Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 10: 2949, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31956324

RESUMEN

Beneficial effects of probiotics on gut microbiota homeostasis and inflammatory immune responses suggested the investigation of their potential clinical efficacy in experimental models of autoimmune diseases. Indeed, administration of two bifidobacteria and lactobacilli probiotic strains prevented disease manifestations in the Lewis rat model of Myasthenia Gravis (EAMG). Here, we demonstrate the clinical efficacy of therapeutic administration of vital bifidobacteria (i.e., from EAMG onset). The mechanisms involved in immunomodulation were investigated with ex vivo and in vitro experiments. Improvement of EAMG symptoms was associated to decreased anti-rat AChR antibody levels, and differential expression of TGFß and FoxP3 immunoregulatory transcripts in draining lymph nodes and spleen of treated-EAMG rats. Exposure of rat bone marrow-derived dendritic cells to bifidobacteria or lactobacilli strains upregulated toll-like receptor 2 mRNA expression, a key molecule involved in bacterium recognition via lipotheicoic acid. Live imaging experiments of AChR-specific effector T cells, co-cultured with BMDCs pre-exposed to bifidobacteria, demonstrated increased percentages of motile effector T cells, suggesting a hindered formation of TCR-peptide-MHC complex. Composition of gut microbiota was studied by 16S rRNA gene sequencing, and α and ß diversity were determined in probiotic treated EAMG rats, with altered ratios between Tenericutes and Verrucomicrobia (phylum level), and Ruminococcaceae and Lachnospiraceae (family level). Moreover, the relative abundance of Akkermansia genus was found increased compared to healthy and probiotic treated EAMG rats. In conclusion, our findings confirms that the administration of vital bifidobacteria at EAMG onset has beneficial effects on disease progression; this study further supports preclinical research in human MG to evaluate probiotic efficacy as supplementary therapy in MG.


Asunto(s)
Bifidobacterium , Miastenia Gravis Autoinmune Experimental/etiología , Probióticos/administración & dosificación , Animales , Autoinmunidad , Movimiento Celular , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Microbioma Gastrointestinal , Secuenciación de Nucleótidos de Alto Rendimiento , Metagenoma , Metagenómica/métodos , ARN Ribosómico 16S , Ratas , Ratas Endogámicas Lew
2.
Front Immunol ; 9: 2399, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30410484

RESUMEN

The guanine nucleotide exchange factor Vav1 is essential for transducing T cell receptor (TCR) signals and plays an important role in T cell development and activation. Previous genetic studies identified a natural variant of Vav1 characterized by the substitution of an arginine (R) residue by a tryptophane (W) at position 63 (Vav1R63W). This variant impacts Vav1 adaptor functions and controls susceptibility to T cell-mediated neuroinflammation. To assess the implication of this Vav1 variant on the susceptibility to antibody-mediated diseases, we used the animal model of myasthenia gravis, experimental autoimmune myasthenia gravis (EAMG). To this end, we generated a knock-in (KI) mouse model bearing a R to W substitution in the Vav1 gene (Vav1R63W) and immunized it with either torpedo acetylcholine receptor (tAChR) or the α146-162 immunodominant peptide. We observed that the Vav1R63W conferred increased susceptibility to EAMG, revealed by a higher AChR loss together with an increased production of effector cytokines (IFN-γ, IL-17A, GM-CSF) by antigen-specific CD4+ T cells, as well as an increased frequency of antigen-specific CD4+ T cells. This correlated with the emergence of a dominant antigen-specific T cell clone in KI mice that was not present in wild-type mice, suggesting an impact on thymic selection and/or a different clonal selection threshold following antigen encounter. Our results highlight the key role of Vav1 in the pathophysiology of EAMG and this was associated with an impact on the TCR repertoire of AChR reactive T lymphocytes.


Asunto(s)
Variación Genética , Miastenia Gravis Autoinmune Experimental/etiología , Miastenia Gravis Autoinmune Experimental/metabolismo , Proteínas Proto-Oncogénicas c-vav/genética , Proteínas Proto-Oncogénicas c-vav/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Citocinas/metabolismo , Susceptibilidad a Enfermedades , Ratones , Miastenia Gravis Autoinmune Experimental/patología , Fenotipo , Receptores Nicotínicos/metabolismo , Especificidad del Receptor de Antígeno de Linfocitos T
3.
Ann N Y Acad Sci ; 1412(1): 54-61, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29125188

RESUMEN

While the majority of myasthenia gravis patients express antibodies targeting the acetylcholine receptor, the second most common cohort instead displays autoantibodies against muscle-specific kinase (MuSK). MuSK is a transmembrane tyrosine kinase found in the postsynaptic membrane of the neuromuscular junction. During development, MuSK serves as a signaling hub, coordinating the alignment of the pre- and postsynaptic components of the synapse. Adult mice that received repeated daily injections of IgG from anti-MuSK+ myasthenia gravis patients developed muscle weakness, associated with neuromuscular transmission failure. MuSK autoantibodies are predominantly of the IgG4 type. They suppress the kinase activity of MuSK and the phosphorylation of target proteins in the postsynaptic membrane. Loss of postsynaptic acetylcholine receptors is the primary cause of neuromuscular transmission failure. MuSK autoantibodies also disrupt the capacity of the motor nerve terminal to adaptively increase acetylcholine release in response to the reduced postsynaptic responsiveness to acetylcholine. The passive IgG transfer model of MuSK myasthenia gravis has been used to test candidate treatments. Pyridostigmine, a first-line cholinesterase inhibitor drug, exacerbated the disease process, while 3,4-diaminopyridine and albuterol were found to be beneficial in this mouse model.


Asunto(s)
Miastenia Gravis Autoinmune Experimental/etiología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/inmunología , Animales , Inhibidores de la Colinesterasa/farmacología , Femenino , Humanos , Inmunización Pasiva , Ratones , Proteínas Musculares/metabolismo , Miastenia Gravis Autoinmune Experimental/inmunología , Miastenia Gravis Autoinmune Experimental/fisiopatología , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores Colinérgicos/inmunología , Receptores Colinérgicos/metabolismo , Sinapsis/inmunología , Sinapsis/fisiología
4.
Oncotarget ; 7(7): 7550-62, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26771137

RESUMEN

Abnormal overexpression of CXCL13 is observed in many inflamed tissues and in particular in autoimmune diseases. Myasthenia gravis (MG) is a neuromuscular disease mainly mediated by anti-acetylcholine receptor autoantibodies. Thymic hyperplasia characterized by ectopic germinal centers (GCs) is a common feature in MG and is correlated with high levels of anti-AChR antibodies. We previously showed that the B-cell chemoattractant, CXCL13 is overexpressed by thymic epithelial cells in MG patients. We hypothesized that abnormal CXCL13 expression by the thymic epithelium triggered B-cell recruitment in MG. We therefore created a novel transgenic (Tg) mouse with a keratin 5 driven CXCL13 expression. The thymus of Tg mice overexpressed CXCL13 but did not trigger B-cell recruitment. However, in inflammatory conditions, induced by Poly(I:C), B cells strongly migrated to the thymus. Tg mice were also more susceptible to experimental autoimmune MG (EAMG) with stronger clinical signs, higher titers of anti-AChR antibodies, increased thymic B cells, and the development of germinal center-like structures. Consequently, this mouse model finally mimics the thymic pathology observed in human MG. Our data also demonstrated that inflammation is mandatory to reveal CXCL13 ability to recruit B cells and to induce tertiary lymphoid organ development.


Asunto(s)
Linfocitos B/patología , Quimiocina CXCL13/fisiología , Inflamación/complicaciones , Miastenia Gravis Autoinmune Experimental/patología , Hiperplasia del Timo/fisiopatología , Animales , Linfocitos B/metabolismo , Células Cultivadas , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Citometría de Flujo , Centro Germinal/metabolismo , Centro Germinal/patología , Humanos , Técnicas para Inmunoenzimas , Inflamación/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miastenia Gravis Autoinmune Experimental/etiología , Miastenia Gravis Autoinmune Experimental/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
Eur J Immunol ; 45(5): 1339-47, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25676041

RESUMEN

The role of Th17 cells in the pathogenesis of autoantibody-mediated diseases is unclear. Here, we assessed the contribution of Th17 cells to the pathogenesis of experimental autoimmune myasthenia gravis (EAMG), which is induced by repetitive immunizations with Torpedo californica acetylcholine receptor (tAChR). We show that a significant fraction of tAChR-specific CD4(+) T cells is producing IL-17. IL-17(ko) mice developed fewer or no EAMG symptoms, although the frequencies of tAChR-specific CD4(+) T cells secreting IL-2, IFN-γ, or IL-21, and the percentage of FoxP3(+) Treg cells were similar to WT mice. Even though the total anti-tAChR antibody levels were equal, the complement fixating IgG2b subtype was reduced in IL-17(ko) as compared to WT mice. Most importantly, pathogenic anti-murine AChR antibodies were significantly lower in IL-17(ko) mice. Furthermore, we confirmed the role of Th17 cells in EAMG pathogenesis by the reconstitution of TCR ß/δ(ko) mice with WT or IL-17(ko) CD4(+) T cells. In conclusion, we show that the level of IgG2b and the loss of B-cell tolerance, which results in pathogenic anti-murine AChR-specific antibodies, are dependent on IL-17 production by CD4(+) T cells. Thus, we describe here for the first time how Th17 cells are involved in the induction of classical antibody-mediated autoimmunity.


Asunto(s)
Linfocitos B/inmunología , Interleucina-17/biosíntesis , Miastenia Gravis Autoinmune Experimental/inmunología , Células Th17/inmunología , Traslado Adoptivo , Animales , Antígenos/administración & dosificación , Tolerancia Inmunológica , Inmunoglobulina G/biosíntesis , Interleucina-17/deficiencia , Interleucina-17/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miastenia Gravis Autoinmune Experimental/etiología , Receptores de Antígenos de Linfocitos T alfa-beta/deficiencia , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/deficiencia , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Receptores Colinérgicos/inmunología , Torpedo/inmunología
6.
Muscle Nerve ; 45(2): 209-16, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22246876

RESUMEN

INTRODUCTION: HLA-DQ8 transgenic mice develop ocular myasthenia gravis (oMG), which then progresses to generalized MG (gMG) when immunized with the human acetylcholine receptor (H-AChR) α subunit. Because the fetal AChR γ subunit is expressed in adult extraocular muscles, we anticipated that γ subunit immunization would generate an immune response to mouse AChR and induce MG in mice. RESULTS: H-AChR γ subunit immunization in HLA-DQ8 mice induced an autoimmune response to mouse AChR and led to the destruction of AChR in the neuromuscular junction (NMJ) by anti-AChR antibody and complement activation, and it triggered upregulation of AChR gene transcription. CONCLUSION: Our findings indicate that oMG may be induced by immunity to the AChR γ subunit.


Asunto(s)
Inmunización/efectos adversos , Miastenia Gravis Autoinmune Experimental/etiología , Receptores Nicotínicos/inmunología , Animales , Anticuerpos Antiidiotipos/sangre , Anticuerpos Antiidiotipos/inmunología , Proteínas de Arabidopsis/metabolismo , Proliferación Celular , Complemento C3/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Antígenos HLA-DQ/genética , Humanos , Transferasas Intramoleculares/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Transgénicos , Fuerza Muscular/genética , Miastenia Gravis Autoinmune Experimental/sangre , Miastenia Gravis Autoinmune Experimental/genética , Miastenia Gravis Autoinmune Experimental/patología , Unión Neuromuscular/inmunología , Trastornos de la Motilidad Ocular/etiología , Trastornos de la Motilidad Ocular/inmunología , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo
8.
Clin Immunol ; 141(1): 36-48, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21570918

RESUMEN

RAGE belongs to immunoglobulin superfamily and serves as a ligand for various immunoregulatory molecules including S100B that has been demonstrated important to T cell mediated autoimmune diseases. In this context, we hypothesized that RAGE could also impact B cell mediated, T cell-dependent autoimmune diseases. This was tested using myasthenia gravis (MG) animal model, EAMG. We show that expression of both RAGE and S100B are increased during EAMG and the interaction between RAGE and S100B affected the Th1/Th2/Th17/Treg cell equilibrium, up-regulate AChR-specific T cell proliferation. Furthermore, addition of S100B in vitro stimulated splenocyte activity linked to COX-2 up-regulation. NS-398, a selective COX-2 inhibitor, effectively diminished S100B mediated activity of AChR-specific antibody secreting splenocytes. These findings suggested that a reciprocal relationship between RAGE and S100B promoted the development of EAMG, highlighting the importance of understanding the mechanisms of EAMG disease as a means of developing new therapies for the treatment of MG.


Asunto(s)
Miastenia Gravis Autoinmune Experimental/etiología , Miastenia Gravis Autoinmune Experimental/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Autoanticuerpos/biosíntesis , Proliferación Celular , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Femenino , Técnicas In Vitro , Ligandos , Miastenia Gravis Autoinmune Experimental/inmunología , Miastenia Gravis Autoinmune Experimental/patología , Proteína Básica de Mielina/genética , Proteína Básica de Mielina/inmunología , Factores de Crecimiento Nervioso/metabolismo , Factores de Crecimiento Nervioso/farmacología , Nitrobencenos/farmacología , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Ratas , Ratas Endogámicas Lew , Receptor para Productos Finales de Glicación Avanzada , Receptores Colinérgicos/genética , Receptores Colinérgicos/inmunología , Receptores Colinérgicos/metabolismo , Subunidad beta de la Proteína de Unión al Calcio S100 , Proteínas S100/metabolismo , Proteínas S100/farmacología , Sulfonamidas/farmacología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/patología , Regulación hacia Arriba
9.
Ann Neurol ; 65(1): 67-75, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19194881

RESUMEN

OBJECTIVE: Complement mediated injury of the neuromuscular junction is considered a primary disease mechanism in human myasthenia gravis and animal models of experimentally acquired myasthenia gravis (EAMG). We utilized active and passive models of EAMG to investigate the efficacy of a novel C5 complement inhibitor rEV576, recombinantly produced protein derived from tick saliva, in moderating disease severity. METHODS: Standardized disease severity assessment, serum complement hemolytic activity, serum cytotoxicity, acetylcholine receptor (AChR) antibody concentration, IgG subclassification, and C9 deposition at the neuromuscular junction were used to assess the effect of complement inhibition on EAMG induced by administration of AChR antibody or immunization with purified AChR. RESULTS: Administration of rEV576 in passive transfer EAMG limited disease severity as evidenced by 100% survival rate and a low disease severity score. In active EAMG, rats with severe and mild EAMG were protected from worsening of disease and had limited weight loss. Serum complement activity (CH(50)) in severe and mild EAMG was reduced to undetectable levels during treatment, and C9 deposition at the neuromuscular junction was reduced. Treatment with rEV576 resulted in reduction of toxicity of serum from severe and mild EAMG rats. Levels of total AChR IgG, and IgG(2a) antibodies were similar, but unexpectedly, the concentration of complement fixing IgG(1) antibodies was lower in a group of rEV576-treated animals, suggesting an effect of rEV576 on cellular immunity. INTERPRETATION: Inhibition of complement significantly reduced weakness in two models of EAMG. C5 inhibition could prove to be of significant therapeutic value in human myasthenia gravis.


Asunto(s)
Complemento C5/antagonistas & inhibidores , Proteínas Inactivadoras de Complemento/uso terapéutico , Miastenia Gravis Autoinmune Experimental/prevención & control , Animales , Anticuerpos/efectos adversos , Proteínas de Artrópodos , Línea Celular Tumoral , Complemento C9/metabolismo , Proteínas Inactivadoras de Complemento/inmunología , Proteínas Inactivadoras de Complemento/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Inmunoglobulina G/sangre , Inmunoglobulina G/clasificación , Proteínas de Insectos/inmunología , Proteínas de Insectos/uso terapéutico , Fuerza Muscular/efectos de los fármacos , Miastenia Gravis Autoinmune Experimental/etiología , Miastenia Gravis Autoinmune Experimental/inmunología , Miastenia Gravis Autoinmune Experimental/patología , Unión Neuromuscular/efectos de los fármacos , Unión Neuromuscular/metabolismo , Ratas , Ratas Endogámicas Lew , Receptores Colinérgicos/inmunología , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/patología , Proteínas y Péptidos Salivales/sangre , Proteínas y Péptidos Salivales/inmunología , Proteínas y Péptidos Salivales/uso terapéutico , Índice de Severidad de la Enfermedad , Factores de Tiempo , Pérdida de Peso/efectos de los fármacos
10.
J Neuroimmunol ; 201-202: 111-20, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18667242

RESUMEN

Currently used non-specific immunosuppressive drugs often require intervention in myasthenia gravis (MG) and clinical improvement varies widely. To analyze the therapeutic effect of mycophenolate mofetil (MMF) in experimental autoimmune MG (EAMG), rats were immunized with acetylcholine receptors (AChRs) and subsequently treated with MMF or vehicle. MMF treatment resulted in a significant suppression of anti-rat AChR antibody titers. Interestingly, no abnormalities of neuromuscular transmission and adverse side effects were detected in MMF-treated EAMG animals. Moreover, anti-rat AChR antibody titers correlated to an improvement of clinical outcome. In conclusion, our data suggest that MMF acts as a potent immunosuppressant drug in EAMG.


Asunto(s)
Inmunosupresores/uso terapéutico , Miastenia Gravis Autoinmune Experimental/tratamiento farmacológico , Ácido Micofenólico/análogos & derivados , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Potenciales de Acción/efectos de la radiación , Análisis de Varianza , Animales , Anticuerpos/sangre , Modelos Animales de Enfermedad , Electromiografía/métodos , Femenino , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Miastenia Gravis Autoinmune Experimental/sangre , Miastenia Gravis Autoinmune Experimental/etiología , Miastenia Gravis Autoinmune Experimental/patología , Ácido Micofenólico/uso terapéutico , Ratas , Ratas Endogámicas Lew , Receptores Colinérgicos/inmunología , Receptores Colinérgicos/metabolismo , Estadísticas no Paramétricas , Factores de Tiempo , Proteínas de Transporte Vesicular de Acetilcolina/metabolismo
11.
Invest Ophthalmol Vis Sci ; 48(11): 5101-11, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17962462

RESUMEN

PURPOSE: To establish a novel model of autoimmune ocular myasthenia gravis (oMG) in mice and study the pathogenic mechanisms of oMG. METHODS: oMG was induced in HLA-DQ8 transgenic, HLA-DR3 transgenic, major histocompatibility complex (MHC) class II-deficient, C57BL/6, and C57BL/10 mice by immunization with an Escherichia coli plasmid expressing the recombinant human acetylcholine receptor (AChR) alpha subunit. RESULTS: All strains of immunized mice developed ocular myasthenia gravis with varying disease incidence and severity. HLA-DQ8 transgenic mice were highly susceptible to oMG. Mice with oMG had serum autoantibodies to the mouse extraocular AChR, pathologic deposits of IgG, C3, and C5b-C9 in their extraocular and limb neuromuscular junctions, and droopiness of eyelids. HLA-DR3 transgenic and MHC class II-deficient mice were relatively resistant to oMG induced by AChR alpha subunit immunization and had minimal ocular abnormalities. CONCLUSIONS: These findings suggest that oMG pathogenesis could be triggered by immunity to the human AChR alpha subunit and that MHC class II molecule is required for human AChR alpha subunit presentation and CD4 cell-mediated anti-AChR antibody class switching. Differential oMG susceptibility observed in DQ8 and DR3 transgenic mice correlated with the intensity of lymphocytes to respond to the human AChR alpha subunit. This new model of oMG will be a valuable tool for studying the mechanism of oMG and gMG pathogenesis in humans and for preclinical therapeutic analysis.


Asunto(s)
Modelos Animales de Enfermedad , Miastenia Gravis Autoinmune Experimental/etiología , Trastornos de la Motilidad Ocular/etiología , Animales , Western Blotting , Complemento C3/inmunología , Ensayo de Inmunoadsorción Enzimática , Escherichia coli/genética , Femenino , Expresión Génica , Antígenos HLA-DQ/genética , Antígeno HLA-DR3/genética , Activación de Linfocitos , Complejo Mayor de Histocompatibilidad/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente , Miastenia Gravis Autoinmune Experimental/genética , Miastenia Gravis Autoinmune Experimental/patología , Trastornos de la Motilidad Ocular/genética , Trastornos de la Motilidad Ocular/patología , Plásmidos , Radioinmunoensayo , Receptores Nicotínicos/inmunología
12.
Neuromuscul Disord ; 16(2): 137-43, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16427283

RESUMEN

Components of the complement cascade and circulating immune complexes play important roles in both experimental autoimmune myasthenia gravis and myasthenia gravis in humans. Thus far, no serological factor has been identified to predict the clinical severity of either myasthenia gravis. Upon immunization with acetylcholine receptor, levels of complement factors C1q, C3 and CIC increase with time in sera from C57BL/6 (B6) mice. Both these and plasma samples from myasthenia gravis patients also contain anti-C1q antibodies. The serum levels of anti-C1q antibodies but not C1q, C3 and CIC are significantly correlated with the clinical severity in the experimental myasthenia mice. However, this correlation is not observed in myasthenia gravis patients.


Asunto(s)
Anticuerpos Antiidiotipos/sangre , Complemento C1q/inmunología , Miastenia Gravis Autoinmune Experimental/inmunología , Miastenia Gravis/inmunología , Adulto , Animales , Complemento C1/análisis , Complemento C1/inmunología , Complemento C1q/análisis , Complemento C3/análisis , Complemento C3/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/análisis , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Debilidad Muscular/fisiopatología , Miastenia Gravis/sangre , Miastenia Gravis/etiología , Miastenia Gravis/fisiopatología , Miastenia Gravis Autoinmune Experimental/sangre , Miastenia Gravis Autoinmune Experimental/etiología , Miastenia Gravis Autoinmune Experimental/fisiopatología , Unión Neuromuscular/química , Unión Neuromuscular/patología , Valor Predictivo de las Pruebas , Índice de Severidad de la Enfermedad
13.
Artículo en Inglés | MEDLINE | ID: mdl-16201276

RESUMEN

To investigate the underlying mechanism of the exacerbation of myasthenia gravis by aminoglycoside antibiotics. C57/BL6 mice were immunized with acetylcholine receptor (AChR), extracted from electric organ of Narcine timilei according to Xu Haopeng's methods, in complete Fruends adjuvant (CFA) to establish experimental autoimmune myasthenia gravis (EAMG). EAMG mice were divided randomly into 5 groups: MG group, NS group and three antibiotics groups. The clinical symptom scores of mice were evaluated on d7 after the last immunization and d14 of antibiotics treatment. Repetitive nerve stimulation (RNS) was performed and the levels of anti-AChR antibody (AChR-Ab) were tested at the same time. The mean clinical symptom grades of gentamycin group (1.312, 2.067), amikacin group (1.111, 1.889) and etimicin group (1.263, 1.632) were significantly higher than those of MG group (1.000, 1.200) (P<0.05). The positive rates of RNS of three antibiotics groups were 69.23%, 58.82% and 63.16% respectively, which were significantly higher than those of MG group and NS group (40.00%, 40.00%, P<0.05). The AChR-Ab level in serum and the expression of AChR on neuromuscular junction (NMJ) of mice in three antibiotics groups were also higher than those of MG group. Our results indicated that aminoglycoside antibiotics could aggravate the symptom of myasthenia gravis. The exacerbation of myasthenia gravis by these antibiotics probably involves competitively restraining the release of acetylcholine from presynaptic membrane, impairing the depolarization of postsynaptic membrane, depressing the irritability of myocyte membrane around the end-plate membrane and consequently leading to the blockade of neuromuscular junction.


Asunto(s)
Aminoglicósidos/efectos adversos , Antibacterianos/efectos adversos , Miastenia Gravis Autoinmune Experimental/tratamiento farmacológico , Amicacina/efectos adversos , Animales , Femenino , Gentamicinas/efectos adversos , Ratones , Ratones Endogámicos C57BL , Miastenia Gravis Autoinmune Experimental/etiología , Miastenia Gravis Autoinmune Experimental/inmunología , Receptores Colinérgicos/inmunología
14.
J Neuroimmunol ; 153(1-2): 16-25, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15265659

RESUMEN

Lymphocyte costimulation via the inducible costimulatory molecule (ICOS) is required for effective humoral immunity development. Following immunization with Torpedo acetylcholine receptor (AChR), ICOS gene knockout (KO) mice were highly resistant to clinical experimental autoimmune myasthenia gravis (EAMG) development, had less serum AChR-specific immunoglobulins (Igs), and exhibited a diminutive germinal center (GC) reaction in secondary lymphoid tissues. Lymphocyte proliferation and both Th1 and Th2 differentiation in response to AChR and the AChR dominant alpha146-162 peptide were inhibited by the ICOS gene deficiency. ICOS-mediated lymphocyte costimulation is thus vital to the induction of T cell-mediated humoral immunity to AChR and the development of clinical EAMG.


Asunto(s)
Formación de Anticuerpos/inmunología , Antígenos de Diferenciación de Linfocitos T/fisiología , Miastenia Gravis Autoinmune Experimental/etiología , Receptores Colinérgicos/inmunología , Linfocitos T/inmunología , Animales , Recuento de Células , División Celular/fisiología , Complemento C3/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática/métodos , Citometría de Flujo/métodos , Centro Germinal/metabolismo , Inmunización/métodos , Epítopos Inmunodominantes/metabolismo , Cambio de Clase de Inmunoglobulina/fisiología , Inmunoglobulina G/metabolismo , Inmunohistoquímica/métodos , Proteína Coestimuladora de Linfocitos T Inducibles , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miastenia Gravis Autoinmune Experimental/inmunología , Unión Neuromuscular/metabolismo , Péptidos/inmunología , Radioinmunoensayo/métodos , Receptores Colinérgicos/sangre , Factores de Tiempo , Torpedo
15.
Immunogenetics ; 56(1): 61-4, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15004728

RESUMEN

The nicotinic acetylcholine receptor (nAChR) is the autoantigen in seropositive myasthenia gravis (MG), a T-cell-dependent B-cell-mediated autoimmune disease. The nAChR is a pentameric transmembrane receptor comprising alpha alpha beta gamma delta chains. During early postnatal development the nAChR gamma chain is replaced by the nAChR epsilon chain. We tested the myasthenogenicity in experimental autoimmune myasthenia gravis (EAMG) of the native nAChR derived from the electric ray Torpedo californica (T-nAChR) in various inbred and MHC -congenic rat strains. Differences in the disease course emerged dependent on the MHC haplotype and non-MHC genes. Interestingly, no tested rat strain was completely resistant to EAMG, but there were strong differences in disease severity mainly depending on the MHC haplotype. In the LEW non-MHC genome, the B-cell response and the severity of EAMG were dependent on the expressed MHC haplotype. This study underscores the influence of genetic factors on disease severity, disease course and on the degree of the emerging antibody responses in EAMG.


Asunto(s)
Genes MHC Clase II , Miastenia Gravis Autoinmune Experimental/genética , Miastenia Gravis Autoinmune Experimental/inmunología , Receptores Nicotínicos/inmunología , Animales , Animales Congénicos , Femenino , Genoma , Haplotipos , Humanos , Inmunización , Miastenia Gravis Autoinmune Experimental/etiología , Ratas , Ratas Endogámicas Lew , Ratas Endogámicas , Torpedo
16.
Ann N Y Acad Sci ; 998: 284-307, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-14592887

RESUMEN

Although the symptoms of myasthenia gravis (MG) and experimental MG (EAMG) are caused by autoantibodies, CD4(+) T cells specific for the target antigen, the nicotinic acetylcholine receptor, and the cytokines they secrete, have an important role in these diseases. CD4(+) T cells have a pathogenic role, by permitting and facilitating the synthesis of high-affinity anti-AChR antibodies. Th1 CD4(+) cells are especially important because they drive the synthesis of anti-AChR complement-fixing IgG subclasses. Binding of those antibodies to the muscle AChR at the neuromuscular junction will trigger the complement-mediated destruction of the postsynaptic membrane. Thus, IL-12, a crucial cytokine for differentiation of Th1 cells, is necessary for development of EAMG. Th2 cells secrete different cytokines, with different effects on the pathogenesis of EAMG. Among them, IL-10, which is a potent growth and differentiation factor for B cells, facilitates the development of EAMG. In contrast, IL-4 appears to be involved in the differentiation of AChR-specific regulatory CD4(+) T cells, which can prevent the development of EAMG and its progression to a self-maintaining, chronic autoimmune disease. Studies on the AChR-specific CD4(+) cells commonly present in the blood of MG patients support a crucial role of CD4(+) T cells in the development of MG. Circumstantial evidence supports a pathogenic role of IL-10 also in human MG. On the other hand, there is no direct or circumstantial evidence yet indicating a role of IL-4 in the modulatory or immunosuppressive circuits in MG.


Asunto(s)
Citocinas/metabolismo , Miastenia Gravis/patología , Linfocitos T/patología , Animales , Anticuerpos/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Citocinas/clasificación , Citocinas/inmunología , Modelos Animales de Enfermedad , Humanos , Inmunización , Ratones , Ratones SCID , Ratones Transgénicos , Músculos/metabolismo , Músculos/fisiopatología , Músculos/trasplante , Miastenia Gravis/inmunología , Miastenia Gravis/metabolismo , Miastenia Gravis Autoinmune Experimental/etiología , Miastenia Gravis Autoinmune Experimental/genética , Miastenia Gravis Autoinmune Experimental/metabolismo , Subunidades de Proteína , Receptores Colinérgicos/inmunología , Linfocitos T/clasificación , Linfocitos T/inmunología , Factores de Tiempo
18.
J Neuroimmunol ; 135(1-2): 56-61, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12576224

RESUMEN

Antibodies (Abs) specifically directed against the muscular acetylcholine receptor (AChR) mediate the pathogenesis of myasthenia gravis (MG). The animal model experimental autoimmune MG (EAMG) can be induced by passive transfer or by active immunization of anti-AChR Abs. We report a new EAMG mouse model that generates human anti-AChR Abs upon immunization with Torpedo AChR (tAChR). Mice transgenic for human mu, gamma1, and kappa germ line genes (HuMAb-Mice) were immunized with tAChR. Serum titers of anti-tAChR Abs were in the nanomolar range, and anti-rodent AChR Abs were in picomolar range. Some HuMAb-Mice had signs of muscle weakness, clearly indicating their susceptibility to EAMG. Human Ab-mouse AChR complexes were found at the neuromuscular junction, while AChR loss was up to 65%. Spleen and lymph nodes were used for producing hybridomas. Of the anti-tAChR monoclonal Ab-producing hybridomas, 2% had cross-reactivity with rodent AChR and none with human AChR. Immunization with a fusion protein, Trx-Halpha1-210, displaying the human main immunogenic region did not result in EAMG or the generation of human anti-human AChR monoclonal Abs. These experiments show that the HuMAb-Mouse represents a suitable model to generate and study the effects of human anti-AChR Abs in vivo.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Genes de Inmunoglobulinas , Miastenia Gravis Autoinmune Experimental/inmunología , Receptores Colinérgicos/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Autoanticuerpos/sangre , Humanos , Hibridomas/inmunología , Inmunización , Ratones , Ratones Transgénicos , Miastenia Gravis Autoinmune Experimental/etiología , Miastenia Gravis Autoinmune Experimental/genética , Torpedo
19.
J Clin Invest ; 110(9): 1269-74, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12417565

RESUMEN

Myasthenia gravis (MG) is an autoimmune neuromuscular transmission disorder characterized by loss of acetylcholine receptors (AChR's) due primarily to the production of anti-AChR autoantibodies. In this study we investigated whether the presence of decay-accelerating factor (DAF or CD55), an intrinsic complement regulator, protects against the development of disease. Experimental autoimmune MG was induced in Daf1(-/-) mice (devoid of neuromuscular DAF protein) and their Daf1(+/+) littermates by injection of rat anti-AChR mAb McAb-3. After twenty-four hours, grip strength assessment revealed that Daf1(-/-) mice exhibited hold times of less than 30 seconds, compared with more than 8 minutes for the Daf1(+/+) controls. The weakness was reversed by edrophonium, consistent with a myasthenic disorder. Immunohistochemistry revealed greatly augmented C3b deposition localized at postsynaptic junctions, and radioimmunoassays showed more profound reductions in AChR levels. Electron microscopy demonstrated markedly greater junctional damage in the Daf1(-/-) mice compared with the Daf1(+/+) littermates. Control studies showed equivalent levels of other cell surface regulators, i.e., Crry and CD59. The results demonstrate that mice that lack DAF are markedly more susceptible to anti-AChR-induced MG, which simulates the primary mechanism in the human disease, and strongly suggest that in disease flares complement inhibitors might have therapeutic value.


Asunto(s)
Antígenos CD55/fisiología , Miastenia Gravis Autoinmune Experimental/etiología , Animales , Antígenos CD59/análisis , Complemento C3b/análisis , Susceptibilidad a Enfermedades , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miastenia Gravis Autoinmune Experimental/prevención & control , Receptores Colinérgicos/inmunología
20.
J Immunol ; 169(3): 1579-86, 2002 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12133987

RESUMEN

Autoantibodies with reactivity against the postjunctional muscle receptor for acetylcholine receptor are able to interfere with contractile function of skeletal muscles and cause the symptoms of myasthenia gravis (MG) in humans, as well as in experimental animal models of MG. In the study described below using a rat model of MG, it was observed that exposure to acetylcholine receptor-reactive Abs also induced increased levels of chemokine (i.e., monocyte chemoattractant protein 1) production by skeletal muscle cells. This was true of both cultured rat myocytes exposed in vitro and rat muscle exposed in vivo following passive Ab transfer. Increased monocyte chemoattractant protein 1 production may explain the increased trafficking of leukocytes through muscle following Ab transfer described in this and other reports. These observations may also be relevant to the induction of disease symptoms in experimental animal models of MG, since numerous reports from this and other laboratories indicate that the cytokine environment provided by leukocytes trafficking through muscle may play a pivotal role in disease progression.


Asunto(s)
Anticuerpos/inmunología , Quimiocina CCL2/biosíntesis , Músculo Esquelético/metabolismo , Miastenia Gravis Autoinmune Experimental/etiología , Receptores Colinérgicos/inmunología , Traslado Adoptivo , Animales , Línea Celular , Movimiento Celular , Femenino , Leucocitos/fisiología , Músculo Esquelético/citología , Músculo Esquelético/inmunología , Ratas , Ratas Endogámicas Lew , Receptores de IgG/fisiología , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...