Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Neurology ; 102(12): e209482, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38781559

ABSTRACT

The role of immunosenescence, particularly the natural process of thymic involution during aging, is increasingly acknowledged as a factor contributing to the development of autoimmune diseases and cancer. Recently, a concern has been raised about deleterious consequences of the surgical removal of thymic tissue, including for patients who undergo thymectomy for myasthenia gravis (MG) or resection of a thymoma. This review adopts a multidisciplinary approach to scrutinize the evidence concerning the long-term risks of cancer and autoimmunity postthymectomy. We conclude that for patients with acetylcholine receptor antibody-positive MG and those diagnosed with thymoma, the removal of the thymus offers prominent benefits that well outweigh the potential risks. However, incidental removal of thymic tissue during other thoracic surgeries should be minimized whenever feasible.


Subject(s)
Myasthenia Gravis , Thymectomy , Thymoma , Thymus Gland , Thymus Neoplasms , Humans , Thymectomy/adverse effects , Thymectomy/methods , Myasthenia Gravis/surgery , Thymus Gland/surgery , Thymus Neoplasms/surgery , Thymus Neoplasms/complications , Thymoma/surgery , Thymoma/complications , Postoperative Complications/etiology , Autoimmune Diseases/surgery
2.
PLoS One ; 18(10): e0287654, 2023.
Article in English | MEDLINE | ID: mdl-37816000

ABSTRACT

OBJECTIVE: High-dose prednisone use, lasting several months or longer, is the primary initial therapy for myasthenia gravis (MG). Upwards of a third of patients do not respond to treatment. Currently no biomarkers can predict clinical responsiveness to corticosteroid treatment. We conducted a discovery-based study to identify treatment responsive biomarkers in MG using sera obtained at study entry to the thymectomy clinical trial (MGTX), an NIH-sponsored randomized, controlled study of thymectomy plus prednisone versus prednisone alone. METHODS: We applied ultra-performance liquid chromatography coupled with electro-spray quadrupole time of flight mass spectrometry to obtain comparative serum metabolomic and lipidomic profiles at study entry to correlate with treatment response at 6 months. Treatment response was assessed using validated outcome measures of minimal manifestation status (MMS), MG-Activities of Daily Living (MG-ADL), Quantitative MG (QMG) score, or a strictly defined composite measure of response. RESULTS: Increased serum levels of phospholipids were associated with treatment response as assessed by QMG, MMS, and the Responders classification, but all measures showed limited overlap in metabolomic profiles, in particular the MG-ADL. A panel including histidine, free fatty acid (13:0), γ-cholestenol and guanosine was highly predictive of the strictly defined treatment response measure. The AUC in Responders' prediction for these markers was 0.90 irrespective of gender, age, thymectomy or baseline prednisone use. Pathway analysis suggests that xenobiotic metabolism could play a major role in treatment resistance. There was no association with outcome and gender, age, thymectomy or baseline prednisone use. INTERPRETATION: We have defined a metabolomic and lipidomic profile that can now undergo validation as a treatment predictive marker for MG patients undergoing corticosteroid therapy. Metabolomic profiles of outcome measures had limited overlap consistent with their assessing distinct aspects of treatment response and supporting unique biological underpinning for each outcome measure. Interindividual variation in prednisone metabolism may be a determinate of how well patients respond to treatment.


Subject(s)
Activities of Daily Living , Myasthenia Gravis , Humans , Prednisone/adverse effects , Glucocorticoids/therapeutic use , Myasthenia Gravis/drug therapy , Combined Modality Therapy , Thymectomy/methods , Treatment Outcome
4.
Sci Immunol ; 8(85): eadj4913, 2023 07 14.
Article in English | MEDLINE | ID: mdl-37418544

ABSTRACT

Antigen activated naïve B cells undergoing germinal center responses have distinct metabolic requirements.


Subject(s)
B-Lymphocytes , Germinal Center , Lymphocyte Activation , Antigens
5.
J Immunol ; 205(7): 1743-1751, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32839239

ABSTRACT

Myasthenia gravis (MG) is an autoimmune disease in which Abs target neuromuscular junction proteins, in particular the acetylcholine receptor. We previously identified the antiapoptotic protein survivin in the autoreactive B cells and plasma cells of MG patients. To further define the role of survivin in MG, we have assessed PBMCs from 29 patients with MG and 15 controls. We confirmed the increased expression of survivin in CD20+ lymphocytes from MG patients compared with controls. Furthermore, the CD20+ population of cells from MG patients contained a higher percentage of extracellular survivin compared with controls. The analysis of CD4+ cells showed an increased percentage of intracellular survivin in MG patients compared with controls, whereas the extracellular survivin CD4+ percentage was unaffected. In an experimental mouse model of MG, we assessed the therapeutic potential of an Ab raised to a modified survivin peptide but cross-reactive to survivin. Ab treatment reduced disease severity, lowered acetylcholine receptor-specific Abs, and decreased CD19+ survivin+ splenocytes. The ability to target survivin through Ab recognition of autoreactive cells offers the potential for a highly specific therapeutic agent for MG.


Subject(s)
B-Lymphocytes/metabolism , Myasthenia Gravis/immunology , Survivin/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antibodies/metabolism , Antigens, CD20/metabolism , Autoantigens/immunology , B-Lymphocytes/immunology , Disease Models, Animal , Female , Humans , Immunity, Humoral , Male , Mice , Mice, Inbred C57BL , Middle Aged , Peptides/immunology , Young Adult
6.
Mol Ther Methods Clin Dev ; 13: 484-492, 2019 Jun 14.
Article in English | MEDLINE | ID: mdl-31193726

ABSTRACT

Complement-mediated damage to the neuromuscular junction (NMJ) is a key mechanism of pathology in myasthenia gravis (MG), and therapeutics inhibiting complement have shown evidence of efficacy in the treatment of MG. In this study, we describe the development of a subcutaneously administered N-acetylgalactosamine (GalNAc)-conjugated small interfering RNA (siRNA) targeting the C5 component of complement that silences C5 expression in the liver (ALN-CC5). Treatment of wild-type rodents with ALN-CC5 resulted in robust and durable suppression of liver C5 expression. Dose-dependent serum C5 suppression was observed in non-human primates, with a lowering of serum C5 of up to 97.5% and the concomitant inhibition of serum complement activity. C5 silencing was efficacious in ameliorating disease symptoms in two standard rat models of MG, demonstrating the key role of circulating C5 in pathology at the NMJ. Improvement in disease activity scores and NMJ pathology was observed at intermediate levels of complement activity inhibition, suggesting that complete ablation of complement activity may not be required for efficacy in MG. The pre-clinical studies of ALN-CC5 and efficacy of C5 silencing in rat models of MG support further clinical development of ALN-CC5 as a potential therapeutic for the treatment of MG and other complement-mediated disorders.

7.
PLoS One ; 13(10): e0205464, 2018.
Article in English | MEDLINE | ID: mdl-30308012

ABSTRACT

BACKGROUND: A characteristic pathology of early onset myasthenia gravis is thymic hyperplasia with ectopic germinal centers (GC). However, the mechanisms that trigger and maintain thymic hyperplasia are poorly characterized. Dysregulation of small, non-coding microRNAs (miRNAs) and their target genes has been identified in the pathology of several autoimmune diseases. We assessed the miRNA and mRNA profiles of the MG thymus and have investigated their role in GC formation and maintenance. METHODS: MG thymus samples were assessed by histology and grouped based upon the appearance of GC; GC positive and GC negative. A systems biology approach was used to study the differences between the groups. Our study included miRNA and mRNA profiling, quantitative real-time PCR validation, miRNA target identification, pathway analysis, miRNA-mRNA reciprocal expression pairing and interaction. RESULTS: Thirty-eight mature miRNAs and forty-six annotated mRNA transcripts were differentially expressed between the two groups (>1.5 fold change, ANOVA p<0.05). The miRNAs were found to be involved in immune response pathways and identified in other autoimmune diseases. The cellular and molecular functions of the mRNAs showed involvement in cell death and cell survival, cellular proliferation, cytokine signaling and extra-cellular matrix reorganization. Eleven miRNA and mRNA pairs were reciprocally regulated. The Regulator of G protein Signalling 13 (RGS13), known to be involved in GC regulation, was identified in specimens with GC and was paired with downregulation of miR-452-5p and miR-139-5p. MiRNA target sites were validated by dual luciferase assay. Transfection of miRNA mimics led to down regulation of RGS13 expression in Raji cells. CONCLUSION: Our study indicates a distinct miRNA and mRNA expression pattern in ectopic GC in MG thymus. These miRNAs and mRNAs are involved in regulatory pathways common to inflammation and immune response, cell cycle regulation and anti-apoptotic pathways suggesting their involvement in support of GC formation in the thymus. We demonstrate for the first time that miR-139-5p and miR-452-5p negatively regulate RGS13 expression.


Subject(s)
Gene Expression Profiling/methods , Germinal Center/chemistry , MicroRNAs/genetics , Myasthenia Gravis/genetics , RGS Proteins/genetics , Adult , Cell Line , Gene Expression Regulation , Gene Regulatory Networks , Humans , Middle Aged , RNA, Messenger/genetics , Signal Transduction , Systems Biology/methods , Thymus Gland , Young Adult
8.
Ann N Y Acad Sci ; 1413(1): 136-142, 2018 02.
Article in English | MEDLINE | ID: mdl-29356015

ABSTRACT

Because of the failure of many promising therapeutics identified in preclinical evaluation, funding sources have established guidelines for increased rigor in animal evaluations. The myasthenia gravis (MG) community of scientists has developed guidelines for preclinical assessment for potential MG treatments. Here, we provide a focused summary of these recommendations and the role of complement in disease development in experimental models of MG.


Subject(s)
Autoantibodies/immunology , Complement Inactivator Proteins/pharmacology , Complement System Proteins/immunology , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , Myasthenia Gravis, Autoimmune, Experimental/immunology , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Cholinergic/immunology , Animals , Complement System Proteins/genetics , Disease Models, Animal , Immunization, Passive/methods , Immunoglobulin G/immunology , Macaca mulatta , Mice , Mice, Inbred C57BL , Myasthenia Gravis, Autoimmune, Experimental/pathology , Rats , Rats, Inbred Lew
9.
Front Neurol ; 8: 230, 2017.
Article in English | MEDLINE | ID: mdl-28620344

ABSTRACT

Biomarkers that assess treatment response for patients with the autoimmune disorder, myasthenia gravis (MG), have not been evaluated to a significant extent. We hypothesized the pro-inflammatory cytokine, osteopontin (OPN), may be associated with variability of response to glucocorticoids (GCs) in patients with MG. A cohort of 250 MG patients treated with standardized protocol of GCs was recruited, and plasma OPN and polymorphisms of its gene, secreted phosphoprotein 1 (SPP1), were evaluated. Mean OPN levels were higher in patients compared to healthy controls. Carriers of rs11728697*T allele (allele definition: one of two or more alternative forms of a gene) were more frequent in the poorly GC responsive group compared to the GC responsive group indicating an association of rs11728697*T allele with GC non-responsiveness. One risk haplotype (AGTACT) was identified associated with GC non-responsiveness compared with GC responsive MG group. Genetic variations of SPP1 were found associated with the response to GC among MG patients.

10.
Cytokine ; 96: 279-285, 2017 08.
Article in English | MEDLINE | ID: mdl-28599246

ABSTRACT

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.


Subject(s)
Autoimmunity/immunology , Interleukin-17/genetics , Interleukin-17/physiology , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/physiopathology , Animals , Autoantibodies , Cytokines/immunology , Disease Models, Animal , Forkhead Transcription Factors/genetics , Humans , Immunoglobulin G/blood , Interleukin-17/deficiency , Mice , Mice, Knockout , Proto-Oncogene Proteins c-bcl-6/genetics , Real-Time Polymerase Chain Reaction , Severity of Illness Index , T-Lymphocytes, Regulatory
11.
Front Physiol ; 7: 524, 2016.
Article in English | MEDLINE | ID: mdl-27891095

ABSTRACT

The differential susceptibility of skeletal muscle by myasthenia gravis (MG) is not well understood. We utilized RNA expression profiling of extraocular muscle (EOM), diaphragm (DIA), and extensor digitorum (EDL) of rats with experimental autoimmune MG (EAMG) to evaluate the hypothesis that muscles respond differentially to injury produced by EAMG. EAMG was induced in female Lewis rats by immunization with acetylcholine receptor purified from the electric organ of the Torpedo. Six weeks later after rats had developed weakness and serum antibodies directed against the AChR, animals underwent euthanasia and RNA profiling performed on DIA, EDL, and EOM. Profiling results were validated by qPCR. Across the three muscles between the experiment and control groups, 359 probes (1.16%) with greater than 2-fold changes in expression in 7 of 9 series pairwise comparisons from 31,090 probes were identified with approximately two-thirds being increased. The three muscles shared 16 genes with increased expression and 6 reduced expression. Functional annotation demonstrated that these common expression changes fell predominantly into categories of metabolism, stress response, and signaling. Evaluation of specific gene function indicated that EAMG led to a change to oxidative metabolism. Genes related to muscle regeneration and suppression of immune response were activated. Evidence of a differential immune response among muscles was not evident. Each muscle had a distinct RNA profile but with commonality in gene categories expressed that are focused on muscle repair, moderation of inflammation, and oxidative metabolism.

12.
Cytokine ; 78: 44-6, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26618234

ABSTRACT

To better define the role of IL-17A in myasthenia gravis (MG), we assessed plasma concentrations in 69 adult patients with MG prior to initiation of immunosuppression and monitored their clinical course for the subsequent 2years with quantitative MG scores (QMGS) and Osserman classification. IL-17A was higher among patients than healthy control subjects. Early-onset women without thymoma had greater elevations of IL-17A. Logistic regression analysis indicated that the absence of thymoma rather than women gender or early-onset was the significant determinant associated with IL-17A elevation. Elevated IL-17A levels were associated with more severe MG. In summary, IL-17A has role in the pathogenesis of a subgroup of patients with early-onset women with MG with greater disease severity who are most likely to have thymic hyperplasia. This subgroup may be a target for IL-17 treatments, which are under development.


Subject(s)
Interleukin-17/blood , Myasthenia Gravis/blood , Adult , Female , Humans , Male , Myasthenia Gravis/complications , Myasthenia Gravis/immunology , Thymoma/complications , Thymus Hyperplasia/complications , Thymus Neoplasms/complications
14.
Exp Neurol ; 270: 3-10, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25743217

ABSTRACT

Antibodies against the muscle acetylcholine receptor (AChR) are the most common cause of myasthenia gravis (MG). Passive transfer of AChR antibodies from MG patients into animals reproduces key features of human disease, including antigenic modulation of the AChR, complement-mediated damage of the neuromuscular junction, and muscle weakness. Similarly, AChR antibodies generated by active immunization in experimental autoimmune MG models can subsequently be passively transferred to other animals and induce weakness. The passive transfer model is useful to test therapeutic strategies aimed at the effector mechanism of the autoantibodies. Here we summarize published and unpublished experience using the AChR passive transfer MG model in mice, rats and rhesus monkeys, and give recommendations for the design of preclinical studies in order to facilitate translation of positive and negative results to improve MG therapies.


Subject(s)
Myasthenia Gravis, Autoimmune, Experimental , Receptors, Cholinergic/immunology , Animals , Autoantibodies/immunology , Autoantigens/immunology , Guidelines as Topic , Humans , Macaca mulatta , Mice , Rats
15.
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
16.
J Immunol ; 193(5): 2127-34, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25057008

ABSTRACT

We recently demonstrated that hepatic stellate cells induce the differentiation of myeloid-derived suppressor cells (MDSCs) from myeloid progenitors. In this study, we found that adoptive transfer of these MDSCs effectively reversed disease progression in experimental autoimmune myasthenia gravis (EAMG), a T cell-dependent and B cell-mediated model for myasthenia gravis. In addition to ameliorated disease severity, MDSC-treated EAMG mice showed suppressed acetylcholine receptor (AChR)-specific T cell responses, decreased levels of serum anti-AChR IgGs, and reduced complement activation at the neuromuscular junctions. Incubating MDSCs with B cells activated by anti-IgM or anti-CD40 Abs inhibited the proliferation of these in vitro-activated B cells. Administering MDSCs into mice immunized with a T cell-independent Ag inhibited the Ag-specific Ab production in vivo. MDSCs directly inhibit B cells through multiple mechanisms, including PGE2, inducible NO synthase, and arginase. Interestingly, MDSC treatment in EAMG mice does not appear to significantly inhibit their immune response to a nonrelevant Ag, OVA. These results demonstrated that hepatic stellate cell-induced MDSCs concurrently suppress both T and B cell autoimmunity, leading to effective treatment of established EAMG, and that the MDSCs inhibit AChR-specific immune responses at least partially in an Ag-specific manner. These data suggest that MDSCs could be further developed as a novel approach to treating myasthenia gravis and, even more broadly, other diseases in which T and B cells are involved in pathogenesis.


Subject(s)
B-Lymphocytes/immunology , Myasthenia Gravis, Autoimmune, Experimental , Myeloid Cells , T-Lymphocytes/immunology , Animals , Autoantibodies/immunology , B-Lymphocytes/pathology , Dinoprostone/immunology , Hepatic Stellate Cells/immunology , Hepatic Stellate Cells/pathology , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Mice , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/pathology , Myasthenia Gravis, Autoimmune, Experimental/therapy , Myeloid Cells/immunology , Myeloid Cells/pathology , Myeloid Cells/transplantation , Receptors, Nicotinic/immunology , T-Lymphocytes/pathology
17.
PLoS One ; 9(7): e102635, 2014.
Article in English | MEDLINE | ID: mdl-25032816

ABSTRACT

Prednisone is often used for the treatment of autoimmune and inflammatory diseases but they suffer from variable therapeutic responses and significant adverse effects. Serum biological markers that are modulated by chronic corticosteroid use have not been identified. Myasthenia gravis is an autoimmune neuromuscular disorder caused by antibodies directed against proteins present at the post-synaptic surface of neuromuscular junction resulting in weakness. The patients with myasthenia gravis are primarily treated with prednisone. We analyzed the metabolomic profile of serum collected from patients prior to and after 12 weeks of prednisone treatment during a clinical trial. Our aim was to identify metabolites that may be treatment responsive and be evaluated in future studies as potential biomarkers of efficacy or adverse effects. Ultra-performance liquid chromatography coupled with electro-spray quadrupole time of flight mass spectrometry was used to obtain comparative metabolomic and lipidomic profile. Untargeted metabolic profiling of serum showed a clear distinction between pre- and post-treatment groups. Chronic prednisone treatment caused upregulation of membrane associated glycerophospholipids: phosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, 1, 2-diacyl-sn glycerol 3 phosphate and 1-Acyl-sn-glycero-3-phosphocholine. Arachidonic acid (AA) and AA derived pro-inflammatory eicosanoids such as 18-carboxy dinor leukotriene B4 and 15 hydroxyeicosatetraenoic acids were reduced. Perturbations in amino acid, carbohydrate, vitamin and lipid metabolism were observed. Chronic prednisone treatment caused increase in membrane associated glycerophospholipids, which may be associated with certain adverse effects. Decrease of AA and AA derived pro-inflammatory eicosanoids demonstrate that immunosuppression by corticosteroid is via suppression of pro-inflammatory pathways. The study identified metabolomic fingerprints that can now be validated as prednisone responsive biomarkers for the improvement in diagnostic accuracy and prediction of therapeutic outcome.


Subject(s)
Metabolome , Myasthenia Gravis/blood , Myasthenia Gravis/drug therapy , Prednisone/therapeutic use , Adult , Female , Humans , Lipid Metabolism , Male , Metabolomics , Myasthenia Gravis/metabolism , Neuromuscular Junction
18.
PLoS One ; 9(7): e102231, 2014.
Article in English | MEDLINE | ID: mdl-25050620

ABSTRACT

The mechanisms that underlie the development and maintenance of autoimmunity in myasthenia gravis are poorly understood. In this investigation, we evaluate the role of survivin, a member of the inhibitor of apoptosis protein family, in humans and in two animal models. We identified survivin expression in cells with B lymphocyte and plasma cells markers, and in the thymuses of patients with myasthenia gravis. A portion of survivin-expressing cells specifically bound a peptide derived from the alpha subunit of acetylcholine receptor indicating that they recognize the peptide. Thymuses of patients with myasthenia gravis had large numbers of survivin-positive cells with fewer cells in the thymuses of corticosteroid-treated patients. Application of a survivin vaccination strategy in mouse and rat models of myasthenia gravis demonstrated improved motor assessment, a reduction in acetylcholine receptor specific autoantibodies, and a retention of acetylcholine receptor at the neuromuscular junction, associated with marked reduction of survivin-expressing circulating CD20+ cells. These data strongly suggest that survivin expression in cells with lymphocyte and plasma cell markers occurs in patients with myasthenia gravis and in two animal models of myasthenia gravis. Survivin expression may be part of a mechanism that inhibits the apoptosis of autoreactive B cells in myasthenia gravis and other autoimmune disorders.


Subject(s)
Cell Survival , Inhibitor of Apoptosis Proteins/physiology , Myasthenia Gravis/metabolism , Adult , Aged , Animals , B-Lymphocytes/immunology , Case-Control Studies , Disease Models, Animal , Female , Humans , Leukocytes, Mononuclear/metabolism , Male , Mice, Inbred C57BL , Middle Aged , Myasthenia Gravis/immunology , Neuromuscular Junction/metabolism , Rats, Inbred Lew , Receptors, Cholinergic/immunology , Receptors, Cholinergic/metabolism , Survivin , Thymus Gland/metabolism , Young Adult
19.
Muscle Nerve ; 49(5): 749-56, 2014 May.
Article in English | MEDLINE | ID: mdl-24037951

ABSTRACT

INTRODUCTION: The site of pathology in myasthenia gravis (MG) is the neuromuscular junction (NMJ). Our goal was to determine the ability to direct complement inhibition to the NMJ. METHODS: A single-chain antibody directed against the alpha subunit of the acetylcholine receptor was synthesized (scFv-35) and coupled to decay-accelerating factor (DAF, scFv-35-DAF). scFv-35-DAF was tested in a passive model of experimentally acquired MG. RESULTS: Administration of scFv-35-DAF to mice deficient in intrinsic complement inhibitors produced no weakness despite confirmation of its localization to the NMJ and no evidence of tissue destruction related to complement activation. Rats with experimentally acquired MG treated with scFV-35-DAF showed less weakness and a reduction of complement deposition. CONCLUSIONS: We demonstrate a method to effectively target a therapeutic agent to the NMJ.


Subject(s)
CD55 Antigens/administration & dosage , Drug Delivery Systems/methods , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , Neuromuscular Junction , Receptors, Cholinergic , Single-Chain Antibodies/therapeutic use , Animals , Mice , Myasthenia Gravis, Autoimmune, Experimental/immunology , Rats , Single-Chain Antibodies/immunology
20.
Muscle Nerve ; 47(1): 33-40, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23042232

ABSTRACT

INTRODUCTION: Intrinsic mouse complement regulators influence the severity of passively induced experimental acquired myasthenia gravis (EAMG). To assess the potential influence of CD59b in the absence of CD59a background, we used the mCD59ab(-/-) mouse model to re-evaluate mCD59 in protecting the neuromuscular junction (NMJ). METHODS: EAMG was induced with monoclonal antibody to the acetylcholine receptor (AChR) in Daf1(-/-) , CD59ab(-/-) , Daf1(-/-) CD59ab(-/-) , and wild-type C57Bl/6 mice. Animals were monitored throughout the experiment. Diaphragms were analyzed for NMJ injury. RESULTS: Daf1(-/-) CD59ab(-/-) mice required euthanasia 24 hours after disease induction because of severe weakness. Histological assessment demonstrated reduced AChR density, simplification of synaptic folds, and disrupted mitochondria. CD59ab-deficient mice demonstrated mild weakness and reduction in weight after 24 hours. In contrast, Daf1(-/-) had more severe weakness at 60 hours. The NMJ of EAMG-induced Daf1(-/-) and CD59ab(-/-) mice demonstrated similar AChR density. CONCLUSION: NMJs of CD59 and DAF mice are protected from complement-mediated injury of passive EAMG.


Subject(s)
CD59 Antigens/immunology , Cell Membrane/immunology , Complement System Proteins/immunology , Myasthenia Gravis, Autoimmune, Experimental/immunology , Neuromuscular Junction/immunology , Animals , Cell Membrane/pathology , Mice , Mice, Transgenic , Myasthenia Gravis, Autoimmune, Experimental/pathology , Neuromuscular Junction/pathology , Receptors, Cholinergic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...