Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 16.171
Filter
1.
Front Public Health ; 12: 1421211, 2024.
Article in English | MEDLINE | ID: mdl-39257951

ABSTRACT

Objective: We investigated the risk factors associated with severe or critical Coronavirus disease 2019 (COVID-19) infection due to the Omicron variant in patients with myasthenia gravis (MG) and determined the potential effect of COVID-19 on myasthenic exacerbation during the Omicron pandemic. Methods: This retrospective study included 287 patients with MG in Tianjin, China. Clinical data of the patients were collected using electronic questionnaires, databases, and clinical records. Results: The overall infection rate was 84.7%. Advanced age, comorbidities, generalized phenotype, and MG instability were drivers of COVID-19 severity, and post-COVID-19 myasthenic exacerbation. The concurrent use of a steroid-sparing agent did not affect COVID-19 susceptibility or severity. It did lower the risk of myasthenic exacerbation after COVID-19 infection. Patients with severe COVID-19 experienced myasthenic exacerbation earlier than patients with non-severe infection (p < 0.001). The severity of COVID-19 (Hazards Ratio = 3.04, 95% CI: 1.41-6.54, p = 0.004) and the clinical phenotype (Hazards Ratio = 3.29, 95% CI: 1.63-6.63, p < 0.001) emerged as independent risk factors for early MG exacerbation. Conclusion: Generally, patients with MG appear to be susceptible to the Omicron strains. Immunotherapy for MG did not increase COVID-19 susceptibility or severity. We do not advocate an immediate cessation of ongoing immunosuppressive treatments once a COVID-19 infection is diagnosed. Instead, a judicious evaluation of the risks and benefits, tailored to each individual, is recommended.


Subject(s)
COVID-19 , Myasthenia Gravis , SARS-CoV-2 , Humans , COVID-19/epidemiology , COVID-19/complications , Retrospective Studies , Male , Female , Middle Aged , China/epidemiology , Adult , Risk Factors , Aged , Severity of Illness Index , Comorbidity
2.
BMC Neurol ; 24(1): 355, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39304804

ABSTRACT

BACKGROUND: Several cases of autoimmune disease onset after treatment for Cushing's syndrome have been reported. CASE PRESENTATION: Herein, we report a case of myasthenia gravis crisis in a 51-year-old woman 2 months after adrenalectomy for adrenal Cushing's syndrome accompanied by takotsubo cardiomyopathy. The resolution of excessive endogenous cortisol after adrenalectomy may have triggered the onset of previously latent myasthenia gravis. CONCLUSIONS: Observing the similarities in symptoms between myasthenia gravis and adrenal crisis, which can sometimes be challenging to differentiate, is essential. Moreover, the presence of takotsubo cardiomyopathy as a non-motor manifestation of myasthenic crisis must be noted.


Subject(s)
Adrenalectomy , Cushing Syndrome , Myasthenia Gravis , Takotsubo Cardiomyopathy , Humans , Myasthenia Gravis/complications , Myasthenia Gravis/diagnosis , Myasthenia Gravis/surgery , Takotsubo Cardiomyopathy/etiology , Female , Middle Aged , Adrenalectomy/adverse effects , Cushing Syndrome/surgery , Cushing Syndrome/etiology , Cushing Syndrome/complications
3.
Front Immunol ; 15: 1423547, 2024.
Article in English | MEDLINE | ID: mdl-39315106

ABSTRACT

Myasthenia gravis (MG) and idiopathic inflammatory myopathy (IIM) are autoimmune diseases of the nervous system, and their main clinical manifestation is muscle weakness. The concurrent presence of both conditions in the same patient is clinically rare and easily missed. Here, we report the case of a 74-year-old woman who went to the doctor with fluctuating weakness of the limbs and muscle pain. By analyzing the patient's history and the results of repeated frequency electrical stimulation, chest computed tomography, thigh muscle magnetic resonance imaging, serum antibody detection, lymph node biopsy, etc., she was finally diagnosed with MG-concomitant IIM with squamous cell carcinoma of the thymus. Acetylcholine receptor antibody, titin antibody, ryanodine receptor antibody, anti-JO-1 antibody, and Ro-52 antibody tests were positive. MG-concomitant IIM is often associated with thymoma. The immunopathology mechanism may be different from that of pure MG or IIM, which needs further research.


Subject(s)
Autoantibodies , Myasthenia Gravis , Myositis , Thymoma , Thymus Neoplasms , Humans , Myasthenia Gravis/immunology , Myasthenia Gravis/complications , Myasthenia Gravis/diagnosis , Female , Thymoma/complications , Thymoma/immunology , Thymoma/diagnosis , Aged , Myositis/immunology , Myositis/diagnosis , Myositis/complications , Autoantibodies/blood , Autoantibodies/immunology , Thymus Neoplasms/complications , Thymus Neoplasms/immunology , Thymus Neoplasms/diagnosis
4.
Allergol Immunopathol (Madr) ; 52(5): 59-64, 2024.
Article in English | MEDLINE | ID: mdl-39278852

ABSTRACT

Acquired myasthenia (AM), a debilitating autoimmune disease, is typically characterized by skeletal muscle fatigue and weakness. Despite advances in myasthenia gravis treatment, current approaches remain unsatisfactory and many result in unexpected side effects. Traditional Chinese medicine has shown great potential in the treatment of myasthenia gravis, including relieving myasthenic symptoms, improving patients' quality of life, and reducing Western medicine side effects. This study investigates the protective effects and mechanism of BZYQD in mice with acquired myasthenia. BZYQD alleviates the reduced grip strength and increased expression of MAFbx and MuRF-1 in mice with acquired myasthenia. It also reduces levels of pro-inflammatory factors IL-1ß, IL-6, and TNF-α in the mouse serum. In addition, BZYQD reduces ROS accumulation and the mitochondrial ROS production rate, while increasing ATP levels and mitochondrial membrane potential in mice with acquired myasthenia. Moreover, BZYQD decreases the expression of p-JAK2, p-STAT3, and p-AKT in the skeletal muscle of mice with acquired myasthenia. In summary, BZYQD reduces inflammation, enhances mitochondrial function, and regulates the JAK2/STAT3/AKT signaling pathway to treat acquired myasthenia.


Subject(s)
Drugs, Chinese Herbal , Janus Kinase 2 , Mitochondria , Proto-Oncogene Proteins c-akt , STAT3 Transcription Factor , Signal Transduction , Animals , Janus Kinase 2/metabolism , STAT3 Transcription Factor/metabolism , Drugs, Chinese Herbal/pharmacology , Mice , Signal Transduction/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Myasthenia Gravis/drug therapy , Myasthenia Gravis/immunology , Female , Inflammation/drug therapy , Inflammation/immunology , Cytokines/metabolism , Disease Models, Animal , Humans , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , Myasthenia Gravis, Autoimmune, Experimental/immunology , SKP Cullin F-Box Protein Ligases/metabolism , Tripartite Motif Proteins/metabolism , Reactive Oxygen Species/metabolism , Muscle Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
5.
Front Immunol ; 15: 1397916, 2024.
Article in English | MEDLINE | ID: mdl-39346912

ABSTRACT

Introduction: NK cells are dysfunctional in myasthenia gravis (MG), but the mechanism is unclear. This study aims to measure associations and underlying mechanisms between the NK cells and the development of MG. Methods: Twenty healthy controls (HCs) and 53 MG patients who did not receive glucocorticoids and immunosuppressants were collected. According to the Myasthenia Gravis Foundation of America (MGFA) classification, MG patients were categorized into MGFA I group (n = 18) and MGFA II-IV group (n = 35). Flow cytometry, cell sorting, ELISA, mRNA-sequencing, RT-qPCR, western blot, and cell culture experiments were performed to evaluate the regulatory mechanism of exhausted NK cells. Results: Peripheral NK cells in MGFA II-IV patients exhibit exhausted phenotypes than HCs, marked by the dramatic loss of total NK cells, CD56dimCD16- NK cells, elevated PD1 expression, reduced NKG2D expression, impaired cytotoxic activity (perforin, granzyme B, CD107a) and cytokine secretion (IFN-γ). Plasma IL-6 and IL-21 are elevated in MG patients and mainly derived from the aberrant expansion of monocytes and Tfh cells, respectively. IL-6/IL-21 cooperatively induced NK-cell exhausted signature via upregulating SOCS2 and inhibiting the phosphorylation of STAT5. SOCS2 siRNA and IL-2 supplement attenuated the IL-6/IL-21-mediated alteration of NK-cell phenotypes and function. Discussion: Inhibition of IL-6/IL-21/SOCS2/STAT5 pathway and recovery of NK-cell ability to inhibit autoimmunity may be a new direction in the treatment of MG.


Subject(s)
Killer Cells, Natural , Myasthenia Gravis , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Myasthenia Gravis/immunology , Male , Female , Adult , Middle Aged , Interleukins/metabolism , Interleukin-6/metabolism , Interleukin-6/blood , Signal Transduction , Case-Control Studies
6.
Proc Natl Acad Sci U S A ; 121(39): e2408324121, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39288173

ABSTRACT

Myasthenia gravis (MG) is a chronic and severe disease of the skeletal neuromuscular junction (NMJ) in which the effects of neurotransmitters are attenuated, leading to muscle weakness. In the most common forms of autoimmune MG, antibodies attack components of the postsynaptic membrane, including the acetylcholine receptor (AChR) or muscle-specific kinase (MuSK). MuSK, a master regulator of NMJ development, associates with the low-density lipoprotein-related receptor 4 (Lrp4) to form the signaling receptor for neuronal Agrin, a nerve-derived synaptic organizer. Pathogenic antibodies to MuSK interfere with binding between MuSK and Lrp4, inhibiting the differentiation and maintenance of the NMJ. MuSK MG can be debilitating and refractory to treatments that are effective for AChR MG. We show here that recombinant antibodies, derived from MuSK MG patients, cause severe neuromuscular disease in mice. The disease can be prevented by a MuSK agonist antibody, presented either prophylactically or after disease onset. These findings suggest a therapeutic alternative to generalized immunosuppression for treating MuSK MG by selectively and directly targeting the disease mechanism.


Subject(s)
Myasthenia Gravis , Neuromuscular Junction , Receptor Protein-Tyrosine Kinases , Receptors, Cholinergic , Animals , Receptor Protein-Tyrosine Kinases/immunology , Receptor Protein-Tyrosine Kinases/metabolism , Mice , Neuromuscular Junction/drug effects , Neuromuscular Junction/immunology , Receptors, Cholinergic/immunology , Receptors, Cholinergic/metabolism , Myasthenia Gravis/immunology , Myasthenia Gravis/drug therapy , Humans , LDL-Receptor Related Proteins/immunology , Autoantibodies/immunology , Female , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , Antibodies/immunology , Antibodies/pharmacology , Disease Models, Animal , Fatty Acids, Monounsaturated
7.
Cells ; 13(18)2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39329732

ABSTRACT

Myasthenia gravis (MG) is an antibody-mediated autoimmune disorder characterized by altered neuromuscular transmission, which causes weakness and fatigability in the skeletal muscles. The etiology of MG is complex, being associated with multiple genetic and environmental factors. Over recent years, progress has been made in understanding the immunological alterations implicated in the disease, but the exact pathogenesis still needs to be elucidated. A pathogenic interplay between innate immunity and autoimmunity contributes to the intra-thymic MG development. Epigenetic changes are critically involved in both innate and adaptive immune response regulation. They can act as (i) pathological factors besides genetic predisposition and (ii) co-factors contributing to disease phenotypes or patient-specific disease course/outcomes. This article reviews the role of non-coding RNAs (ncRNAs) as epigenetic factors implicated in MG. Particular attention is dedicated to microRNAs (miRNAs), whose expression is altered in MG patients' thymuses and circulating blood. The long ncRNA (lncRNA) contribution to MG, although not fully characterized yet, is also discussed. By summarizing the most recent and fast-growing findings on ncRNAs in MG, we highlight the therapeutic potential of these molecules for achieving immune regulation and their value as biomarkers for the development of personalized medicine approaches to improve disease care.


Subject(s)
Myasthenia Gravis , Precision Medicine , RNA, Untranslated , Humans , Myasthenia Gravis/immunology , Myasthenia Gravis/genetics , Myasthenia Gravis/therapy , Myasthenia Gravis/pathology , Precision Medicine/methods , RNA, Untranslated/genetics , Epigenesis, Genetic , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , Animals , Autoimmunity/genetics
8.
Neurol Neuroimmunol Neuroinflamm ; 11(6): e200313, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39250722

ABSTRACT

BACKGROUND AND OBJECTIVES: In this retrospective longitudinal study, we aimed at exploring the role of (a) MuSK-immunoglobulin G (IgG) levels, (b) predominant MuSK-IgG subclasses, and (c) antibody affinity as candidate biomarkers of severity and outcomes in MuSK-MG, using and comparing different antibody testing techniques. METHODS: Total MuSK-IgGs were quantified with radioimmunoassay (RIA), ELISA, flow cytometry, and cell-based assay (CBA) serial dilutions using HEK293 cells transfected with MuSK-eGFP. MuSK-IgG subclasses were measured by flow cytometry. SAffCon assay was used for determining MuSK-IgG affinity. RESULTS: Forty-three serum samples were obtained at different time points from 20 patients with MuSK-MG (median age at onset: 48 years, interquartile range = 27.5-72.5; women, 16/20), with 9 of 20 (45%) treated with rituximab. A strong correlation between MuSK-IgG levels measured by flow cytometry and RIA titers was found (rs = 0.74, 95% CI 0.41-0.89, p = 0.0003), as well as a moderate correlation between CBA end-point titers and RIA titers (rs = 0.47, 95% CI 0.01-0.77, p = 0.0414). A significant correlation was found between MuSK-IgG flow cytometry levels and disease severity (rs = 0.39, 95% CI 0.06-0.64, p = 0.0175; mixed-effects model estimate: 2.296e-06, std. error: 1.024e-06, t = 2.243, p = 0.032). In individual patients, clinical improvement was associated with decrease in MuSK-IgG levels, as measured by either flow cytometry or CBA end-point titration. In all samples, MuSK-IgG4 was the most frequent isotype (mean ± SD: 90.95% ± 13.89). A significant reduction of MuSK-IgG4 and, to a lesser extent, of MuSK-IgG2, was seen in patients with favorable clinical outcomes. A similar trend was confirmed in the subgroup of rituximab-treated patients. In a single patient, MuSK-IgG affinity increased during symptom exacerbation (KD values: 62 nM vs 0.6 nM) while total MuSK-IgG and IgG4 levels remained stable, suggesting that affinity maturation may be a driver of clinical worsening. DISCUSSION: Our data support the quantification of MuSK antibodies by flow cytometry. Through a multimodal investigational approach, we showed that total MuSK-IgG levels, MuSK-IgG4 and MuSK-IgG2 levels, and MuSK-IgG affinity may represent promising biomarkers of disease outcomes in MuSK-MG.


Subject(s)
Biomarkers , Immunoglobulin G , Myasthenia Gravis , Receptors, Cholinergic , Humans , Female , Male , Middle Aged , Myasthenia Gravis/blood , Myasthenia Gravis/immunology , Myasthenia Gravis/drug therapy , Myasthenia Gravis/diagnosis , Adult , Retrospective Studies , Aged , Immunoglobulin G/blood , Biomarkers/blood , Receptors, Cholinergic/immunology , Longitudinal Studies , Autoantibodies/blood , Receptor Protein-Tyrosine Kinases/immunology , Receptor Protein-Tyrosine Kinases/blood , HEK293 Cells , Rituximab/pharmacology , Immunologic Factors/pharmacology
9.
Vestn Oftalmol ; 140(4): 86-91, 2024.
Article in Russian | MEDLINE | ID: mdl-39254395

ABSTRACT

Myasthenia gravis is an autoimmune disease characterized by muscle weakness and pathological fatigue due to autoaggressive phenomena with the formation of antibodies directed against various structures of the neuromuscular synapse. In most patients, the disease begins with the involvement of extraocular muscles, presenting with symptoms such as intermittent ptosis of the upper eyelid and/or binocular diplopia. In 15% of cases, clinical manifestations are limited to impairment of the levator palpebrae superioris and extraocular muscles, characteristic of the ocular form of myasthenia gravis. Specialists often encounter challenges in diagnosing this form, as serological and electrophysiological studies may be uninformative, necessitating diagnosis based on patient history and clinical picture. This literature review outlines the key aspects of the pathogenesis, clinical manifestations, methods of diagnosis and treatment of ocular myasthenia gravis.


Subject(s)
Myasthenia Gravis , Oculomotor Muscles , Myasthenia Gravis/diagnosis , Myasthenia Gravis/physiopathology , Myasthenia Gravis/therapy , Myasthenia Gravis/complications , Humans , Oculomotor Muscles/physiopathology , Diagnosis, Differential
10.
Virulence ; 15(1): 2404225, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39267271

ABSTRACT

The THαß host immunological pathway contributes to the response to infectious particles (viruses and prions). Furthermore, there is increasing evidence for associations between autoimmune diseases, and particularly type 2 hypersensitivity disorders, and the THαß immune response. For example, patients with systemic lupus erythematosus often produce anti-double stranded DNA antibodies and anti-nuclear antibodies and show elevated levels of type 1 interferons, type 3 interferons, interleukin-10, IgG1, and IgA1 throughout the disease course. These cytokines and antibody isotypes are associated with the THαß host immunological pathway. Similarly, the type 2 hypersensitivity disorders myasthenia gravis, Graves' disease, graft-versus-host disease, autoimmune hemolytic anemia, immune thrombocytopenia, dermatomyositis, and Sjögren's syndrome have also been linked to the THαß pathway. Considering the potential associations between these diseases and dysregulated THαß immune responses, therapeutic strategies such as anti-interleukin-10 or anti-interferon α/ß could be explored for effective management.


Subject(s)
Lupus Erythematosus, Systemic , Humans , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/complications , Sjogren's Syndrome/immunology , Graft vs Host Disease/immunology , Autoimmune Diseases/immunology , Cytokines/immunology , Myasthenia Gravis/immunology , Anemia, Hemolytic, Autoimmune/immunology , Graves Disease/immunology , Graves Disease/complications , Dermatomyositis/immunology
11.
J Cardiothorac Surg ; 19(1): 510, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39227955

ABSTRACT

BACKGROUND: Myasthenia gravis is an autoimmune disease with high prevalence of thymus disorders, in which, thymectomy is considered one of the therapeutic approaches in improving the patients' clinical outcomes. Today, thoracoscopic thymectomy has received significant attention than the classic transsternal approach due to fewer complication. Therefore, this study was designed with the aim of investigating the therapeutic outcomes of thymectomy in patients with myasthenia gravis in the Afzalipour Hospital of Kerman between 2011 and 2021. METHODS: The current study is a descriptive analytical study on patients with myasthenia gravis who underwent surgical thymectomy within 2011-2021. Demographic and clinical characteristics of patients from the time of operation to three years of follow-up were extracted and recorded from clinical records or by phone calls. Data were analyzed using SPSS software. RESULTS: The data of 70 patients who underwent surgical thymectomy were analyzed. Thymectomy caused a significant reduction in the severity of the disease according to the Osserman classification (P = 0.001). It also significantly reduced the use of corticosteroids (P = 0.001) and IVIG (P = 0.015) compared to the time before the surgery. Sixty-two patients (88.57%) needed to take less medicine than before surgery. Left VATS was associated with less post-operative severity of the disease (P = 0.023). There were only two deaths during the follow-up period. CONCLUSION: Overall, the findings of the present study demonstrated that thoracoscopic thymectomy is a useful surgical approach that leads to faster recovery, reducing the severity of the disease, need for medication, and complications in patients with myasthenia gravis, In comparison with the transsternal approach.


Subject(s)
Myasthenia Gravis , Thymectomy , Humans , Myasthenia Gravis/surgery , Thymectomy/methods , Male , Female , Adult , Middle Aged , Treatment Outcome , Retrospective Studies , Young Adult , Thoracoscopy/methods , Adolescent , Thoracic Surgery, Video-Assisted/methods , Follow-Up Studies
12.
Nervenarzt ; 95(10): 920-931, 2024 Oct.
Article in German | MEDLINE | ID: mdl-39302417

ABSTRACT

This article deals with peripheral neuroimmunological diseases and briefly outlines the currently most important aspects and treatment developments. Idiopathic inflammatory myopathies have different mechanisms of development, manifestations and prognoses. New classification systems and more specific treatment concepts have been developed. The IIMs include different subgroups. These entities can have specific autoantibodies. Diagnostically, a muscle biopsy is generally desirable for a precise diagnosis and is essential in unclear cases. Primary systemic vasculitides can be divided into different groups based on the predominant pattern of involvement, while secondary vasculitides and single organ vasculitides are also differentiated. Vasculitic myopathy cannot be equated with myositis and a reliable distinction is currently only possible by a muscle biopsy. Treatment concepts should be developed on an interdisciplinary basis. Chronic inflammatory demyelinating polyneuropathy is the most frequent immune-mediated neuropathy and is characterized by a predominant demyelination of the motor and sensory nerves. The disease course runs in phases or is progressive and leads to significant disability and reduction in quality of life, despite current standard treatment. Novel treatment approaches are currently undergoing clinical trials. Myasthenia gravis, with the leading symptom of exercise-induced muscle weakness, is caused by autoantibodies against structures of the neuromuscular endplate. Autoantibody testing is the most important pillar in the diagnosis and is now also increasingly guiding treatment decisions. Overall, peripheral neuroimmunological diseases represent a heterogeneous group. Increasing knowledge of the pathophysiology is the key to numerous developments in diagnostics and treatment, which could lead to far-reaching practical changes in the future.


Subject(s)
Peripheral Nervous System Diseases , Humans , Peripheral Nervous System Diseases/immunology , Peripheral Nervous System Diseases/diagnosis , Peripheral Nervous System Diseases/therapy , Peripheral Nervous System Diseases/physiopathology , Diagnosis, Differential , Myositis/diagnosis , Myositis/immunology , Myositis/therapy , Myositis/classification , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/diagnosis , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/therapy , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/immunology , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/physiopathology , Myasthenia Gravis/immunology , Myasthenia Gravis/diagnosis , Myasthenia Gravis/therapy , Myasthenia Gravis/classification , Autoantibodies/immunology , Vasculitis/diagnosis , Vasculitis/therapy , Vasculitis/immunology , Vasculitis/classification
13.
Neuromuscul Disord ; 43: 39-43, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39190980

ABSTRACT

The aim of this study was to evaluate clinical and serological differences between the ocular myasthenia gravis (oMG) and generalized MG (gMG). This study is a retrospective chart review, in which data was collected from patients fulfilling 2 of 3 diagnostic MG criteria (positive antibodies, evidence of neuromuscular transmission defect on neurophysiological examination, positive effect of pyridostigmine treatment). 350 patients were included and data concerning demographics and MG medical history were collected. Patients with oMG accounted for 15.7 % of the included patients. The two subgroups differed significantly in oMG having a later age at onset, lower AChR antibody-titers, longer doctor-to-diagnosis delay and less intensive MG treatment. Additionally, patients with oMG were faster at reaching a well-controlled disease state. Thymus pathology, number of antibody-positive (95.9 % of gMG and 94.5 % of oMG), sex, number of other autoimmune diseases and delay before drug stability did not differ between oMG and gMG. In conclusion, oMG is presumably a milder form of gMG characterized by lower AChR antibody-titers, a milder phenotype, and a quicker response to a less aggressive treatment. But otherwise, oMG and gMG show very similar characteristics, including the same frequency of positive AChR antibodies, which seems new compared to previous reports.


Subject(s)
Autoantibodies , Myasthenia Gravis , Receptors, Cholinergic , Humans , Myasthenia Gravis/diagnosis , Myasthenia Gravis/blood , Myasthenia Gravis/immunology , Female , Male , Receptors, Cholinergic/immunology , Retrospective Studies , Middle Aged , Adult , Autoantibodies/blood , Aged , Young Adult , Age of Onset , Adolescent
14.
J Neuroimmunol ; 395: 578441, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39216158

ABSTRACT

INTRODUCTION: Dendritic cells (DCs) are crucial to form ectopic germinal centers (GCs) in the hyperplastic thymus (HT), which are typically found in anti-acetylcholine receptor autoantibody-positive myasthenia gravis (MG) patients. However, the characteristics of such DCs in the HT and their roles in thymic hyperplasia formation remain unclear. METHODS: We collected thymic tissue from MG patients and patients who underwent cardiac surgery. The tissues were cut into sections for immunohistochemistry and immunofluorescence or digested into a single cell suspension for flow cytometry. RESULTS: In addition to formation of ectopic GCs, we found that the proportion of the medulla in the thymic parenchyma was higher than that in the cortex (areacortex/areamedulla, 1.279 vs. 0.6576) in the HT of MG patients. The density of conventional dendritic cells (cDCs) in the HT was 131 ± 64.36 per mm2, whereas in normal thymic tissue, the density was 59.17 ± 22.54 per mm2. The more abundant cDCs expressed co-stimulatory molecules (CD80 and CD86) strongly. Moreover, the more abundant subset was mainly CD141+ DCs (cDC1s), accounting for an increase from 15% to 29%. However, these increased cDC1s appeared to be unrelated to Hassall's corpuscles and ectopic GCs. CONCLUSION: Thymic hyperplasia in MG patients is manifested as an increase in the proportion of the thymic medulla accompanied by increases in the density and functional activation as well as changes in the subset composition of cDCs.


Subject(s)
Dendritic Cells , Myasthenia Gravis , Thymus Gland , Thymus Hyperplasia , Humans , Myasthenia Gravis/pathology , Myasthenia Gravis/immunology , Dendritic Cells/pathology , Male , Female , Adult , Middle Aged , Thymus Hyperplasia/pathology , Thymus Gland/pathology , Thymus Gland/immunology , Aged , Young Adult , Hyperplasia/pathology , Adolescent
15.
J Neuroimmunol ; 395: 578431, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39142025

ABSTRACT

Efgartigimod was the first-in-class neonatal Fc receptor antagonist approved for the treatment of acetylcholine receptor antibody positive (AChR+), Myasthenia Gravis Foundation of America (MGFA) Class II-IV generalized myasthenia gravis (gMG) patients. As a novel therapy, the clinical experiences are still lacking, especially for the use of efgartigimod in manifest and impending myasthenic crisis (IMC). We reported three AChR+, gMG patients, two with myasthenic crisis (MC) and one with IMC, treated with efgartigimod. MGFA class, MG-Activity of Daily Living score (MG-ADL), Quantitative MG score (QMG), and Muscle Research Council sum score (MRC), concentration of anti-AChR antibody, IgG, globulin, and albumin, subsets of T and B lymphocyte were evaluated or measured before, during and after efgartigimod treatment. All patients showed fast and robust response to efgartigimod with marked improvement in MGFA, MG-ADL, QMG, and MRC scores. Patient 1 did not respond effectively to IVIg but was successfully rescued by add-on efgartigimod. She extubated at 7 days after the first infusion and got rid of NIV after 14-days treatment. Patient 2 and patient 3 directly used efgartigimod when symptoms were not ameliorated by adjusting of oral drugs. Patient 2 wean from BiPAP at seven days after the first infusion. Patient 3 in IMC status, overcame the severe dysphagia at three days after the first infusion. Clinical symptoms continued to improve 1-2 weeks after discharge. Concentration of anti-AChR antibody, IgG and globulin were remarkably reduced by efgartigimod treatment. Our study supported that efgartigimod could act as a fast-acting rescue therapy for patients with MC or IMC. Larger studies from multicenter are required to provide further evidence.


Subject(s)
Antibodies, Monoclonal, Humanized , Myasthenia Gravis , Adult , Aged , Female , Humans , Male , Middle Aged , Myasthenia Gravis/drug therapy , Antibodies, Monoclonal, Humanized/therapeutic use
16.
Adv Rheumatol ; 64(1): 61, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39169436

ABSTRACT

Bruton's tyrosine kinase (BTK), a nonreceptor tyrosine kinase, plays a remarkable role in the transmission and amplification of extracellular signals to intracellular signaling pathways. Various types of cells use the BTK pathway to communicate, including hematopoietic cells particularly B cells and T cells. The BTK pathway plays a role in controlling the proliferation, survival, and functions of B cells as well as other myeloid cells. First, second, and third-generation BTK inhibitors are currently being evaluated for the treatment of immune-mediated diseases in addition to B cell malignancies. In this article, the available evidence on the action mechanisms of BTK inhibitors is reviewed. Then, the most recent data obtained from preclinical studies and ongoing clinical trials for the treatment of autoimmune diseases, such as pemphigus vulgaris, pemphigus foliaceus, bullous pemphigoid, systemic lupus erythematosus, Sjögren's disease, rheumatoid arthritis, systemic sclerosis, multiple sclerosis, myasthenia gravis, and inflammatory diseases such as psoriasis, chronic spontaneous urticaria, atopic dermatitis, and asthma are discussed. In addition, adverse effects and complications associated with BTK inhibitors as well as factors predisposing patients to BTK inhibitors complications are discussed.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase , Autoimmune Diseases , Pemphigus , Protein Kinase Inhibitors , Signal Transduction , Humans , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Autoimmune Diseases/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pemphigus/drug therapy , Pemphigus/immunology , Pyrimidines/therapeutic use , Piperidines/therapeutic use , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Myasthenia Gravis/drug therapy , Multiple Sclerosis/drug therapy , Pyrazoles/therapeutic use , Pyrazoles/pharmacology , Nitriles/therapeutic use , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Adenine/analogs & derivatives , Adenine/therapeutic use , Asthma/drug therapy , B-Lymphocytes/immunology , Sjogren's Syndrome/drug therapy , Psoriasis/drug therapy , Psoriasis/immunology , Scleroderma, Systemic/drug therapy , Protein-Tyrosine Kinases/antagonists & inhibitors , Benzamides , Imidazoles , Pyrazines
17.
Front Immunol ; 15: 1409480, 2024.
Article in English | MEDLINE | ID: mdl-39148733

ABSTRACT

Background: Efgartigimod (Efgartigimod alpha fcab, Vyvgart™) is a pioneering neonatal Fc receptor (FcRn) antagonist for the treatment of severe autoimmune diseases mediated by pathogenic immunoglobulin G (IgG) autoantibodies, including myasthenia gravis (MG). It is a well-tolerated drug with minor side effects, such as headache and upper respiratory (lung) and urinary tract infections. Here, we present a case of Kaposi's varicelliform eruption (KVE) and herpetic conjunctivitis related to efgartigimod in a 60-year-old patient with ocular MG (OMG). Case description: A 60-year-old Chinese male suffered from acetylcholine receptor antibody positive (AChR Ab+) OMG for 8 years. During this period, he underwent first-line treatment with systemic corticosteroids, cyclosporine, cyclophosphamide, and so on, but had poor symptom improvement. On the recommendation of his attending neurologist, he received one cycle of intravenous efgartigimod (10mg/kg, once weekly for 4 weeks). The patient experienced fever, widespread painful blisters, and edema on the face on the third day after his last intravenous infusion. The patient also complained of increased secretions and a foreign body sensation in both eyes. Laboratory tests confirmed infection with herpes simplex virus (HSV). A diagnosis of efgartigimod-associated KVE and herpetic conjunctivitis was made. After intravenous administration (5mg/kg, 3 times a day, every 8 hours) for 10 days, the patient was cured without residual complications. Conclusions: This case is the first report of a patient with KVE and herpetic conjunctivitis related to efgartigimod in PubMed. This is rare and unusual. Clinicians should be alert to the rare symptoms related to efgartigimod.


Subject(s)
Kaposi Varicelliform Eruption , Myasthenia Gravis , Humans , Male , Middle Aged , Myasthenia Gravis/drug therapy , Myasthenia Gravis/chemically induced , Myasthenia Gravis/immunology , Myasthenia Gravis/diagnosis , Kaposi Varicelliform Eruption/drug therapy , Herpes Simplex/drug therapy , Herpes Simplex/diagnosis , Herpes Simplex/immunology , Conjunctivitis, Viral/drug therapy , Conjunctivitis, Viral/diagnosis
18.
J Neuroinflammation ; 21(1): 197, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39113081

ABSTRACT

BACKGROUND: Myasthenia gravis (MG) is an autoimmune disease characterized by pathogenic antibodies that target structures of the neuromuscular junction. The evidence suggests that the regulation of long noncoding RNAs (lncRNAs) that is mediated by transcription factors (TFs) plays a key role in the pathophysiology of MG. Nevertheless, the detailed molecular mechanisms of lncRNAs in MG remain largely undetermined. METHODS: Using microarray analysis, we analyzed the lncRNA levels in MG. By bioinformatics analysis, LINC01566 was found to potentially play an important role in MG. First, qRT‒PCR was performed to verify the LINC1566 expressions in MG patients. Then, fluorescence in situ hybridization was conducted to determine the localization of LINC01566 in CD4 + T cells. Finally, the impact of LINC01566 knockdown or overexpression on CD4 + T-cell function was also analyzed using flow cytometry and CCK-8 assay. A dual-luciferase reporter assay was used to validate the binding of the TF FOSL1 to the LINC01566 promoter. RESULTS: Based on the lncRNA microarray and differential expression analyses, we identified 563 differentially expressed (DE) lncRNAs, 450 DE mRNAs and 19 DE TFs in MG. We then constructed a lncRNA-TF-mRNA network. Through network analysis, we found that LINC01566 may play a crucial role in MG by regulating T-cell-related pathways. Further experiments indicated that LINC01566 is expressed at low levels in MG patients. Functionally, LINC01566 is primarily distributed in the nucleus and can facilitate CD4 + T-cell apoptosis and inhibit cell proliferation. Mechanistically, we hypothesized that LINC01566 may negatively regulate the expressions of DUSP3, CCR2, FADD, SIRPB1, LGALS3 and SIRPB1, which are involved in the T-cell activation pathway, to further influence the cellular proliferation and apoptosis in MG. Moreover, we found that the effect of LINC01566 on CD4 + T cells in MG was mediated by the TF FOSL1, and in vitro experiments indicated that FOSL1 can bind to the promoter region of LINC01566. CONCLUSIONS: In summary, our research revealed the protective roles of LINC01566 in clinical samples and cellular experiments, illustrating the potential roles and mechanism by which FOSL1/LINC01566 negatively regulates CD4 + T-cell activation in MG.


Subject(s)
CD4-Positive T-Lymphocytes , Lymphocyte Activation , Myasthenia Gravis , Proto-Oncogene Proteins c-fos , RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Myasthenia Gravis/metabolism , Myasthenia Gravis/immunology , Myasthenia Gravis/genetics , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , Proto-Oncogene Proteins c-fos/metabolism , Female , Male , Middle Aged , Adult
19.
J Neurol Sci ; 464: 123154, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39142082

ABSTRACT

INTRODUCTION/AIMS: The common presentations of statin intolerance are muscle-specific symptoms. Although statins are one type of drug reported to cause myasthenic worsening, myasthenic worsening has not been recognized as statin intolerance. The purpose of the present study is to investigate in a large cohort the safety profiles of statins in patients with myasthenia gravis (MG). METHODS: A total of 1710 consecutive patients with MG who visited sites associated with the Japan MG registry 2021 group between April and October 2021 were reviewed. Statin-associated myasthenic worsening was defined as worsening of any myasthenic symptoms on statin use and improvement of the symptom by stopping the statin or by undertaking additional treatment with patient and doctor confirmation. RESULTS: Among the 400 patients who used statins, 8 (2%) patients experienced statin intolerance and 6 (1.5%) patients experienced myasthenic worsening. No patients developed MG on the statin. Ptosis was a main symptom of myasthenic worsening in 4 (67%) patients. Atorvastatin was used in all patients with statin-associated myasthenic worsening. The symptoms of statin intolerance and statin-associated myasthenic worsening were improved within 2 months and 3 months, respectively, in all patients by cessation of statin use. DISCUSSION: Regarding statin-associated myasthenic worsening, prevalence was low, and severity was mild; with cessation of statin use, symptoms improved within a few months, and outcomes were generally good. Although statins can be used in MG patients with little concern, statin-associated myasthenic worsening should be noted in addition to the classical statin intolerance associated with statin use.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Myasthenia Gravis , Humans , Myasthenia Gravis/drug therapy , Myasthenia Gravis/chemically induced , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Male , Female , Aged , Middle Aged , Disease Progression , Registries , Aged, 80 and over , Adult , Japan/epidemiology , Cohort Studies , Atorvastatin/adverse effects , Atorvastatin/therapeutic use
20.
Sci Rep ; 14(1): 19523, 2024 08 22.
Article in English | MEDLINE | ID: mdl-39174610

ABSTRACT

Mechanical ventilation in myasthenic crisis is not standardized and is at high risk of failure. We investigated liberation from mechanical ventilation during myasthenic crisis using a prolonged spontaneous breathing trials (SBT) and sequential pulmonary function tests (PFT). In this retrospective monocenter study, we included patients admitted for a first episode of myasthenic crisis between January 2001 and January 2018. The primary outcome was the incidence of weaning failure upon first extubation in our cohort of patients with MC. Secondary objectives were to determine risk factors and outcome associated with weaning failure upon first extubation in MC. We also compared the characteristics of patients with prolonged weaning. 126 episodes of MC were analyzed. Patient's age was 64 [42-76] years with 72/126 (56.5%) being women. The median delay between weaning initiation and first extubation was 6 [3-10] days and the median total length of MV was 14 [10-23] days. 118/126 (93.7%) patients underwent prolonged SBT of 8 h or more prior to first extubation. The overall weaning failure rate was 18/126 (14.3%). Extubation was more often successful when the factor precipitating the myasthenic crisis was identified (86/108 (79.6%) vs. 8/18 (44.4%); p = 0.004), whereas PFT was similar in failure or successes. Most weaning failures upon first extubation attempt (11/18; 61%) were attributed to an insufficient stabilization of myasthenia gravis. Duration of mechanical ventilation, an infectious trigger and maximal inspiratory pressure upon intubation were independent risk factors for prolonged weaning. In myasthenic crisis, a standardized protocol including prolonged SBT and respiratory function tests might improve the success of first extubation without prolonging mechanical ventilation. The results of this single center study warrant further evaluation in interventional trials.


Subject(s)
Myasthenia Gravis , Respiration, Artificial , Ventilator Weaning , Humans , Female , Male , Myasthenia Gravis/therapy , Myasthenia Gravis/physiopathology , Middle Aged , Retrospective Studies , Aged , Adult , Respiration, Artificial/methods , Respiration, Artificial/adverse effects , Respiratory Function Tests , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL