Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Arthritis Rheumatol ; 69(10): 2052-2061, 2017 10.
Article in English | MEDLINE | ID: mdl-28666081

ABSTRACT

OBJECTIVE: Both environmental and genetic factors are important in the development of antiphospholipid antibodies (aPL) in patients with antiphospholipid syndrome (APS). Currently, the only available data on predisposing genetic factors have been obtained from epidemiologic studies, without mechanistic evidence. Therefore, we studied the influence of major histocompatibility complex (MHC) class II alleles on the production of aPL in a mouse model of APS. METHODS: Three groups of mice, MHC class II-deficient (MHCII-/- ) mice, MHCII-/- mice transgenic for human HLA-DQ6 (DQ6), DQ8, or DR4 alleles, and the corresponding wild-type (WT) mouse strains were immunized; half were immunized with human ß2 -glycoprotein I (ß2 GPI), and the other half were immunized with control ovalbumin (OVA) protein. Thrombus formation in vivo, tissue factor activity in carotid and peritoneal macrophages, and serum levels of tumor necrosis factor (TNF), IgG anticardiolipin (aCL), antibodies, and anti-OVA antibodies were determined. RESULTS: Immunization with ß2 GPI induced significant production of aCL and anti-ß2 GPI in WT mice compared with control mice immunized with OVA (P < 0.001) but diminished aCL (P < 0.001) and anti-ß2 GPI (P = 0.016) production in MHCII-/- mice. Anti-ß2 GPI production was fully restored in DQ6 and DQ8 mice, while levels of anti-ß2 GPI in DR4 mice and aCL in all transgenic lines were only partially restored (P < 0.001 to P < 0.046). Thrombus size in WT mice was twice that in MHCII-/- mice (P < 0.001) but similar to that in all transgenic lines. Carotid and peritoneal macrophage tissue factor levels decreased by >50% in MHCII-/- mice compared with wild-type B6 mice and were restored in DQ8 mice but not DR4 mice or DQ6 mice. TNF levels decreased 4-fold in MHCII-/- mice (P < 0.001) and were not restored in transgenic mice. CONCLUSION: Our mechanistic study is the first to show that MHC class II alleles influence not only quantitative aPL production but also the pathogenic capacity of induced aPL.


Subject(s)
Antibodies, Antiphospholipid/immunology , Genes, MHC Class II/genetics , HLA-DQ Antigens/genetics , HLA-DR4 Antigen/genetics , Alleles , Animals , Antibodies, Anticardiolipin/immunology , Carotid Arteries/immunology , Disease Models, Animal , Humans , Immunization , Immunoglobulin G/immunology , Macrophages/immunology , Macrophages, Peritoneal/immunology , Mice , Mice, Knockout , Mice, Transgenic , Ovalbumin/immunology , Severity of Illness Index , Thrombosis , Tumor Necrosis Factor-alpha/immunology , beta 2-Glycoprotein I/immunology
2.
J Neuroimmunol ; 295-296: 84-92, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27235354

ABSTRACT

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


Subject(s)
Immunoglobulin G/immunology , Myasthenia Gravis, Autoimmune, Experimental/chemically induced , Myasthenia Gravis, Autoimmune, Experimental/immunology , Receptor Protein-Tyrosine Kinases/toxicity , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Autoantibodies/blood , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Freund's Adjuvant/toxicity , Immunization , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myasthenia Gravis, Autoimmune, Experimental/pathology , Neuromuscular Junction/immunology , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Receptors, Cholinergic , Severity of Illness Index , Statistics, Nonparametric
3.
Immunol Lett ; 168(2): 306-12, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26493475

ABSTRACT

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


Subject(s)
HLA-DQ Antigens/immunology , HLA-DR3 Antigen/immunology , Myasthenia Gravis, Autoimmune, Experimental/immunology , Oculomotor Muscles/immunology , Animals , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Freund's Adjuvant/immunology , HLA-DQ Antigens/genetics , HLA-DR3 Antigen/genetics , Humans , Immunization/methods , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin M/blood , Immunoglobulin M/immunology , Lymphocytes/immunology , Lymphocytes/pathology , Mice, Transgenic , Microscopy, Fluorescence , Myasthenia Gravis, Autoimmune, Experimental/chemically induced , Myasthenia Gravis, Autoimmune, Experimental/genetics , Oculomotor Muscles/metabolism , Oculomotor Muscles/pathology , Protein Subunits/immunology , Receptors, Cholinergic/immunology
4.
J Neuroimmunol ; 281: 68-72, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25867470

ABSTRACT

Myasthenia gravis is an autoimmune disease characterized by muscle weakness due to neuromuscular junction (NMJ) damage by anti-acetylcholine receptor (AChR) auto-antibodies and complement. In experimental autoimmune myasthenia gravis (EAMG), which is induced by immunization with Torpedo AChR in CFA, anti-AChR IgG2b and IgG1 are the predominant isotypes in the circulation. Complement activation by isotypes such as IgG2b plays a crucial role in EAMG pathogenesis; this suggested the possibility that IgG1, which does not activate complement through the classical pathway, may suppress EAMG. In this study, we show that AChR-immunized BALB/c mice genetically deficient for IgG1 produce higher levels of complement-activating isotypes of anti-AChR, especially IgG3 and IgG2a, and develop increased IgG3/IgG2a deposits at the NMJ, as compared to wild type (WT) BALB/c mice. Consistent with this, AChR-immunized IgG1(-/-) BALB/c mice lose muscle strength and muscle AChR to a greater extent than AChR-immunized WT mice. These observations demonstrate that IgG1 deficiency leads to increased severity of EAMG associated with an increase in complement activating IgG isotypes. Further studies are needed to dissect the specific role or mechanism of IgG1 in limiting EAMG and that of EAMG exacerbating role of complement activating IgG3 and IgG2a in IgG1 deficiency.


Subject(s)
Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/metabolism , Animals , Female , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Torpedo
5.
Exp Neurol ; 270: 11-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25697844

ABSTRACT

Myasthenia gravis (MG) is an autoimmune disorder characterized by generalized muscle weakness due to neuromuscular junction (NMJ) dysfunction brought by acetylcholine receptor (AChR) antibodies in most cases. Although steroids and other immunosuppressants are effectively used for treatment of MG, these medications often cause severe side effects and a complete remission cannot be obtained in many cases. For pre-clinical evaluation of more effective and less toxic treatment methods for MG, the experimental autoimmune myasthenia gravis (EAMG) induced by Torpedo AChR immunization has become one of the standard animal models. Although numerous compounds have been recently proposed for MG mostly by using the active immunization EAMG model, only a few have been proven to be effective in MG patients. The variability in the experimental design, immunization methods and outcome measurements of pre-clinical EAMG studies make it difficult to interpret the published reports and assess the potential for application to MG patients. In an effort to standardize the active immunization EAMG model, we propose standard procedures for animal care conditions, sampling and randomization of mice, experimental design and outcome measures. Utilization of these standard procedures might improve the power of pre-clinical EAMG experiments and increase the chances for identifying promising novel treatment methods that can be effectively translated into clinical trials for MG.


Subject(s)
Immunization/standards , Myasthenia Gravis, Autoimmune, Experimental , Animals , Autoantibodies/immunology , Guidelines as Topic , Immunization/methods , Mice , Receptors, Cholinergic/immunology
6.
Rev Neurosci ; 25(4): 575-83, 2014.
Article in English | MEDLINE | ID: mdl-24731953

ABSTRACT

While the complement system is desired for protective immunity, antibody- and complement-mediated neuromuscular junction (NMJ) destruction, a hallmark of myasthenia gravis (MG) or experimental autoimmune MG (EAMG), is a significant concern. Evidence suggests that the binding of complement factors to the pathogenic anti-acetylcholine receptor (AChR) autoantibody induces the formation of membrane attack complexes (MAC), which ultimately lead to NMJ destruction and muscle weakness. Studies corroborating the evidence show that the complement (C3-C6)-deficient or complement inhibitor (anti-C1q, soluble CR1, anti-C6, and C5 inhibiting peptide)-treated animals are highly resistant to EAMG induction, whereas the deficiency of the naturally occurring complement inhibitors, such as the decay-accelerating factor (DAF), increases EAMG susceptibility. Notably, the complement-inhibited animals do not exhibit significant immunosuppression but only a marginal reduction in the production of certain cytokines and immunoglobulin isotypes. A preliminary clinical trial using antibody-based C5 inhibitor eculizumab has been shown to be of potential use for MG treatment. The inhibition of the classic complement pathway (CCP) alone appears to be enough to suppress EAMG, suggesting that the complement inhibitors targeting specifically the classic pathway could effectively treat MG without causing immunosuppressive and other side effects. For instance, a recent non-antibody-based therapeutic approach selectively targeting the CCP component C2 by small interfering RNA (siRNA) has proven useful in EAMG treatment. The treatment strategies developed for MG might also be beneficial for other complement-mediated autoimmune diseases.


Subject(s)
Complement Inactivating Agents/therapeutic use , Myasthenia Gravis/therapy , Animals , Antibodies, Monoclonal/therapeutic use , Humans , Immunosuppression Therapy , Myasthenia Gravis/immunology
7.
Clin Immunol ; 151(2): 155-63, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24589747

ABSTRACT

Myasthenia gravis (MG) is an autoimmune disease characterized by muscle weakness associated with acetylcholine receptor (AChR), muscle-specific receptor kinase (MuSK) or low-density lipoprotein receptor-related protein 4 (LRP4)-antibodies. MuSK-antibodies are predominantly of the non-complement fixing IgG4 isotype. The MuSK associated experimental autoimmune myasthenia gravis (EAMG) model was established in mice to investigate immunoglobulin (Ig) and cytokine responses related with MuSK immunity. C57BL/6 (B6) mice immunized with 30µg of recombinant human MuSK in incomplete or complete Freund's adjuvant (CFA) showed significant EAMG susceptibility (>80% incidence). Although mice immunized with 10µg of MuSK had lower EAMG incidence (14.3%), serum MuSK-antibody levels were comparable to mice immunized with 30µg MuSK. While MuSK immunization stimulated production of all antibody isotypes, non-complement fixing IgG1 was the dominant anti-MuSK Ig isotype in both sera and neuromuscular junctions. Moreover, MuSK immunized IgG1 knockout mice showed very low serum MuSK-antibody levels. Sera and MuSK-stimulated lymph node cell supernatants of MuSK immunized mice showed significantly higher levels of IL-4 and IL-10 (but not IFN-γ and IL-12), than those of CFA immunized mice. Our results suggest that through activation of Th2-type cells, anti-MuSK immunity promotes production of IL-4, which in turn activates anti-MuSK IgG1, the mouse analog of human IgG4. These findings might provide clues for the pathogenesis of other IgG4-related diseases as well as development of disease specific treatment methods (e.g. specific IgG4 inhibitors) for MuSK-related MG.


Subject(s)
Immunoglobulin G/metabolism , Interleukin-10/metabolism , Interleukin-4/metabolism , Myasthenia Gravis/immunology , Receptor Protein-Tyrosine Kinases/immunology , Animals , Antibody Specificity , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/immunology , Immunization , Immunoglobulin G/genetics , Interleukin-10/genetics , Interleukin-4/genetics , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Knockout
8.
Autoimmun Rev ; 12(9): 904-11, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23537510

ABSTRACT

The complement system is profoundly involved in the pathogenesis of acetylcholine receptor (AChR) antibody (Ab) related myasthenia gravis (MG) and its animal model experimental autoimmune myasthenia gravis (EAMG). The most characteristic finding of muscle pathology in both MG and EAMG is the abundance of IgG and complement deposits at the nerve-muscle junction (NMJ), suggesting that AChR-Ab induces muscle weakness by complement pathway activation and consequent membrane attack complex (MAC) formation. This assumption has been supported with EAMG resistance of complement factor C3 knockout (KO), C4 KO and C5 deficient mice and amelioration of EAMG symptoms following treatment with complement inhibitors such as cobra venom factor, soluble complement receptor 1, anti-C1q, anti-C5 and anti-C6 Abs. Moreover, the complement inhibitor decay accelerating factor (DAF) KO mice exhibit increased susceptibility to EAMG. These findings have brought forward improvisation of novel therapy methods based on inhibition of classical and common complement pathways in MG treatment.


Subject(s)
Complement System Proteins/immunology , Myasthenia Gravis/immunology , Neuromuscular Junction/pathology , Animals , Autoantibodies/immunology , Complement Activation , Humans , Myasthenia Gravis/drug therapy , Myasthenia Gravis/metabolism , Myasthenia Gravis/pathology , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/metabolism , Myasthenia Gravis, Autoimmune, Experimental/pathology
9.
Curr Protoc Immunol ; Chapter 15: Unit 15.8., 2013.
Article in English | MEDLINE | ID: mdl-23392639

ABSTRACT

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.


Subject(s)
Fish Proteins/immunology , Muscle, Skeletal/immunology , Myasthenia Gravis, Autoimmune, Experimental/immunology , Neuromuscular Junction/drug effects , Receptors, Nicotinic/immunology , Synaptic Transmission/drug effects , Animals , Autoantibodies/biosynthesis , Autoantibodies/immunology , Complement System Proteins/immunology , Electromyography , Female , Fish Proteins/administration & dosage , Fish Proteins/isolation & purification , Immunoglobulin G/immunology , Male , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Myasthenia Gravis, Autoimmune, Experimental/metabolism , Myasthenia Gravis, Autoimmune, Experimental/physiopathology , Neuromuscular Junction/physiopathology , Radioimmunoassay , Receptors, Nicotinic/administration & dosage , Receptors, Nicotinic/isolation & purification , Torpedo/physiology
10.
J Autoimmun ; 42: 94-104, 2013 May.
Article in English | MEDLINE | ID: mdl-23410585

ABSTRACT

Activation of complement components is crucial in the progression and severity of myasthenia gravis and experimental autoimmune myasthenia gravis (EAMG). Mice deficient in complement component C4 or treated with monoclonal antibody to C1q are resistant to EAMG. In this study, we show that inhibition of complement cascade activation by suppressing the expression of a critical low-abundant protein, C2, in the classical complement pathway, significantly improved clinical and immunopathological manifestations of EAMG. Two weeks after a second booster immunization with acetylcholine receptor, when mice exhibit muscle weakness, i.p. injection of C2 siRNA significantly suppressed C2 mRNA in the blood cells and liver of EAMG mice. Treatment of EAMG mice with C2 siRNA, once a week for 5 weeks, significantly improved muscle strength, which was further evidenced by functional AChR preservation in muscle, reduction in number of C3 and membrane-attack complexes at neuro-muscular junctions in forelimb muscle sections, and a transient decrease in serum IgG2b levels. Our study shows for the first time that siRNA-mediated suppression of C2, a component of the classical complement system, following established disease, can effectively contribute to the remission of EAMG. Therefore, C2 siRNA mediated therapy can be applied in all complement mediated autoimmune diseases.


Subject(s)
Complement C2/antagonists & inhibitors , Liver/immunology , Muscles/immunology , Myasthenia Gravis, Autoimmune, Experimental/therapy , RNA, Small Interfering/genetics , Animals , Complement Activation/genetics , Complement C2/genetics , Complement Membrane Attack Complex/metabolism , Female , Humans , Immunoglobulin G/blood , Mice , Mice, Inbred C57BL , Muscles/pathology , Receptors, Cholinergic/immunology
11.
J Neuroimmunol ; 256(1-2): 49-54, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23266128

ABSTRACT

CD21, a major complement receptor expressed on B cells, is associated with autoimmune disorders. In the present study, we investigated the role of CD21 in pathogenesis of myasthenia gravis (MG) in relationship to anti-acetylcholine receptor (AchR) IgG (anti-AchR IgG) secretion. We detected increased surface expression of CD21 on AchR specified B cells as well as decreased surface expression of CD21 on total B cells in peripheral blood of patients with generalized MG (gMG). In addition, the serum concentrations of soluble secreted CD21 (sCD21) were decreased in patients with gMG. We also found that the level of CD21(+) AchR specified B cells correlated positively with serum anti-AchR IgG level, while the serum concentration of soluble CD21 correlated negatively with serum anti-AchR IgG level. Our data suggests that CD21 might facilitate its function on AchR specified B cell activation, resulting in the secretion of anti-AchR IgG.


Subject(s)
B-Lymphocytes/metabolism , Gene Expression Regulation/physiology , Myasthenia Gravis/pathology , Receptors, Cholinergic/metabolism , Receptors, Complement 3d/metabolism , Adult , Antigens, CD19/immunology , Antigens, CD19/metabolism , Autoantibodies , B-Lymphocytes/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Male , Middle Aged , Myasthenia Gravis/immunology , Receptors, Cholinergic/genetics , Receptors, Cholinergic/immunology , Receptors, Complement 3d/genetics , Statistics as Topic
12.
Ann N Y Acad Sci ; 1274: 133-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23252908

ABSTRACT

Experimental autoimmune myasthenia gravis (EAMG) in mice has been used to unravel the pathogenic mechanisms and to be used as a preclinical model of myasthenia gravis (MG). Induction of predominantly ocular EAMG in HLA-DQ8 transgenic mice immunized with acetylcholine receptor (AChR) subunits demonstrated the importance of nonconformationally expressed AChR subunits in extraocular muscle involvement. Wild-type (WT) and CD4(+) T cell knockout (KO) C57BL/6 mice developed EAMG upon immunization with AChR in incomplete Freund's adjuvant plus lipopolysaccharide. AChR-specific IgG2(+) B cell frequencies, estimated by Alexa-conjugated AChR, and AChR-reactive IgG2b levels significantly correlated with the clinical grades of EAMG in WT mice. CD4(+) T cell-deficient EAMG mice exhibited AChR antibodies mainly of the IgG2b isotype, emphasizing T helper-independent B cell activation pathways in EAMG induction. These novel EAMG models have suggested that diverse immunopathological mechanisms might contribute to EAMG or MG pathogenesis.


Subject(s)
Myasthenia Gravis, Autoimmune, Experimental/immunology , Receptors, Cholinergic/metabolism , Animals , Autoantibodies/immunology , Cytokines/immunology , HLA-DQ Antigens/immunology , HLA-DQ Antigens/metabolism , Immunoglobulin G/immunology , Mice , Mice, Knockout , Myasthenia Gravis, Autoimmune, Experimental/pathology , Receptors, Cholinergic/immunology
13.
J Neuroimmunol ; 249(1-2): 1-7, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22626443

ABSTRACT

The potential of lipopolysaccharide (LPS) to induce antigen-specific B cell responses to acetylcholine receptor (AChR) in myasthenia gravis (MG) was evaluated in wild type (WT) and CD4-/- C57BL/6 mice. The WT mice immunized with AChR in LPS developed an MG-like disease (LPS-EAMG) similar to that induced by immunization with AChR in complete Freund's adjuvant (CFA-EAMG). CD4-/- mice were resistant to CFA-EAMG but susceptible to LPS-EAMG. LPS abrogated EAMG resistance in CD4-/- mice by increasing high-affinity anti-AChR IgG2b in sera and enhancing immune complex deposition in muscle.


Subject(s)
Adjuvants, Immunologic/pharmacology , CD4-Positive T-Lymphocytes/immunology , Lipopolysaccharides/pharmacology , Lymphocyte Activation/immunology , Myasthenia Gravis, Autoimmune, Experimental/immunology , Animals , Antigen-Antibody Complex/immunology , Autoantibodies/blood , Autoantibodies/immunology , Autoantigens/immunology , Enzyme-Linked Immunosorbent Assay , Freund's Adjuvant/immunology , Freund's Adjuvant/pharmacology , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Receptors, Cholinergic/immunology
14.
Muscle Nerve ; 45(2): 209-16, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22246876

ABSTRACT

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.


Subject(s)
Immunization/adverse effects , Myasthenia Gravis, Autoimmune, Experimental/etiology , Receptors, Nicotinic/immunology , Animals , Antibodies, Anti-Idiotypic/blood , Antibodies, Anti-Idiotypic/immunology , Arabidopsis Proteins/metabolism , Cell Proliferation , Complement C3/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , HLA-DQ Antigens/genetics , Humans , Intramolecular Transferases/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Muscle Strength/genetics , Myasthenia Gravis, Autoimmune, Experimental/blood , Myasthenia Gravis, Autoimmune, Experimental/genetics , Myasthenia Gravis, Autoimmune, Experimental/pathology , Neuromuscular Junction/immunology , Ocular Motility Disorders/etiology , Ocular Motility Disorders/immunology , Protein Subunits/genetics , Protein Subunits/metabolism
15.
Curr Protoc Immunol ; Chapter 15: Unit 15.23, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22048803

ABSTRACT

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.


Subject(s)
Myasthenia Gravis, Autoimmune, Experimental/immunology , Receptors, Nicotinic/immunology , Animals , Electric Organ/immunology , Electromyography , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , Muscle, Skeletal/immunology , Neuromuscular Junction/immunology , Receptors, Nicotinic/isolation & purification , Torpedo/immunology
16.
Cell Immunol ; 271(2): 292-8, 2011.
Article in English | MEDLINE | ID: mdl-21861992

ABSTRACT

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.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Myasthenia Gravis, Autoimmune, Experimental/blood , Myasthenia Gravis, Autoimmune, Experimental/immunology , Receptors, Cholinergic/blood , Animals , Autoantibodies/blood , Cyclic AMP/analogs & derivatives , Flow Cytometry , Immunization , Immunoglobulin G/blood , Immunoglobulin M/blood , Mice , Mice, Inbred C57BL , Torpedo/immunology
17.
J Autoimmun ; 37(2): 136-43, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21636248

ABSTRACT

Myasthenia gravis (MG) is a T cell-dependent and antibody-mediated disease in which the target antigen is the skeletal muscle acetylcholine receptor (AChR). In the last few decades, several immunological factors involved in MG pathogenesis have been discovered mostly by studies utilizing the experimental autoimmune myasthenia gravis (EAMG) model. Nevertheless, MG patients are still treated with non-specific global immunosuppression that is associated with severe chronic side effects. Due to the high heterogeneity of AChR epitopes and antibody responses involved in MG pathogenesis, the specific treatment of MG symptoms have to be achieved by inhibiting the complement factors and cytokines involved in anti-AChR immunity. EAMG studies have clearly shown that inhibition of the classical and common complement pathways effectively and specifically diminish the neuromuscular junction destruction induced by anti-AChR antibodies. The inborn or acquired deficiencies of IL-6, TNF-α and TNF receptor functions are associated with the lowest EAMG incidences. Th17-type immunity has recently emerged as an important contributor of EAMG pathogenesis. Overall, these results suggest that inhibition of the complement cascade and the cytokine networks alone or in combination might aid in development of future treatment models that would reduce MG symptoms with highest efficacy and lowest side effect profile.


Subject(s)
Complement System Proteins/immunology , Cytokines/immunology , Myasthenia Gravis/drug therapy , Myasthenia Gravis/immunology , Receptors, Cholinergic/immunology , Animals , Autoantibodies/immunology , Complement Activation/drug effects , Disease Models, Animal , Humans , Mice , Molecular Targeted Therapy/trends , Th17 Cells/immunology
18.
J Neuroimmunol ; 234(1-2): 165-7, 2011 May.
Article in English | MEDLINE | ID: mdl-21481948

ABSTRACT

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.


Subject(s)
Estrogen Receptor alpha/deficiency , Myasthenia Gravis, Autoimmune, Experimental/genetics , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/pathology , Animals , Antibodies/blood , Cytokines/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Genetic Predisposition to Disease/genetics , Hand Strength/physiology , Immunization/adverse effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Myasthenia Gravis, Autoimmune, Experimental/physiopathology , Receptors, Cholinergic/immunology , Time Factors
19.
Inflamm Res ; 59(3): 197-205, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19768385

ABSTRACT

OBJECTIVE: To observe the therapeutic effect of RelB-silenced dendritic cells (DCs) in experimental autoimmune myasthenia gravis (EAMG), and further to investigate the mechanism of this immune system therapeutic. METHODS: (1) RelB-silenced DCs and control DCs were prepared and the supernatants were collected for IL-12p70, IL-6, and IL-23 measurement by ELISA. (2) RelB-silenced DCs and control DCs were co-cultured with AChR-specific T cells, and the supernatant was collected to observe the IL-17, IFN-gamma, IL-4 production. (3) EAMG mice with clinical scores of 1 to 2 were collected and enrolled randomly into the RelB-silenced DC group or the control DC group. RelB-silenced DCs or an equal amount of control DCs were injected intravenously on days 0, 7, and 14 after enrollment. Clinical scores were evaluated every other day. Twenty days after allotment, serum from individual mice was collected to detect serum concentrations of anti-mouse AChR IgG, IgG1, IgG2b, and IgG2c. The splenocytes were isolated for analysis of lymphocyte proliferative responses, cytokine (IL-17, IFN-gamma, IL-4) production, and protein levels of RORgammat, T-bet, GATA-3, and FoxP3 (the special transcription factors of Th17, Th1, Th2, and Treg, respectively). RESULTS: (1) RelB-silenced DCs produced significantly reduced amounts of IL-12p70, IL-6, and IL-23, as compared with control DCs. (2) RelB-silenced DCs spurred on the CD4(+) T cells from Th1/Th17 to the Th2 cell subset in the co-culture system. (3) Treatment with RelB-silenced DCs effectively reduced myasthenic symptoms and levels of serum anti-acetylcholine receptor autoantibody in mice with ongoing EAMG. Th17-related markers (RORgammat, IL-17) and Th1-related markers (T-bet, IFN-gamma) were downregulated, whereas Th2 markers (IL-4 and GATA3) and Treg marker (FoxP3) were upregulated. CONCLUSIONS: RelB-silenced DCs were able to create a particular cytokine environment that was absent of inflammatory cytokines. RelB-silenced DCs provide a practical means to normalize the differentiation of the four T-cell subsets (Th17, Th1, Th2, and Treg) in vivo, and thus possess therapeutic potential in Th1/Th17-dominant autoimmune disorders such as myasthenia gravis.


Subject(s)
Dendritic Cells/cytology , Forkhead Transcription Factors/metabolism , Myasthenia Gravis, Autoimmune, Experimental/therapy , T-Lymphocytes, Regulatory/pathology , Th1 Cells/pathology , Th2 Cells/pathology , Transcription Factor RelB/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Proliferation , Cell- and Tissue-Based Therapy , Cytokines/metabolism , Dendritic Cells/metabolism , Female , Immunoglobulin G/blood , Mice , Mice, Inbred C57BL , Myasthenia Gravis, Autoimmune, Experimental/metabolism , Myasthenia Gravis, Autoimmune, Experimental/pathology , Receptors, Cholinergic/immunology , Receptors, Cholinergic/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/pathology , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/metabolism , Th2 Cells/metabolism , Transcription Factor RelB/genetics
20.
J Neuroimmunol ; 208(1-2): 40-5, 2009 Mar 31.
Article in English | MEDLINE | ID: mdl-19193448

ABSTRACT

Classical complement pathway factor, C4 is required for experimental autoimmune myasthenia gravis (EAMG) pathogenesis. C4 is also a central component of the mannose binding lectin (MBL) pathway suggesting that this pathway might also be involved in MG pathogenesis. However, MBL gene deficient mice displayed intact anti-acetylcholine receptor (AChR)-immune response and neuromuscular junction (NMJ) IgG and complement accumulation following AChR-immunization. Moreover, no significant difference was observed between the serum MBL levels of 77 anti-AChR antibody positive generalized MG patients and 105 healthy controls. Therefore, MBL pathway does not play a role in EAMG/MG pathogenesis.


Subject(s)
Complement Pathway, Mannose-Binding Lectin/immunology , Mannose-Binding Lectin/blood , Myasthenia Gravis/immunology , Signal Transduction/immunology , Adult , Aged , Animals , Complement Pathway, Mannose-Binding Lectin/genetics , Female , Humans , Immunoglobulin G/biosynthesis , Male , Mannose-Binding Lectin/deficiency , Mannose-Binding Lectin/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myasthenia Gravis/blood , Receptors, Cholinergic/immunology , Signal Transduction/genetics , Torpedo
SELECTION OF CITATIONS
SEARCH DETAIL
...