption, and can remove antibodies in an MG patient effectively. Whole blood immunoadsorption improved clinical manifestation and neuromuscular function of the EAMG rabbits.


Assuntos
Autoanticorpos , Hemofiltração/métodos , Imunoadsorventes/uso terapêutico , Miastenia Gravis Autoimune Experimental/terapia , Animais , Autoanticorpos/imunologia , Celulose/química , Celulose/uso terapêutico , Feminino , Imunoadsorventes/química , Miastenia Gravis Autoimune Experimental/induzido quimicamente , Miastenia Gravis Autoimune Experimental/imunologia , Coelhos , Receptores Colinérgicos/química , Triptofano/química , Triptofano/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...