Abstract

This study is aimed at exploring the expression pattern and methylation level of G0S2 in the peripheral blood mononuclear cells (PBMCs) of myasthenia gravis (MG) patients with positive acetylcholine receptor (AChR) autoantibodies and revealing the relationship between the G0S2 methylation pattern and MG. The relationship between the NFAT family members and G0S2 was explored to reveal the regulatory mechanism of G0S2 in the pathogenesis and treatment of AChR MG. Moreover, we attempted to demonstrate the potential therapeutic mechanism of tacrolimus in AChR MG. The relative G0S2 expression level in the PBMCs of healthy people was compared with that in the PBMCs of AChR MG patients with quantitative real-time PCR (qRT-PCR). The methylation frequency of the G0S2 promoter was detected by bisulfite sequencing PCR (BSP) and pyrosequencing. A dual-luciferase reporter system was used to reveal the relationship between the G0S2 promoter and nuclear factor of activated T cells 5 (NFAT5). The qRT-PCR results showed that G0S2 expression was significantly upregulated in the B cells and CD8+ T cells of AChR MG patients but not in the CD4+ T cells, and these expression differences were significantly associated with a decrease in G0S2 methylation. NFAT5, which was speculated to bind to island 1 (p1) in the G0S2 promoter, may regulate the lymphocyte balance by regulating G0S2 gene expression but failed to affect the methylation of the G0S2 promoter. Tacrolimus therapy-induced methylation and overexpression of NFAT5 could significantly reduce the expression of G0S2 in AChR MG patients. The G0S2 gene was remarkably upregulated in the PBMCs of MG patients. NFAT5 may affect transcription initiation and downregulate G0S2 expression through p1 in the promoter, thus controlling G0S2 gene expression and regulating the lymphocyte balance. Therefore, G0S2 could be an immune regulatory factor in both AChR MG occurrence and treatment with tacrolimus.

Highlights

  • Myasthenia gravis (MG) is an autoimmune disease characterized by a transmission disorder of the neuromuscular junction

  • Compared with the mRNA microarray analysis results, the G0S2 gene showed a more significant upregulation in the peripheral blood of MG patients. quantitative real-time PCR (qRT-PCR) revealed that the expression level of G0S2 was sharply upregulated in the peripheral blood mononuclear cells (PBMCs) of MG patients (Figure 1)

  • The G0S2 gene was originally discovered in PBMCs and was associated with the cell cycle

Read more

Summary

Introduction

Myasthenia gravis (MG) is an autoimmune disease characterized by a transmission disorder of the neuromuscular junction. The molecular immunopathology of MG is due to the presence of circulating autoantibodies that target the acetylcholine receptor (AChR), muscle-specific tyrosine kinase (MuSK), or low-density lipoprotein (LDL) receptor-related protein 4 (LRP4). Patient-derived AChR, MuSK, and LRP4 autoantibodies in MG are demonstrably pathogenic (B cells in the pathophysiology of myasthenia gravis), and patients most often harbor only one of these autoantibody specificities. In approximately 75–85% of MG patients, AChR autoantibodies are detectable [1, 2]. Our understanding of MG immunopathology remains incomplete. It appears that the mechanism used by B cells for autoantibody production in AChR and MuSK MG differs, details on both are needed to understand the immunopathology that will guide the development of more effective therapies [3]

Objectives
Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call