Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Neuroreport ; 32(9): 803-807, 2021 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-33994526

RESUMEN

Myasthenia gravis (MG) is an autoimmune disease with autoantibodies against the mainly nicotinic acetylcholine receptor (AChR). High mobility group box1 (HMGB1) acts as a danger signal and drives the pathogenesis of autoimmune-mediated diseases. However, the role of HMGB1 in the pathogenesis of MG is not fully understood. Therefore, in this study, we analyzed serum levels of HMGB1 and immunohistochemical HMGB1 staining of muscle tissues in the passive transfer MG model to investigate the role of HMGB1 in MG. As a result, serum HMGB1 levels tended to be higher and the quantitative score of muscle pathology showed greater HMGB1 deposition (P = 0.02) along with sparser AChR staining and more severe inflammation in the passive transfer MG rats (n = 6) than those in control rats (n = 6). These findings indicate that HMGB1 is an important mediator and biomarker for inflammation in the pathogenesis of MG and can be a therapeutic target in MG.


Asunto(s)
Proteína HMGB1/metabolismo , Músculo Esquelético/metabolismo , Miastenia Gravis Autoinmune Experimental/metabolismo , Animales , Femenino , Proteína HMGB1/sangre , Miastenia Gravis Autoinmune Experimental/sangre , Ratas
2.
Immunol Invest ; 50(6): 671-684, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32597289

RESUMEN

Antigen-specific immune responses are crucially involved in both multiple sclerosis (MS) and myasthenia gravis (MG). Teriflunomide is an immunomodulatory agent approved for treatment of MS through inhibition of lymphocyte proliferation. MG associated with muscle-specific tyrosine kinase (MuSK) antibodies often manifests with a severe disease course, prompting development of effective treatment methods. To evaluate whether teriflunomide treatment may ameliorate MuSK-autoimmunity, experimental autoimmune MG (EAMG) was induced by immunizing C57BL/6 (B6) mice three times with MuSK in complete Freund's adjuvant (CFA) (n = 17). MuSK-immunized mice were treated daily with teriflunomide (n = 8) or PBS (n = 9) starting from the third immunization (week 8) to termination (week 14). Clinical severity of EAMG was monitored. Immunological alterations were evaluated by measurement of anti-MuSK IgG, neuromuscular junction deposits, and flow cytometric analysis of lymph node cells. In MS patients under teriflunomide treatment, the peripheral blood B cell subset profile was analyzed. B6 mice treated with teriflunomide displayed relatively preserved body weight, lower EAMG prevalence, reduced average clinical grades, higher inverted screen scores, diminished anti-MuSK antibody and NMJ deposit levels. Amelioration of EAMG findings was associated with reduced memory B cell ratios in the lymph nodes. Similarly, MS patients under teriflunomide treatment showed reduced memory B cell, plasma cell, and plasmablast ratios. Teriflunomide treatment has effectively ameliorated MuSK-autoimmunity and thus may putatively be used in long-term management of MuSK-MG as an auxiliary treatment method. Teriflunomide appears to exert beneficial effects through inhibition of effector B cells.


Asunto(s)
Subgrupos de Linfocitos B/efectos de los fármacos , Crotonatos/administración & dosificación , Hidroxibutiratos/administración & dosificación , Esclerosis Múltiple/tratamiento farmacológico , Miastenia Gravis Autoinmune Experimental/tratamiento farmacológico , Nitrilos/administración & dosificación , Proteínas Tirosina Quinasas Receptoras/inmunología , Receptores Colinérgicos/inmunología , Toluidinas/administración & dosificación , Adulto , Animales , Subgrupos de Linfocitos B/inmunología , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Esclerosis Múltiple/sangre , Esclerosis Múltiple/diagnóstico , Esclerosis Múltiple/inmunología , Miastenia Gravis Autoinmune Experimental/sangre , Miastenia Gravis Autoinmune Experimental/diagnóstico , Miastenia Gravis Autoinmune Experimental/inmunología , Proteínas Tirosina Quinasas Receptoras/administración & dosificación , Receptores Colinérgicos/administración & dosificación , Resultado del Tratamiento
3.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 32(11): 1471-1474, 2016 Nov.
Artículo en Chino | MEDLINE | ID: mdl-27774937

RESUMEN

Objective To investigate the effects of tetramethylpyrazine (TMP) on experimental autoimmune myasthenia gravis (EAMG) in rats and explore the possible immune regulation mechanism. Methods Lewis rats were randomly divided into 4 groups: control group, EAMG group, TMP low-dose group (TMP-L, 10 mg/kg) and TMP high-dose group (TMP-H, 20 mg/kg). Except the control group, the other 3 groups were subjected to EAMG modeling. The body mass was determined and the symptoms of muscular weakness in rats were scored by Lennon EAMG criteria. The expressions of acetylcholine receptor (AChR), IgG and C3 in neuromuscular junctions were examined by immunofluorescence. The levels of R97-116 antibody (IgG1, IgG2a, IgG2b), tumor necrosis factor α (TNF-α), interferon γ (IFN-γ) and interleukin 10 (IL-10) were detected by ELISA. Results TMP decreased the scores in the evaluation of the symptoms of muscle weakness in EAMG rats. Moreover, TMP administration significantly inhibited AChR expression and increased IgG and C3 expression in neuromuscular junctions of EAMG rats. In addition, after TMP treament, the levels of IgG1, IgG2a and TNF-α were remarkably reduced, while IL-10 level was elevated. However, there was no significant change in serum levels of IgG2b and IFN-γ. Conclusion TMP can exert inhibitory effects on Th1, Th2 and B cells and modulate the levels of IgG1, IgG2a, TNF-α and IL-10. All these effects contribute to relieve the symptoms of muscle weakness in EAMG rats.


Asunto(s)
Citocinas/sangre , Miastenia Gravis Autoinmune Experimental/sangre , Miastenia Gravis Autoinmune Experimental/tratamiento farmacológico , Pirazinas/uso terapéutico , Animales , Ensayo de Inmunoadsorción Enzimática , Femenino , Inmunoglobulina G/sangre , Interferón gamma/sangre , Interleucina-10/sangre , Ratas , Ratas Endogámicas Lew , Factor de Necrosis Tumoral alfa/sangre
4.
J Neuroinflammation ; 13(1): 121, 2016 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-27220674

RESUMEN

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease that involves the selective loss of the upper and lower motor neurons (MNs). Neuroinflammation has been implicated in the pathogenesis of the sporadic form of the disease. We earlier developed immune-mediated animal models of ALS and demonstrated humoral and cellular immune reactions in the nervous system and in the sera of patients and animals. The accumulation of immunoglobulin G (IgG), an elevated intracellular level of calcium, ultrastructural alterations in the MNs, and activation of the microglia were noted in the spinal cord of ALS patients. Similar alterations developed in mice inoculated intraperitoneally with IgG from ALS patients or from an immune-mediated goat model. METHODS: We have now examined whether the intraperitoneal injection of mice with IgG from sporadic ALS patients or from immunized goats with the homogenate of the anterior horn of the bovine spinal cord is associated with changes in the pro-inflammatory (TNF-α and IL-6) and anti-inflammatory (IL-10) cytokines in the spinal cord and serum of the mice. The levels of cytokines were measured by ELISA. RESULTS: Intraperitoneally administered IgG from the ALS patients induced subclinical signs of MN disease, while the injection of IgG from immunized goats resulted in a severe respiratory dysfunction and limb paralysis 24 h after the injections. Significantly increased levels of TNF-α and IL-10 were detected in the spinal cord of the mice injected with the human ALS IgG. The level of IL-6 increased primarily in the serum. The IgG from the immunized goats induced highly significant increases in the levels of all three cytokines in the serum and the spinal cord of mice. CONCLUSIONS: Our earlier experiments had proved that when ALS IgG or IgG from immune-mediated animal models was inoculated into mice, it was taken up in the MNs and had the ability to initiate damage in them. The pathological process was paralleled by microglia recruitment and activation in the spinal cord. The present experiment revealed that these forms of IgG cause significant increases in certain cytokine levels locally in the spinal cord and in the serum of the inoculated mice. These results suggest that IgG directed to the MNs may be an initial element in the damage to the MNs both in human ALS and in its immune-mediated animal models.


Asunto(s)
Esclerosis Amiotrófica Lateral/sangre , Citocinas/metabolismo , Inmunoglobulina G/administración & dosificación , Médula Espinal/metabolismo , Esclerosis Amiotrófica Lateral/inmunología , Análisis de Varianza , Animales , Calcio/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Cabras , Humanos , Inyecciones Intraperitoneales , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Actividad Motora/efectos de los fármacos , Neuronas Motoras/patología , Neuronas Motoras/ultraestructura , Miastenia Gravis Autoinmune Experimental/sangre , Miastenia Gravis Autoinmune Experimental/inmunología , Miastenia Gravis Autoinmune Experimental/patología , Factor de Necrosis Tumoral alfa/metabolismo
5.
Genet Mol Res ; 14(3): 7782-92, 2015 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-26214459

RESUMEN

It is expensive to induce experimental autoimmune myasthenia gravis (EAMG) by active immunity, and difficult to obtain natural acetylcholine receptor (AChR). We sought a new method of inducing EAMG by immunizing rats with artificially synthesized AChR. The AChR mRNA in TE671 cells was extracted and reverse transcribed. The inclusion body was purified and protein concentration was determined, and the EAMG animal model was used for induction. The serum was extracted from rat blood. The antibody titer was determined using enzyme-linked immunosorbant assay (ELISA). The concentration of decay accelerating factor (DAF) in the rat serum was determined by ELISA, and the metabolism of serum rDAF was determined by western blot. We evaluated the inhibition of rDAF by determining the 50% complement hemolysis unit in the rat serum. The extracellular domain (ECD) nucleotide sequence clone produced by polymerase chain reaction was completely consistent with that in the human gene bank; it was induced by isopropyl ß-D-1-thiogalactopyranoside to express the protein after insertion into vector pET16b. Sodium dodecyl sulfate polyacrylamide gel electrophoresis demonstrated that the inclusion body protein was the exact target. The ECD protein was able to bind with mAb35 after dialysis and renaturation, which demonstrated protein activity. The soluble ECD protein was used to immunize rats and obtain the EAMG models. The inhibitory effect of the complement was unsatisfactory owing to high decay rate after rDAF injection into the EAMG models. It is easy to induce the EAMG model by obtaining the AChRTEα1 subunit ECD protein using the substitution method.


Asunto(s)
Antígenos CD55/uso terapéutico , Miastenia Gravis Autoinmune Experimental/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Animales , Antígenos CD55/administración & dosificación , Antígenos CD55/sangre , Modelos Animales de Enfermedad , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inyecciones Intravenosas , Miastenia Gravis Autoinmune Experimental/sangre , Miastenia Gravis Autoinmune Experimental/patología , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacología , Renaturación de Proteína/efectos de los fármacos , Estructura Terciaria de Proteína , Ratas Endogámicas Lew , Proteínas Recombinantes/metabolismo , Solubilidad
6.
J Autoimmun ; 52: 36-43, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24423642

RESUMEN

Tissue plasminogen activator (tPA), a component of the PA/plasmin system, is elevated in inflammatory areas and plays a role in inflammatory neurological disorders. In the present study we explored the involvement of tPA and the potential immunomodulatory activity of tPA in experimental autoimmune myasthenia gravis (EAMG). Mice deficient in tPA (tPA(-/-)) present with a markedly more severe disease than wild type EAMG mice. In an attempt to treat EAMG with an 18aa peptide derived from the PA system inhibitor (PAI-1), designed to tether out the endogenous inhibitor, a significant suppression of disease severity was demonstrated. The more severe disease in tPA(-/-) mice was accompanied by a higher level of anti-AChR antibodies and increased expression of B-cell markers. In view of the essential role of B-cell activating factor (BAFF) in B-cell maturation, the expression of BAFF family components was tested. An increase in BAFF and BAFF receptor was observed in EAMG tPA(-/-) mice, whereas BCMA expression was reduced, consistent with the increased level of pathogenic antibodies and the more severe disease. Given the importance of T regulatory cells (Tregs) in EAMG, they were evaluated and their number was reduced in tPA(-/-) mice, in which EAMG was aggravated, whereas following PAI-1dp treatment, Tregs were replenished and the disease was ameliorated. The results show the involvement of tPA in EAMG, implying a protective role for tPA in EAMG pathogenesis. The amelioration of EAMG by PAI-1dp treatment suggests that the PA system may be considered a potential site for therapeutic intervention in neuroimmune diseases.


Asunto(s)
Miastenia Gravis Autoinmune Experimental/sangre , Linfocitos T Reguladores/inmunología , Activador de Tejido Plasminógeno/metabolismo , Animales , Autoanticuerpos/sangre , Factor Activador de Células B/genética , Factor Activador de Células B/metabolismo , Progresión de la Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miastenia Gravis Autoinmune Experimental/tratamiento farmacológico , Miastenia Gravis Autoinmune Experimental/inmunología , Fragmentos de Péptidos/administración & dosificación , Inhibidor 1 de Activador Plasminogénico/administración & dosificación , Receptores Colinérgicos/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Activador de Tejido Plasminógeno/genética , Regulación hacia Arriba
7.
Neurosci Res ; 77(1-2): 102-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23933211

RESUMEN

Myasthenia gravis (MG) patients with antibodies against muscle specific tyrosine kinase (MuSK+) typically present focal fatigue and atrophy of the facial and bulbar muscles, including the masseter muscle, whereas leg muscles often are clinically spared. This study addresses the regulation of the mTOR signaling pathway in the masseter muscle versus the leg muscle tibialis anterior (TA). We analyzed muscle morphology, protein levels of mTOR components as well as atrogenes and mitochondrial markers in these muscles of healthy control mice and mice with different clinical severity grades of MuSK+ experimental autoimmune MG (EAMG). Protein levels of mTOR components were reduced in the atrophic masseter muscle of MuSK+ EAMG mice, whereas enhanced accumulation of mTOR components was observed in the TA muscles. Two other muscles: omohyoid and soleus showed intermediate spectra. In conclusion, the anabolic mTOR signaling pathway is differentially regulated even in muscles with the same activity pattern in the same neuromuscular disease. This could in part explain the clinical phenotype in MuSK+ EAMG as well as in muscular dystrophies.


Asunto(s)
Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Miastenia Gravis Autoinmune Experimental/inmunología , Miastenia Gravis Autoinmune Experimental/metabolismo , Proteínas Tirosina Quinasas Receptoras/inmunología , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Animales , Femenino , Músculo Masetero/metabolismo , Músculo Masetero/patología , Ratones , Ratones Endogámicos C57BL , Proteínas Musculares/metabolismo , Miastenia Gravis Autoinmune Experimental/sangre , Proteínas Tirosina Quinasas Receptoras/sangre , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Ligasas SKP Cullina F-box/metabolismo , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas/metabolismo
8.
J Neuroimmunol ; 251(1-2): 25-32, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22769060

RESUMEN

Current immunotherapy of myasthenia gravis (MG) is often effective, but entails risks of infection and neoplasia. The "Guided Missile" strategy described here is designed to target and eliminate the individual's unique AChR-specific T cell repertoire, without otherwise interfering with the immune system. We genetically engineered dendritic cells to present AChR epitopes and simultaneously express Fas ligand in an ongoing EAMG model. In both in vitro and in vivo experiments, these engineered cells specifically killed AChR-responsive T cells without otherwise damaging the immune system. AChR antibodies were markedly reduced in the treated mice. Translation of this method to treat human MG is possible.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/trasplante , Inmunoterapia/métodos , Miastenia Gravis Autoinmune Experimental/terapia , Animales , Anticuerpos/sangre , Células Cultivadas , Modelos Animales de Enfermedad , Epítopos/genética , Epítopos/inmunología , Proteína Ligando Fas/genética , Proteína Ligando Fas/inmunología , Femenino , Ingeniería Genética , Ratones , Ratones Endogámicos C57BL , Miastenia Gravis Autoinmune Experimental/sangre , Miastenia Gravis Autoinmune Experimental/inmunología , Receptores Colinérgicos/genética , Receptores Colinérgicos/inmunología , Linfocitos T/inmunología
9.
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
10.
Cell Immunol ; 271(2): 292-8, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21861992

RESUMEN

In myasthenia gravis (MG), the neuromuscular transmission is impaired by antibodies (Abs) specific for muscle acetylcholine receptor (AChR). Anti-AChR Abs can be detected in the serum of MG patients, although their levels do not correlate with disease severity. In this study, we developed a flow cytometric assay for the detection of peripheral blood AChR-specific B cells to characterize B cell phenotypes associated with experimental autoimmune myasthenia gravis (EAMG). Alexa-conjugated AChR was used as a probe for AChR-specific B cells (B220+Ig+). Mice with EAMG had significantly elevated frequencies of AChR-specific IgG2+ and IgM+ B cells. While the frequencies of IgG2+ B cells and plasma anti-AChR IgG2 levels significantly correlated with the clinical grades of EAMG, the frequencies of IgM+ B cells and plasma anti-AChR IgM levels did not. These results indicate that the frequency of AChR-specific and IgG1+ (mouse IgG2 equivalent) peripheral blood B cells and anti-AChR IgG1 levels could be potential biomarkers for MG disease severity.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/metabolismo , Miastenia Gravis Autoinmune Experimental/sangre , Miastenia Gravis Autoinmune Experimental/inmunología , Receptores Colinérgicos/sangre , Animales , Autoanticuerpos/sangre , AMP Cíclico/análogos & derivados , Citometría de Flujo , Inmunización , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Ratones , Ratones Endogámicos C57BL , Torpedo/inmunología
11.
J Biomed Mater Res A ; 98(4): 589-95, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21721115

RESUMEN

The role of immunoadsorption therapy is well established in the management of myasthenia gravis (MG), an autoimmune disorder characterized by muscle weakness and caused by circulating IgG antibodies with specificity against the acetylcholine receptor. Conventional immunoadsorbents that employ recombinant protein A as immobilized ligand suffer from the drawbacks of high cost and low stability. The objective of this work is to assess the safety and efficacy of a synthetic adsorbent for treating MG. Adsorption columns were prepared from a Sepharose-based adsorbent coupled to 4-mercaptoethylpyridine (MEP), which acted as immobilized ligands. Animal model of experimental autoimmune MG (EAMG) using Lewis rats was developed and treated by whole blood perfusion. The results showed that the treatments provided a significant amelioration of clinical weakness for EAMG rats, with clinic score decreasing from 2.08 ± 0.38 to 1.25 ± 0.27. After a treatment session of about 1.5 h, blood cell counts were not significantly changed. Serum levels of total IgG and acetylcholine receptor antibody were reduced by 37.1 ± 6.5% and 35.6 ± 8.6%, respectively. In addition, reduction in complement components C3 (47.1 ± 6.7%), C4 (34.3 ± 3.4%), inflammatory cytokines interleukin-17 (10.4 ± 2.7%), and tumor necrosis factor-α (8.2 ± 3.1%) were also observed. This study demonstrated that MEP-based adsorbent not only removed pathogenic autoantibodies directly from the blood as with protein A adsorbents but also modulated cellular immunity through removal of complement components and related proinflammatory cytokines, thereby providing a potentially superior strategy for the treatment of MG.


Asunto(s)
Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Inmunoadsorbentes/uso terapéutico , Miastenia Gravis Autoinmune Experimental/sangre , Miastenia Gravis Autoinmune Experimental/tratamiento farmacológico , Miastenia Gravis Autoinmune Experimental/inmunología , Piridinas/uso terapéutico , Adsorción , Animales , Citocinas/sangre , Citocinas/inmunología , Femenino , Humanos , Inmunoadsorbentes/química , Ensayo de Materiales , Estructura Molecular , Piridinas/química , Distribución Aleatoria , Ratas , Ratas Endogámicas Lew , Receptores Colinérgicos/genética , Receptores Colinérgicos/inmunología
12.
Mol Immunol ; 46(1): 192-201, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18799218

RESUMEN

Myasthenia gravis (MG) and experimental autoimmune MG (EAMG) are antibody-mediated autoimmune diseases in which the nicotinic acetylcholine receptor (AChR) is the major autoantigen. Previously we have revealed that oral treatment with the less native recombinant fragment of the extracellular domain of the human AChR (Halpha1-205) suppressed ongoing EAMG, whereas the more native recombinant Trx-Halpha1-210 exacerbated EAMG. In this study, we speculated on the role of B-cell epitopes in oral tolerogens for the induction of oral tolerance in EAMG. We developed a B-cell epitope-free AChR fragment (BF-AChR) by removing two major B-cell epitopes (67-76 and 129-145) from Trx-Halpha1-210. BF-AChR exhibited a poor response to EAMG sera and to AChR-specific B- and T-cells while its parent fragment, Trx-Halpha1-210, showed much higher reactivity. Oral administration of BF-AChR ameliorated the symptoms in ongoing myasthenic rats accompanied by a significant decrease in AChR-specific humoral and Th1 cellular responses. The underlying mechanism for BF-AChR-induced oral tolerance was mediated by a shift from Th1 to regulatory T-cell (IL-10(+), CD4(+) TGF-beta(+) or Foxp3(+)) responses. This shift was assessed by changes in the cytokine profile and a deviation in the anti-AChR IgG isotypes from IgG2a/IgG2b to IgG1. Our results suggest that the removal of pathogenic B-cell epitopes from AChR fragments increases tolerogenicity by reducing the activation and proliferation of autoreactive B- and T-cells. Collectively, careful consideration of the immunogenicity of a tolerogen is necessary to induce successful oral tolerance in autoimmune disorders.


Asunto(s)
Epítopos de Linfocito B/inmunología , Miastenia Gravis Autoinmune Experimental/tratamiento farmacológico , Miastenia Gravis Autoinmune Experimental/inmunología , Receptores Colinérgicos/inmunología , Proteínas Recombinantes/uso terapéutico , Administración Oral , Animales , Autoanticuerpos/inmunología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Citocinas/inmunología , Regulación hacia Abajo/efectos de los fármacos , Femenino , Inmunoglobulina G/inmunología , Inmunohistoquímica , Mediadores de Inflamación/inmunología , Miastenia Gravis Autoinmune Experimental/sangre , Ratas , Ratas Endogámicas Lew , Receptores Colinérgicos/administración & dosificación , Proteínas Recombinantes/farmacología , Eliminación de Secuencia , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Células TH1/efectos de los fármacos , Células TH1/inmunología
13.
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
14.
J Neuroimmunol ; 201-202: 128-35, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18632163

RESUMEN

Myasthenia gravis (MG) is frequently treated by corticosteroids such as methylprednisolone. However, continuous treatment with steroids often results in adverse effects. In the present study we evaluated the therapeutic potential of a combination of suboptimal doses of methylprednisolone (Solumedrol) and Pentoxifylline (PTX), a general phosphodiesterase (PDE) inhibitor, in rat experimental autoimmune MG (EAMG). This combined treatment resulted in a pronounced suppressive effect on EAMG and was by far more effective than each of the drugs administered separately at these low doses. The suppressive effect on EAMG was accompanied by decreased humoral and cellular responses to AChR as well as down-regulated mRNA expression levels of Th1 cytokines and IL-10 in lymph node cells and of PDE-4 and cathepsin-l in the muscle. This study demonstrates the potential of PTX as a steroid-sparing agent in the management of myasthenia gravis.


Asunto(s)
Glucocorticoides/uso terapéutico , Metilprednisolona/uso terapéutico , Miastenia Gravis Autoinmune Experimental/tratamiento farmacológico , Pentoxifilina/uso terapéutico , Inhibidores de Fosfodiesterasa/uso terapéutico , Animales , Anticuerpos/sangre , Peso Corporal/efectos de los fármacos , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Miastenia Gravis Autoinmune Experimental/sangre , Miastenia Gravis Autoinmune Experimental/patología , Ratas , Ratas Endogámicas Lew , Receptores Colinérgicos/inmunología , Índice de Severidad de la Enfermedad
15.
Clin Immunol ; 128(2): 172-80, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18502693

RESUMEN

We had previously observed that treatment utilizing granulocyte-macrophage colony-stimulating factor (GM-CSF) had profound effects on the induction of experimental autoimmune myasthenia gravis (EAMG), a well-characterized antibody-mediated autoimmune disease. In this study, we show that EAMG induced by repeated immunizations with acetylcholine receptor (AChR) protein in C57BL6 mice is effectively suppressed by GM-CSF treatment administered at a stage of chronic, well-established disease. In addition, this amelioration of clinical disease is accompanied by down-modulation of both autoreactive T cell, and pathogenic autoantibody responses, a mobilization of DCs with a tolerogenic phenotype, and an expansion of regulatory T cells (Tregs) that potently suppress AChR-stimulated T cell proliferation in vitro. These observations suggest that the mobilization of antigen-specific Tregs in vivo using pharmacologic agents, like GM-CSF, can modulate ongoing anti-AChR immune responses capable of suppressing antibody-mediated autoimmunity.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Miastenia Gravis Autoinmune Experimental/inmunología , Animales , Anticuerpos/sangre , Anticuerpos/inmunología , Especificidad de Anticuerpos , División Celular , Enfermedad Crónica , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Miastenia Gravis Autoinmune Experimental/sangre , Miastenia Gravis Autoinmune Experimental/tratamiento farmacológico , Receptores Colinérgicos/inmunología , Linfocitos T Reguladores/fisiología
16.
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
17.
J Immunol ; 170(1): 604-12, 2003 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-12496449

RESUMEN

Myasthenia gravis (MG) is a T cell-dependent, Ab-mediated autoimmune disease. Ab against muscle acetylcholine receptor (AChR) cause the muscular weakness that characterizes MG and its animal model, experimental MG (EMG). EMG is induced in C57BL6 (B6) mice by three injections of Torpedo AChR (TAChR) in adjuvant. B6 mice develop anti-TAChR Ab that cross-react with mouse muscle AChR, but their CD4+ T cells do not cross-react with mouse AChR sequences. Moreover, murine EMG is not self-maintaining as is human MG, and it has limited duration. Several studies suggest that IL-4 has a protecting function in EMG. Here we show that B6 mice genetically deficient in IL-4 (IL-4-/-) develop long-lasting muscle weakness after a single immunization with TAChR. They develop chronic self-reactive Ab, and their CD4+ T cells respond not only to the TAChR and TAChR subunit peptides, but also to several mouse AChR subunit peptides. These results suggest that in B6 mice, regulatory mechanisms that involve IL-4 contribute to preventing the development of a chronic Ab-mediated autoimmune response to the AChR.


Asunto(s)
Interleucina-4/deficiencia , Interleucina-4/genética , Miastenia Gravis Autoinmune Experimental/genética , Miastenia Gravis Autoinmune Experimental/inmunología , Secuencia de Aminoácidos , Animales , Autoanticuerpos/sangre , Antígeno B7-1/biosíntesis , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Antígenos CD40/biosíntesis , Enfermedad Crónica , Epítopos de Linfocito T/inmunología , Predisposición Genética a la Enfermedad , Inmunización , Inyecciones Subcutáneas , Interferón gamma/metabolismo , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miastenia Gravis Autoinmune Experimental/sangre , Fragmentos de Péptidos/inmunología , Receptores Colinérgicos/administración & dosificación , Receptores Colinérgicos/inmunología , Índice de Severidad de la Enfermedad , Bazo/citología , Bazo/inmunología , Bazo/metabolismo , Torpedo/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA