Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Immunol ; 205(7): 1743-1751, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32839239

RESUMO

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.


Assuntos
Linfócitos B/metabolismo , Miastenia Gravis/imunologia , Survivina/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Anticorpos/metabolismo , Antígenos CD20/metabolismo , Autoantígenos/imunologia , Linfócitos B/imunologia , Modelos Animais de Doenças , Feminino , Humanos , Imunidade Humoral , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Peptídeos/imunologia , Adulto Jovem
2.
Cytokine ; 96: 279-285, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28599246

RESUMO

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.


Assuntos
Autoimunidade/imunologia , Interleucina-17/genética , Interleucina-17/fisiologia , Miastenia Gravis Autoimune Experimental/imunologia , Miastenia Gravis Autoimune Experimental/fisiopatologia , Animais , Autoanticorpos , Citocinas/imunologia , Modelos Animais de Doenças , Fatores de Transcrição Forkhead/genética , Humanos , Imunoglobulina G/sangue , Interleucina-17/deficiência , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-bcl-6/genética , Reação em Cadeia da Polimerase em Tempo Real , Índice de Gravidade de Doença , Linfócitos T Reguladores
3.
Cytokine ; 78: 44-6, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26618234

RESUMO

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.


Assuntos
Interleucina-17/sangue , Miastenia Gravis/sangue , Adulto , Feminino , Humanos , Masculino , Miastenia Gravis/complicações , Miastenia Gravis/imunologia , Timoma/complicações , Hiperplasia do Timo/complicações , Neoplasias do Timo/complicações
4.
J Immunol ; 193(5): 2127-34, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25057008

RESUMO

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.


Assuntos
Linfócitos B/imunologia , Miastenia Gravis Autoimune Experimental , Células Mieloides , Linfócitos T/imunologia , Animais , Autoanticorpos/imunologia , Linfócitos B/patologia , Dinoprostona/imunologia , Células Estreladas do Fígado/imunologia , Células Estreladas do Fígado/patologia , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Camundongos , Miastenia Gravis Autoimune Experimental/imunologia , Miastenia Gravis Autoimune Experimental/patologia , Miastenia Gravis Autoimune Experimental/terapia , Células Mieloides/imunologia , Células Mieloides/patologia , Células Mieloides/transplante , Receptores Nicotínicos/imunologia , Linfócitos T/patologia
5.
Muscle Nerve ; 49(5): 749-56, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24037951

RESUMO

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.


Assuntos
Antígenos CD55/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Miastenia Gravis Autoimune Experimental/tratamento farmacológico , Junção Neuromuscular , Receptores Colinérgicos , Anticorpos de Cadeia Única/uso terapêutico , Animais , Camundongos , Miastenia Gravis Autoimune Experimental/imunologia , Ratos , Anticorpos de Cadeia Única/imunologia
6.
J Clin Invest ; 134(12)2024 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-39105625

RESUMO

Myasthenia gravis (MG) stands as a prototypical antibody-mediated autoimmune disease: it is dependent on T cells and characterized by the presence of autoantibodies targeting proteins located on the postsynaptic surface of skeletal muscle, known as the neuromuscular junction. Patients with MG exhibit a spectrum of weakness, ranging from limited ocular muscle involvement to life-threatening respiratory failure. Recent decades have witnessed substantial progress in understanding the underlying pathophysiology, leading to the delineation of distinct subcategories within MG, including MG linked to AChR or MuSK antibodies as well as age-based distinction, thymoma-associated, and immune checkpoint inhibitor-induced MG. This heightened understanding has paved the way for the development of more precise and targeted therapeutic interventions. Notably, the FDA has recently approved therapeutic inhibitors of complement and the IgG receptor FcRn, a testament to our improved comprehension of autoantibody effector mechanisms in MG. In this Review, we delve into the various subgroups of MG, stratified by age, autoantibody type, and histology of the thymus with neoplasms. Furthermore, we explore both current and potential emerging therapeutic strategies, shedding light on the evolving landscape of MG treatment.


Assuntos
Autoanticorpos , Miastenia Gravis , Miastenia Gravis/imunologia , Miastenia Gravis/terapia , Miastenia Gravis/patologia , Humanos , Autoanticorpos/imunologia , Receptores Colinérgicos/imunologia , Timoma/imunologia , Timoma/patologia , Timoma/terapia , Antígenos de Histocompatibilidade Classe I/imunologia , Receptores Fc/imunologia , Receptores Proteína Tirosina Quinases/imunologia , Receptores Proteína Tirosina Quinases/antagonistas & inibidores
7.
Neurology ; 102(12): e209482, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38781559

RESUMO

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.


Assuntos
Miastenia Gravis , Timectomia , Timoma , Timo , Neoplasias do Timo , Humanos , Timectomia/efeitos adversos , Timectomia/métodos , Miastenia Gravis/cirurgia , Timo/cirurgia , Neoplasias do Timo/cirurgia , Neoplasias do Timo/complicações , Timoma/cirurgia , Timoma/complicações , Complicações Pós-Operatórias/etiologia , Doenças Autoimunes/cirurgia
8.
Muscle Nerve ; 47(1): 33-40, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23042232

RESUMO

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.


Assuntos
Antígenos CD59/imunologia , Membrana Celular/imunologia , Proteínas do Sistema Complemento/imunologia , Miastenia Gravis Autoimune Experimental/imunologia , Junção Neuromuscular/imunologia , Animais , Membrana Celular/patologia , Camundongos , Camundongos Transgênicos , Miastenia Gravis Autoimune Experimental/patologia , Junção Neuromuscular/patologia , Receptores Colinérgicos/imunologia
9.
Sci Immunol ; 8(85): eadj4913, 2023 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-37418544

RESUMO

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


Assuntos
Linfócitos B , Centro Germinativo , Ativação Linfocitária , Antígenos
10.
PLoS One ; 18(10): e0287654, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37816000

RESUMO

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.


Assuntos
Atividades Cotidianas , Miastenia Gravis , Humanos , Prednisona/efeitos adversos , Glucocorticoides/uso terapêutico , Miastenia Gravis/tratamento farmacológico , Terapia Combinada , Timectomia/métodos , Resultado do Tratamento
11.
Ann Neurol ; 65(1): 67-75, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19194881

RESUMO

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


Assuntos
Complemento C5/antagonistas & inibidores , Proteínas Inativadoras do Complemento/uso terapêutico , Miastenia Gravis Autoimune Experimental/prevenção & controle , Animais , Anticorpos/efeitos adversos , Proteínas de Artrópodes , Linhagem Celular Tumoral , Complemento C9/metabolismo , Proteínas Inativadoras do Complemento/imunologia , Proteínas Inativadoras do Complemento/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Imunoglobulina G/sangue , Imunoglobulina G/classificação , Proteínas de Insetos/imunologia , Proteínas de Insetos/uso terapêutico , Força Muscular/efeitos dos fármacos , Miastenia Gravis Autoimune Experimental/etiologia , Miastenia Gravis Autoimune Experimental/imunologia , Miastenia Gravis Autoimune Experimental/patologia , Junção Neuromuscular/efeitos dos fármacos , Junção Neuromuscular/metabolismo , Ratos , Ratos Endogâmicos Lew , Receptores Colinérgicos/imunologia , Rabdomiossarcoma/metabolismo , Rabdomiossarcoma/patologia , Proteínas e Peptídeos Salivares/sangue , Proteínas e Peptídeos Salivares/imunologia , Proteínas e Peptídeos Salivares/uso terapêutico , Índice de Gravidade de Doença , Fatores de Tempo , Redução de Peso/efeitos dos fármacos
12.
Mol Ther Methods Clin Dev ; 13: 484-492, 2019 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-31193726

RESUMO

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.

13.
Ann N Y Acad Sci ; 1132: 220-4, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18567871

RESUMO

Extraocular muscle (EOM) is susceptible to neuromuscular junction disorders, in particular, myasthenia gravis (MG). While EOM physiological characteristics and the ocular motor system requirements contribute to the propensity of ocular motor deficits observed among patients with MG, the authors propose that EOM have immunological features that place the muscles at risk for immune attack. Genomic profiling studies have demonstrated that genes associated with the immune response are differentially expressed in EOM, with particular differences in both classical and alternative complement-mediated immune response pathways. Intrinsic complement regulators are expressed at lower levels at rodent EOM neuromuscular junctions, which would put them at risk for the complement-mediated injury that occurs in MG. In fact, systemic C inhibition in experimental autoimmune MG (EAMG) induced by administration of acetylcholine receptor (AChR) antibodies or immunization with AChR will eliminate complement deposition at junctions of other skeletal muscle, but not EOM. Also, EOM junctions have greater injury in active and passive EAMG by several measures, suggesting that the lack of complement inhibition puts the EOM at risk. Among ocular myasthenia patients, serum AChR antibody levels are low, which would support the concept that EOM junctions are more susceptible to antibody injury than are other junctions. These observations suggest that complement inhibitory therapies may prove to be particularly effective in treatment of ocular myasthenia.


Assuntos
Miastenia Gravis/imunologia , Músculos Oculomotores/imunologia , Animais , Proteínas do Sistema Complemento/imunologia , Proteínas do Sistema Complemento/metabolismo , Suscetibilidade a Doenças/imunologia , Suscetibilidade a Doenças/patologia , Humanos , Miastenia Gravis/genética , Miastenia Gravis/patologia , Músculos Oculomotores/anatomia & histologia
14.
Ann N Y Acad Sci ; 1413(1): 136-142, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29356015

RESUMO

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.


Assuntos
Autoanticorpos/imunologia , Proteínas Inativadoras do Complemento/farmacologia , Proteínas do Sistema Complemento/imunologia , Miastenia Gravis Autoimune Experimental/tratamento farmacológico , Miastenia Gravis Autoimune Experimental/imunologia , Receptores Proteína Tirosina Quinases/imunologia , Receptores Colinérgicos/imunologia , Animais , Proteínas do Sistema Complemento/genética , Modelos Animais de Doenças , Imunização Passiva/métodos , Imunoglobulina G/imunologia , Macaca mulatta , Camundongos , Camundongos Endogâmicos C57BL , Miastenia Gravis Autoimune Experimental/patologia , Ratos , Ratos Endogâmicos Lew
16.
PLoS One ; 13(10): e0205464, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30308012

RESUMO

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.


Assuntos
Perfilação da Expressão Gênica/métodos , Centro Germinativo/química , MicroRNAs/genética , Miastenia Gravis/genética , Proteínas RGS/genética , Adulto , Linhagem Celular , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Pessoa de Meia-Idade , RNA Mensageiro/genética , Transdução de Sinais , Biologia de Sistemas/métodos , Timo , Adulto Jovem
17.
Front Neurol ; 8: 230, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28620344

RESUMO

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.

18.
Neurologist ; 12(5): 231-9, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16990734

RESUMO

BACKGROUND: Although myasthenia gravis (MG) is often considered the best-understood autoimmune disorder and effective treatments have controlled life-threatening complications, the pathogenesis of ocular myasthenia (OM) remains enigmatic, and its clinical consequences offer therapeutic challenges. REVIEW SUMMARY: About half of patients with MG present with visual complaints of droopy eyelids or double vision, and many will remain with purely ocular muscle weakness without generalized weakness, defined as OM. OM may be confused with disorders of the brainstem, ocular motor nerves, and eye muscles. Frustrating for the clinician, confirmatory tests such as the edrophonium test, serum acetylcholine receptor antibodies, and standard electrodiagnostic evaluations may fail to positively identify the clinical suspicion of OM. Patients may derive relief from nonpharmacologic interventions and cholinesterase inhibitors, but most will desire better symptom control with corticosteroids or need other immunosuppression. Early corticosteroid therapy may reduce the probability of generalization of the disease. The reasons for ocular muscle involvement by OM include physiologic and cellular properties of the ocular motor system and the unique immunology of OM, which, when better understood, will lead to novel treatments. CONCLUSIONS: OM is a challenging disorder for the clinician and scientist, with both learning from the other for the betterment of the patient. The future requires answers to why the ocular muscles are so frequently involved by MG, whether the generalization of the disease may be limited by early corticosteroid treatment, and what treatment options may be developed which will improve symptoms without long-term complications.


Assuntos
Miastenia Gravis , Músculos Oculomotores/fisiopatologia , Humanos , Miastenia Gravis/diagnóstico , Miastenia Gravis/etiologia , Miastenia Gravis/terapia
19.
Front Physiol ; 7: 524, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27891095

RESUMO

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.

20.
FASEB J ; 17(8): 893-5, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12670877

RESUMO

Extraocular muscle (EOM) is spared in Duchenne muscular dystrophy. Here, we tested putative EOM sparing mechanisms predicted from existing dystrophinopathy models. Data show that mdx mouse EOM contains dystrophin-glycoprotein complex (DGC)-competent and DGC-deficient myofibers distributed in a fiber type-specific pattern. Up-regulation of a dystrophin homologue, utrophin, mediates selective DGC retention. Counter to the DGC mechanical hypothesis, an intact DGC is not a precondition for EOM sarcolemmal integrity, and active adaptation at the level of calcium homeostasis is not mechanistic in protection. A partial, fiber type-specific retention of antiischemic nitric oxide to vascular smooth muscle signaling is not a factor in EOM sparing, because mice deficient in dystrophin and alpha-syntrophin, which localizes neuronal nitric oxide synthase to the sarcolemma, have normal EOMs. Moreover, an alternative transmembrane protein, alpha7beta1 integrin, does not appear to substitute for the DGC in EOM. Finally, genomewide expression profiling showed that EOM does not actively adapt to dystrophinopathy but identified candidate genes for the constitutive protection of mdx EOM. Taken together, data emphasize the conditional nature of dystrophinopathy and the potential importance of nonmechanical DGC roles and support the hypothesis that broad, constitutive structural cell signaling, and/or biochemical differences between EOM and other skeletal muscles are determinants of differential disease responsiveness.


Assuntos
Distrofia Muscular Animal/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Músculos Oculomotores/metabolismo , Adaptação Fisiológica , Animais , Antígenos CD/genética , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio , Análise por Conglomerados , Proteínas do Citoesqueleto/metabolismo , Distrofina/deficiência , Distrofina/metabolismo , Expressão Gênica , Hemostasia , Imuno-Histoquímica , Cadeias alfa de Integrinas/genética , Proteínas de Membrana/deficiência , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/deficiência , Proteínas Musculares/metabolismo , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/fisiopatologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/fisiopatologia , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo I , Músculos Oculomotores/fisiopatologia , Análise de Sequência com Séries de Oligonucleotídeos , Sarcolema/metabolismo , Utrofina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA