Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Pharmazie ; 73(9): 526-532, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30223936

RESUMEN

Artesunate is a semi-synthetic derivative of a Chinese herb named Artemisia annua L. that is commonly used as an antimalarial agent in the history of traditional Chinese medicine. Many studies have reported artesunate possesses anti-inflammatory and immunoregulation properties. The present study was conducted to explore whether artesunate was effective in experimental autoimmune myasthenia gravis (EAMG) in Lewis rats. Our data showed that artesunate could improve the clinical symptoms and suppress the development of EAMG. Artesunate exerted its immunomodulatory effects by inhibiting lymphocyte proliferation and the expression of costimulatory molecules CD86, modulating Th1/Th2 cytokine expression levels, and enhancing the level of Treg cells. The final result of administration of artesunate was the decreased synthesis of anti-R97-116 IgG, IgG2a, and IgG2b antibodies. The treatment effect of artesunate was more obvious at dose of 10 mg/kg. These date suggest that artesunate might be a potential drug for the treatment of human myasthenia gravis (MG).


Asunto(s)
Artesunato/farmacología , Factores Inmunológicos/farmacología , Miastenia Gravis Autoinmune Experimental/tratamiento farmacológico , Linfocitos T Reguladores/inmunología , Animales , Antimaláricos/administración & dosificación , Antimaláricos/farmacología , Artesunato/administración & dosificación , Proliferación Celular/efectos de los fármacos , Citocinas/inmunología , Relación Dosis-Respuesta a Droga , Femenino , Factores Inmunológicos/administración & dosificación , Miastenia Gravis Autoinmune Experimental/inmunología , Miastenia Gravis Autoinmune Experimental/fisiopatología , Ratas , Ratas Endogámicas Lew , Células TH1/inmunología , Células Th2/inmunología , Regulación hacia Arriba
2.
Ann N Y Acad Sci ; 1412(1): 146-153, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29068559

RESUMEN

The neuromuscular junction (NMJ) forms the synaptic connection between a motor neuron and a skeletal muscle fiber. In order to achieve a sustained muscle contraction, this synapse has to reliably transmit motor neuronal action potentials onto the muscle fiber. To guarantee successful transmission even during intense activation of the NMJ, a safety factor of neuromuscular transmission exists. In the neuromuscular disorder myasthenia gravis (MG), autoantibodies are directed against acetylcholine receptors or, in the rarer variants, against other postsynaptic NMJ proteins. This causes loss of functional acetylcholine receptors, which compromises the safety factor of neuromuscular transmission, leading to the typical fatigable muscle weakness of MG. With intracellular microelectrode measurement of (miniature) endplate potentials at NMJs in ex vivo nerve-muscle preparations from MG animal models, these functional synaptic defects have been determined in much detail. Here, we describe the electrophysiological events at the normal NMJ and the pathoelectrophysiology at NMJs of animal models for MG.


Asunto(s)
Miastenia Gravis Autoinmune Experimental/fisiopatología , Unión Neuromuscular/fisiopatología , Acetilcolina/fisiología , Animales , Señalización del Calcio/fisiología , Fenómenos Electrofisiológicos , Humanos , Técnicas In Vitro , Proteínas Relacionadas con Receptor de LDL , Ratones , Potenciales Postsinápticos Miniatura/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores Colinérgicos/fisiología , Receptores de LDL/fisiología , Sinapsis/fisiología , Transmisión Sináptica/fisiología
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.
J Neuroimmunol ; 312: 24-30, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28912035

RESUMEN

Myasthenia gravis (MG) is caused by autoantibodies, the majority of which target the muscle acetylcholine receptor (AChR). Plasmapheresis and IgG-immunoadsorption are useful therapy options, but are highly non-specific. Antigen-specific immunoadsorption would remove only the pathogenic autoantibodies, reducing the possibility of side effects while maximizing the benefit. We have extensively characterized such adsorbents, but in vivo studies are missing. We used rats with experimental autoimmune MG to perform antigen-specific immunoadsorptions over three weeks, regularly monitoring symptoms and autoantibody titers. Immunoadsorption was effective, resulting in a marked autoantibody titer decrease while the immunoadsorbed, but not the mock-treated, animals showed a dramatic symptom improvement. Overall, the procedure was found to be efficient, suggesting the subsequent initiation of clinical trials.


Asunto(s)
Autoanticuerpos/sangre , Eliminación de Componentes Sanguíneos/métodos , Inmunoadsorbentes/uso terapéutico , Miastenia Gravis Autoinmune Experimental/inmunología , Miastenia Gravis Autoinmune Experimental/terapia , Animales , Peso Corporal , Modelos Animales de Enfermedad , Electromiografía , Femenino , Miastenia Gravis Autoinmune Experimental/metabolismo , Miastenia Gravis Autoinmune Experimental/fisiopatología , Ratas , Ratas Endogámicas Lew , Receptores Colinérgicos/inmunología , Factores de Tiempo , Resultado del Tratamiento
5.
Acta Neuropathol ; 134(6): 869-888, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28756524

RESUMEN

Myasthenia gravis (MG) is a neuromuscular disease caused in most cases by anti-acetyl-choline receptor (AChR) autoantibodies that impair neuromuscular signal transmission and affect skeletal muscle homeostasis. Myogenesis is carried out by muscle stem cells called satellite cells (SCs). However, myogenesis in MG had never been explored. The aim of this study was to characterise the functional properties of myasthenic SCs as well as their abilities in muscle regeneration. SCs were isolated from muscle biopsies of MG patients and age-matched controls. We first showed that the number of Pax7+ SCs was increased in muscle sections from MG and its experimental autoimmune myasthenia gravis (EAMG) mouse model. Myoblasts isolated from MG muscles proliferate and differentiate more actively than myoblasts from control muscles. MyoD and MyoG were expressed at a higher level in MG myoblasts as well as in MG muscle biopsies compared to controls. We found that treatment of control myoblasts with MG sera or monoclonal anti-AChR antibodies increased the differentiation and MyoG mRNA expression compared to control sera. To investigate the functional ability of SCs from MG muscle to regenerate, we induced muscle regeneration using acute cardiotoxin injury in the EAMG mouse model. We observed a delay in maturation evidenced by a decrease in fibre size and MyoG mRNA expression as well as an increase in fibre number and embryonic myosin heavy-chain mRNA expression. These findings demonstrate for the first time the altered function of SCs from MG compared to control muscles. These alterations could be due to the anti-AChR antibodies via the modulation of myogenic markers resulting in muscle regeneration impairment. In conclusion, the autoimmune attack in MG appears to have unsuspected pathogenic effects on SCs and muscle regeneration, with potential consequences on myogenic signalling pathways, and subsequently on clinical outcome, especially in the case of muscle stress.


Asunto(s)
Músculo Esquelético/fisiopatología , Miastenia Gravis Autoinmune Experimental/fisiopatología , Miastenia Gravis/fisiopatología , Células Satélite del Músculo Esquelético/fisiología , Adulto , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/metabolismo , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Tamaño de la Célula , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones Endogámicos C57BL , Persona de Mediana Edad , Músculo Esquelético/patología , Miastenia Gravis/patología , Miastenia Gravis Autoinmune Experimental/patología , Miogenina/metabolismo , ARN Mensajero/metabolismo , Receptores Colinérgicos/inmunología , Regeneración/inmunología , Células Satélite del Músculo Esquelético/patología , Suero/inmunología , Adulto Joven
6.
Cytokine ; 96: 279-285, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28599246

RESUMEN

An array of cytokines influences the pathogenesis of early onset myasthenia gravis (MG) and its animal model, experimental autoimmune myasthenia gravis (EAMG). Patients with MG, in particular those with more severe weakness, have elevations of the pro-inflammatory cytokine IL-17 in the blood. We assessed the role of IL-17A in autoimmunity by inducing EAMG in mice with knockout of IL-17 and found a reduction of EAMG severity, but not a complete ablation of disease. The IL-17ko mice had no evidence of weakness, low levels of acetylcholine receptor antibodies, and retention of acetylcholine receptor at the neuromuscular junction. Splenic germinal center size was reduced in EAMG IL-17ko mice along with elevations of Foxp3 and BCL-6 gene expression, suggesting a shift away from pro-inflammatory signals. The results emphasize the importance of IL-17 in EAMG development and that IL-17 independent pathways drive the autoimmune reaction.


Asunto(s)
Autoinmunidad/inmunología , Interleucina-17/genética , Interleucina-17/fisiología , Miastenia Gravis Autoinmune Experimental/inmunología , Miastenia Gravis Autoinmune Experimental/fisiopatología , Animales , Autoanticuerpos , Citocinas/inmunología , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/genética , Humanos , Inmunoglobulina G/sangre , Interleucina-17/deficiencia , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas c-bcl-6/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Índice de Severidad de la Enfermedad , Linfocitos T Reguladores
7.
Front Biosci (Landmark Ed) ; 22(7): 1033-1051, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28199191

RESUMEN

Properly sustained impulse transmission at the neuromuscular junction (NMJ) is crucial for successful muscle contraction. To guarantee this, NMJs not only possess a considerable safety factor in transmission but also have the ability to adjust the presynaptic acetylcholine release level to cope with any changes in the postsynaptic neurotransmitter sensitivity. This review will provide overview on the discovery and characterization of this synaptic homeostatic mechanism, especially in the condition of the neuromuscular disorder myasthenia gravis (MG) where the postsynaptic transmitter sensitivity at the NMJ becomes severely reduced due to autoimmune attack of acetylcholine receptors. Because homeostatic signalling and adaptation is presumably maximally active in this condition, NMJs from MG animal models are important study objects. Although candidate post- and presynaptic factors as well as the retrograde signals have been proposed, the homeostatic mechanism at the MG NMJ is still incompletely understood. Further identification and functional characterization of key factors is important because these may form new therapeutic targets in MG.


Asunto(s)
Miastenia Gravis/fisiopatología , Unión Neuromuscular/fisiopatología , Acetilcolina/metabolismo , Animales , Homeostasis , Humanos , Contracción Muscular , Miastenia Gravis/patología , Miastenia Gravis Autoinmune Experimental/patología , Miastenia Gravis Autoinmune Experimental/fisiopatología , Unión Neuromuscular/ultraestructura , Transducción de Señal , Transmisión Sináptica
8.
Am J Pathol ; 186(10): 2559-68, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27658713

RESUMEN

Myasthenia gravis (MG) is an autoimmune disease mediated by autoantibodies that target proteins at the neuromuscular junction, primarily the acetylcholine receptor (AChR) and the muscle-specific kinase. Because downstream of kinase 7 (Dok-7) is essential for the full activation of muscle-specific kinase and consequently for dense clustering of AChRs, we hypothesized that reduced levels of Dok-7 increase the susceptibility to passive transfer MG. To test this hypothesis, Dok-7 expression was reduced by transfecting shRNA-coding plasmids into the tibialis anterior muscle of adult rats by in vivo electroporation. Subclinical MG was subsequently induced with a low dose of anti-AChR monoclonal antibody 35. Neuromuscular transmission was significantly impaired in Dok-7-siRNA-electroporated legs compared with the contralateral control legs, which correlated with a reduction of AChR protein levels at the neuromuscular junction (approximately 25%) in Dok-7-siRNA-electroporated muscles, compared with contralateral control muscles. These results suggest that a reduced expression of Dok-7 may play a role in the susceptibility to passive transfer MG, by rendering AChR clusters less resistant to the autoantibody attack.


Asunto(s)
Autoanticuerpos/inmunología , Proteínas Musculares/genética , Miastenia Gravis Autoinmune Experimental/genética , Animales , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Regulación hacia Abajo , Femenino , Silenciador del Gen , Genes Reporteros , Células HEK293 , Humanos , Proteínas Musculares/metabolismo , Músculo Esquelético/inmunología , Músculo Esquelético/fisiopatología , Miastenia Gravis Autoinmune Experimental/inmunología , Miastenia Gravis Autoinmune Experimental/fisiopatología , Unión Neuromuscular/inmunología , Unión Neuromuscular/fisiopatología , Ratas , Ratas Endogámicas Lew , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Colinérgicos/genética , Receptores Colinérgicos/metabolismo , Transmisión Sináptica
9.
J Neuroimmunol ; 289: 130-8, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26616882

RESUMEN

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.


Asunto(s)
Antígenos CD1d/metabolismo , Subgrupos de Linfocitos B/química , Antígenos CD5/metabolismo , Interleucina-10/metabolismo , Miastenia Gravis Autoinmune Experimental/inmunología , Miastenia Gravis Autoinmune Experimental/patología , Traslado Adoptivo/métodos , Animales , Proliferación Celular/fisiología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Interleucina-10/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/metabolismo , Miastenia Gravis Autoinmune Experimental/inducido químicamente , Miastenia Gravis Autoinmune Experimental/fisiopatología , Fragmentos de Péptidos/inmunología , Receptores Nicotínicos/inmunología
10.
Mol Cell Neurosci ; 68: 284-92, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26311508

RESUMEN

We previously demonstrated that atorvastatin induced immature dendritic cells (DCs) derived from spleen in vitro. Administration of these tolerogenic DCs led to amelioration of experimental autoimmune myasthenia gravis (EAMG). The protective effect was mainly mediated by inhibited cellular immune response, including up-regulated regulatory T cells and shifted Th1/Th17 to Th2 cytokines. The present study employed atorvastatin-modified bone marrow-derived DCs (AT-BMDCs) to explore the effect of tolerogenic DCs on humoral immune response of EAMG and further elucidate the underlying mechanisms. Our data showed that AT-BMDCs reduced the quantity and the relative affinity of pathogenic antibodies, suppressed germinal center response, decreased follicular helper T cells and IL-21, and increased regulatory B cells. These results suggest that AT-BMDCs ameliorate EAMG by regulating humoral immune response, thus providing new insights into therapeutic approaches of myasthenia gravis and other autoimmune diseases.


Asunto(s)
Atorvastatina/uso terapéutico , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Inmunidad Humoral/efectos de los fármacos , Miastenia Gravis Autoinmune Experimental/tratamiento farmacológico , Animales , Atorvastatina/farmacología , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Antígenos de Histocompatibilidad Clase II/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Interleucinas/metabolismo , Miastenia Gravis Autoinmune Experimental/fisiopatología , Ratas , Ratas Endogámicas Lew , Receptores Colinérgicos/inmunología , Factores de Tiempo
11.
PLoS One ; 9(12): e110571, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25438154

RESUMEN

Recent studies demonstrate reduced motor-nerve function during autoimmune muscle-specific tyrosine kinase (MuSK) myasthenia gravis (MG). To further understand the basis of motor-nerve dysfunction during MuSK-MG, we immunized female C57/B6 mice with purified rat MuSK ectodomain. Nerve-muscle preparations were dissected and neuromuscular junctions (NMJs) studied electrophysiologically, morphologically, and biochemically. While all mice produced antibodies to MuSK, only 40% developed respiratory muscle weakness. In vitro study of respiratory nerve-muscle preparations isolated from these affected mice revealed that 78% of NMJs produced endplate currents (EPCs) with significantly reduced quantal content, although potentiation and depression at 50 Hz remained qualitatively normal. EPC and mEPC amplitude variability indicated significantly reduced number of vesicle-release sites (active zones) and reduced probability of vesicle release. The readily releasable vesicle pool size and the frequency of large amplitude mEPCs also declined. The remaining NMJs had intermittent (4%) or complete (18%) failure of neurotransmitter release in response to 50 Hz nerve stimulation, presumably due to blocked action potential entry into the nerve terminal, which may arise from nerve terminal swelling and thinning. Since MuSK-MG-affected muscles do not express the AChR γ subunit, the observed prolongation of EPC decay time was not due to inactivity-induced expression of embryonic acetylcholine receptor, but rather to reduced catalytic activity of acetylcholinesterase. Muscle protein levels of MuSK did not change. These findings provide novel insight into the pathophysiology of autoimmune MuSK-MG.


Asunto(s)
Miastenia Gravis Autoinmune Experimental/patología , Miastenia Gravis Autoinmune Experimental/fisiopatología , Conducción Nerviosa , Proteínas Tirosina Quinasas Receptoras/inmunología , Vesículas Sinápticas/metabolismo , Animales , Femenino , Inmunización Pasiva , Ratones , Placa Motora/patología , Placa Motora/fisiopatología , Neuronas Motoras/patología , Miastenia Gravis Autoinmune Experimental/inmunología , Miastenia Gravis Autoinmune Experimental/metabolismo , Neurotransmisores/metabolismo , Estructura Terciaria de Proteína , Ratas , Proteínas Tirosina Quinasas Receptoras/química , Receptores Colinérgicos/metabolismo , Vacunación
12.
Neuromuscul Disord ; 24(8): 726-35, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24878227

RESUMEN

Myasthenia gravis (MG) is an autoimmune disease caused by circulating antibodies that block acetylcholine receptor (AchR) at the neuromuscular junction. There is the cognitive and memory impairment in patients with MG. However, the molecular mechanisms underlying the alteration of central nervous system in MG remain unknown. In the present study, we found that the level of malondialdehyde (MDA) was increased in the brain of experimental autoimmune myasthenia gravis (EAMG). Furthermore, the expression of thioredoxin-1 (Trx-1) and the activity of cAMP response element-binding protein (CREB) were significantly decreased in frontal lobe and hippocampus of mice with EAMG. We also found that the expression of pro-apoptotic C/EBP homologous protein (CHOP) was increased in the frontal lobe and hippocampus of mice. However, the expressions of glucose regulated protein 78 (GRP78/Bip) was not changed in same areas. Inversely, the expressions of pro-caspase-12, pro-caspase-3 and pro-caspase-9 were decreased. These data indicate that Trx-1 mediated endoplasmic reticulum and mitochondria pathways are involved in brain damage in MG. Trx-1 may be a pivotal target for brain protective treatment in MG.


Asunto(s)
Lóbulo Frontal/metabolismo , Hipocampo/metabolismo , Miastenia Gravis Autoinmune Experimental/metabolismo , Tiorredoxinas/metabolismo , Animales , Apoptosis/fisiología , Caspasa 12/metabolismo , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Chaperón BiP del Retículo Endoplásmico , Femenino , Proteínas de Choque Térmico/metabolismo , Malondialdehído/metabolismo , Ratones Endogámicos C57BL , Mitocondrias/fisiología , Debilidad Muscular/fisiopatología , Miastenia Gravis Autoinmune Experimental/fisiopatología , Factor de Transcripción CHOP/metabolismo
13.
Curr Protoc Immunol ; Chapter 15: Unit 15.8., 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23392639

RESUMEN

Myasthenia gravis (MG) is a T cell-dependent antibody-mediated autoimmune neuromuscular disease. Antibodies to the nicotinic acetylcholine receptor (AChR) destroy the AChR, thus leading to defective neuromuscular transmission of electrical impulse and to muscle weakness. This unit is a practical guide to the induction and evaluation of experimental autoimmune myasthenia gravis (EAMG) in the mouse, the animal model for MG. Protocols are provided for the extraction and purification of AChR from the electric organs of Torpedo californica, or the electric ray. The purified receptor is used as an immunogen to induce autoimmunity to AChR, thus causing EAMG. The defect in neuromuscular transmission can also be measured quantitatively by electromyography. In addition, EAMG is frequently characterized by the presence of serum antibodies to AChR, which are measured by radioimmunoassay and by a marked antibody-mediated reduction in the number of muscle AChRs. AChR extracted from mouse muscle is used in measuring serum antibody levels and for quantifying muscle AChR content. Another hallmark of the disease is complement and IgG deposits located at the neuromuscular junction, which can be visualized by immunofluorescence techniques.


Asunto(s)
Proteínas de Peces/inmunología , Músculo Esquelético/inmunología , Miastenia Gravis Autoinmune Experimental/inmunología , Unión Neuromuscular/efectos de los fármacos , Receptores Nicotínicos/inmunología , Transmisión Sináptica/efectos de los fármacos , Animales , Autoanticuerpos/biosíntesis , Autoanticuerpos/inmunología , Proteínas del Sistema Complemento/inmunología , Electromiografía , Femenino , Proteínas de Peces/administración & dosificación , Proteínas de Peces/aislamiento & purificación , Inmunoglobulina G/inmunología , Masculino , Ratones , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Miastenia Gravis Autoinmune Experimental/metabolismo , Miastenia Gravis Autoinmune Experimental/fisiopatología , Unión Neuromuscular/fisiopatología , Radioinmunoensayo , Receptores Nicotínicos/administración & dosificación , Receptores Nicotínicos/aislamiento & purificación , Torpedo/fisiología
14.
J Physiol ; 591(10): 2747-62, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23440963

RESUMEN

In myasthenia gravis, the neuromuscular junction is impaired by the antibody-mediated loss of postsynaptic acetylcholine receptors (AChRs). Muscle weakness can be improved upon treatment with pyridostigmine, a cholinesterase inhibitor, or with 3,4-diaminopyridine, which increases the release of ACh quanta. The clinical efficacy of pyridostigmine is in doubt for certain forms of myasthenia. Here we formally examined the effects of these compounds in the antibody-induced mouse model of anti-muscle-specific kinase (MuSK) myasthenia gravis. Mice received 14 daily injections of IgG from patients with anti-MuSK myasthenia gravis. This caused reductions in postsynaptic AChR densities and in endplate potential amplitudes. Systemic delivery of pyridostigmine at therapeutically relevant levels from days 7 to 14 exacerbated the anti-MuSK-induced structural alterations and functional impairment at motor endplates in the diaphragm muscle. No such effect of pyridostigmine was found in mice receiving control human IgG. Mice receiving smaller amounts of MuSK autoantibodies did not display overt weakness, but 9 days of pyridostigmine treatment precipitated generalised muscle weakness. In contrast, one week of treatment with 3,4-diaminopyridine enhanced neuromuscular transmission in the diaphragm muscle. Both pyridostigmine and 3,4-diaminopyridine increase ACh in the synaptic cleft yet only pyridostigmine potentiated the anti-MuSK-induced decline in endplate ACh receptor density. These results thus suggest that ongoing pyridostigmine treatment potentiates anti-MuSK-induced AChR loss by prolonging the activity of ACh in the synaptic cleft.


Asunto(s)
Debilidad Muscular/fisiopatología , Miastenia Gravis Autoinmune Experimental/fisiopatología , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores Colinérgicos/fisiología , 4-Aminopiridina/análogos & derivados , 4-Aminopiridina/farmacología , Amifampridina , Animales , Autoanticuerpos/farmacología , Inhibidores de la Colinesterasa/farmacología , Potenciales Evocados , Femenino , Humanos , Inmunoglobulina G/farmacología , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/fisiología , Unión Neuromuscular/efectos de los fármacos , Unión Neuromuscular/fisiología , Bromuro de Piridostigmina/farmacología
15.
Ann N Y Acad Sci ; 1274: 127-32, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23252907

RESUMEN

Complement plays an important role in the pathophysiology of experimental autoimmune myasthenia gravis (EAMG). The deposition of IgG at the neuromuscular junction, followed by the activation and observance of C3 at the site, and finally the insertion of the membrane attack complex results in the destruction of the plasma membrane at the neuromuscular junction. Animal models of complement-deficient components show the importance of the mediated lysis in EAMG. These events have regulators that allow for the limitation in the cascade and the ability of the cell to inhibit complement at many places along the pathway. The complement regulatory proteins have many roles in reducing the activation of the complement cascade and the inflammatory pathways. Mice deficient in complement regulatory proteins, decay accelerating factor, and CD59 demonstrate a significant increase in the destruction at the neuromuscular junction. Inhibition of complement-mediated lysis is an attractive therapeutic in MG.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Miastenia Gravis Autoinmune Experimental/inmunología , Animales , Antígenos CD59/inmunología , Antígenos CD59/metabolismo , Complemento C9/inmunología , Complemento C9/metabolismo , Proteínas del Sistema Complemento/deficiencia , Ratones , Miastenia Gravis Autoinmune Experimental/metabolismo , Miastenia Gravis Autoinmune Experimental/fisiopatología , Unión Neuromuscular/inmunología , Unión Neuromuscular/metabolismo
16.
PLoS One ; 7(8): e44148, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22952904

RESUMEN

Myasthenia Gravis (MG) patients suffer from chronic fatigue of skeletal muscles, even after initiation of proper immunosuppressive medication. Since the localization of neuronal nitric oxide synthase (nNOS) at the muscle membrane is important for sustained muscle contraction, we here study the localization of nNOS in muscles from mice with acetylcholine receptor antibody seropositive (AChR+) experimental autoimmune MG (EAMG). EAMG was induced in 8 week-old male mice by immunization with AChRs purified from torpedo californica. Sham-injected wild type mice and mdx mice, a model for Duchenne muscular dystrophy, were used for comparison. At EAMG disease grade 3 (severe myasthenic weakness), the triceps, sternomastoid and masseter muscles were collected for analysis. Unlike in mdx muscles, total nNOS expression as well as the presence of its binding partner syntrophin α-1, were not altered in EAMG. Immunohistological and biochemical analysis showed that nNOS was lost from the muscle membrane and accumulated in the cytosol, which is likely the consequence of blocked neuromuscular transmission. Atrophy of all examined EAMG muscles were supported by up-regulated transcript levels of the atrogenes atrogin-1 and MuRF1, as well as MuRF1 protein, in combination with reduced muscle fiber diameters. We propose that loss of sarcolemmal nNOS provides an additional mechanism for the chronic muscle fatigue and secondary muscle atrophy in EAMG and MG.


Asunto(s)
Fatiga Muscular , Atrofia Muscular/complicaciones , Atrofia Muscular/patología , Miastenia Gravis Autoinmune Experimental/complicaciones , Miastenia Gravis Autoinmune Experimental/patología , Óxido Nítrico Sintasa de Tipo I/deficiencia , Sarcolema/enzimología , Animales , Autoanticuerpos/inmunología , Citosol/enzimología , Modelos Animales de Enfermedad , Inmunización , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Desnervación Muscular , Músculo Esquelético/enzimología , Músculo Esquelético/inmunología , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Atrofia Muscular/inmunología , Atrofia Muscular/fisiopatología , Miastenia Gravis Autoinmune Experimental/inmunología , Miastenia Gravis Autoinmune Experimental/fisiopatología , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico Sintasa de Tipo I/metabolismo , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Colinérgicos/metabolismo , Sarcolema/patología , Pérdida de Peso
17.
Exp Neurol ; 234(2): 506-12, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22326541

RESUMEN

Antibodies directed against the post-synaptic neuromuscular junction protein, muscle specific kinase (MuSK) are found in a small proportion of generalized myasthenia gravis (MuSK-MG) patients. MuSK is a receptor tyrosine kinase which is essential for clustering of the acetylcholine receptors (AChRs) at the neuromuscular junction, but the mechanisms by which MuSK antibodies (MuSK-Abs) affect neuromuscular transmission are not clear. Experimental models of MuSK-MG have been described but there have been no detailed electrophysiological studies and no comparisons between the MuSK-MG and the typical form with AChR-Abs (AChR-MG). Here we studied the electrophysiology of neuromuscular transmission after immunization against MuSK compared with immunization against AChR, and also after passive transfer of IgG from MuSK-MG or AChR-MG patients. Overt clinical weakness was observed in 6/10 MuSK-immunized and 3/9 AChR-immunized mice but not in those injected with patients' IgG. Miniature endplate potentials (MEPPS) were reduced in all weak mice consistent with the reduction in postsynaptic AChRs that was found. However, whereas there was an increase in the quantal release of acetylcholine (ACh) in the weak AChR-immunized mice, no such increase was found in the weak MuSK-immunized mice. Similar trends were found after the passive transfer of purified IgG antibodies from MuSK-MG or AChR-MG patients. Preliminary results showed that MuSK expression was considerably higher at the neuromuscular junctions of the masseter (facial) than in the gastrocnemius (leg) with no reduction in MuSK immunostaining at the neuromuscular junctions. Overall, these results suggest that MuSK antibodies act in at least two ways. Firstly by indirectly affecting MuSK's ability to maintain the high density of AChRs and secondly by interfering with a compensatory presynaptic mechanism that regulates quantal release and helps to preserve neuromuscular function. These results raise questions about how MuSK is involved in retrograde signaling, and the combination of post-synaptic defects with lack of presynaptic compensation may begin to explain the more severe disease in MuSK-MG patients.


Asunto(s)
Autoanticuerpos/inmunología , Debilidad Muscular/fisiopatología , Miastenia Gravis Autoinmune Experimental/fisiopatología , Unión Neuromuscular/fisiopatología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Colinérgicos/metabolismo , Sinapsis/metabolismo , Animales , Modelos Animales de Enfermedad , Inmunización Pasiva , Ratones , Actividad Motora/inmunología , Debilidad Muscular/inmunología , Miastenia Gravis Autoinmune Experimental/inmunología , Unión Neuromuscular/inmunología , Proteínas Tirosina Quinasas Receptoras/inmunología , Receptores Colinérgicos/inmunología , Sinapsis/inmunología , Vacunación
18.
Arch Immunol Ther Exp (Warsz) ; 60(1): 19-30, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22159475

RESUMEN

Myasthenia gravis (MG) is an autoimmune disease caused by an immunological response against the acetylcholine receptor (AChR) at the neuromuscular junction. Anti-AChR antibodies induce degradation of the receptor, activation of complement cascade and destruction of the post-synaptic membrane, resulting in a functional reduction of AChR availability. The pathophysiological role of autoantibodies (auto-Abs) and T helper lymphocytes has been studied in the experimental autoimmune MG (EAMG) models. EAMG models have been employed to investigate the factors involved in the development of MG and to suggest new therapies aimed to preventing or modulating the ongoing disease. EAMG can be induced in susceptible mouse and rat strains, which develop clinical symptoms such as muscular weakness and fatigability, mimicking the human disease. Two major types of EAMG can be induced, passive and active EAMG. Passive transfer MG models, involving the injection of auto-Abs, are helpful for studying the role of complement molecules and their regulatory proteins, which can prevent neuromuscular junction degradation. Active models, induced by immunization, are employed for the analysis of antigen-specific immune responses and their modulation in order to improve disease progression. In this review, we will concentrate on the main pathogenic mechanisms of MG, focusing on recent findings on EAMG experimental models.


Asunto(s)
Autoanticuerpos/inmunología , Modelos Animales de Enfermedad , Miastenia Gravis Autoinmune Experimental/inmunología , Miastenia Gravis Autoinmune Experimental/fisiopatología , Receptores Colinérgicos/inmunología , Animales , Proteínas del Sistema Complemento/inmunología , Humanos , Tolerancia Inmunológica/inmunología , Inmunoterapia/métodos , Miastenia Gravis/inmunología , Miastenia Gravis/patología , Miastenia Gravis/fisiopatología , Miastenia Gravis/terapia , Miastenia Gravis Autoinmune Experimental/patología , Miastenia Gravis Autoinmune Experimental/terapia , Unión Neuromuscular/inmunología , Unión Neuromuscular/patología
19.
Ann N Y Acad Sci ; 1233: 26-33, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21950972

RESUMEN

Acetylcholine receptors (AchRs) and Na(+) channels (NaChs) are concentrated on neuromuscular junction (NMJ) postsynaptic folds; both are depleted in myasthenia gravis (MG), reducing the safety factor (SF) for neuromuscular transmission, especially in extraocular muscles (EOM). Studies of human myasthenic nerve-muscle preparations indicate that loss of endplate AChRs accounts for 59%, and NaChs for 40%, of SF reduction. Rodent models of MG indicate that NaChs and AChRs losses are due to complement-mediated destruction of postsynaptic folding. Saccades in MG show stereotyped, conjugate initial components, similar to normal but different from early disconjugacy with ocular nerve palsies. Loss of AChRs, NaChs, and postsynaptic folding all contribute to SF reduction in MG. EOM seem more susceptible to MG because of poor postsynaptic folding, lower baseline SF, and lower levels of intrinsic complement inhibitors. Initial conjugacy of saccades in MG reflects selective sparing of neuromuscular transmission of fast, pale global fibers, which have better developed postsynaptic folding.


Asunto(s)
Miastenia Gravis/fisiopatología , Músculos Oculomotores/inervación , Músculos Oculomotores/fisiopatología , Transmisión Sináptica/fisiología , Adulto , Anciano , Animales , Estudios de Casos y Controles , Movimientos Oculares/fisiología , Humanos , Masculino , Ratones , Microscopía Electrónica de Transmisión , Persona de Mediana Edad , Placa Motora/fisiopatología , Miastenia Gravis Autoinmune Experimental/patología , Miastenia Gravis Autoinmune Experimental/fisiopatología , Unión Neuromuscular/fisiopatología , Unión Neuromuscular/ultraestructura , Músculos Oculomotores/patología , Nervio Oculomotor/patología , Nervio Oculomotor/fisiopatología , Ratas , Receptores Colinérgicos/fisiología , Movimientos Sacádicos/fisiología , Canales de Sodio/fisiología
20.
J Neuroimmunol ; 234(1-2): 165-7, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21481948

RESUMEN

Autoimmune myasthenia gravis (MG) is characterized by T cell and antibody responses to muscle nicotinic acetylcholine receptor (AChR). It is well known that MG as other autoimmune diseases is more prevalent in women than men and estrogen administration enhances experimental autoimmune MG (EAMG) severity. To determine whether estrogen influences EAMG pathogenesis through estrogen receptor alpha (ERα) activation, ERα knockout (KO) and wild-type (WT) C57BL/6 mice were immunized with AChR. ERα KO mice were equally susceptible to EAMG as WT mice and exhibited comparable antibody and immunopathological responses to AChR, suggesting a lack of involvement of ERα in EAMG pathogenesis.


Asunto(s)
Receptor alfa de Estrógeno/deficiencia , Miastenia Gravis Autoinmune Experimental/genética , Miastenia Gravis Autoinmune Experimental/inmunología , Miastenia Gravis Autoinmune Experimental/patología , Animales , Anticuerpos/sangre , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática/métodos , Predisposición Genética a la Enfermedad/genética , Fuerza de la Mano/fisiología , Inmunización/efectos adversos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miastenia Gravis Autoinmune Experimental/fisiopatología , Receptores Colinérgicos/inmunología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA