...
首页> 外文期刊>Theranostics >ASIC1a induces synovial inflammation via the Casup2+/sup/NFATc3/ RANTES pathway
【24h】

ASIC1a induces synovial inflammation via the Casup2+/sup/NFATc3/ RANTES pathway

机译:ASIC1A通过Ca 2 + / sup> / nfatc3 / rantes路径诱​​导滑膜炎症

获取原文
           

摘要

Rationale: Synovial inflammation is one of the main pathological features of rheumatoid arthritis (RA) and is a key factor leading to the progression of RA. Understanding the regulatory mechanism of synovial inflammation is crucial for the treatment of RA. Acid-sensing ion channel 1a (ASIC1a) is an Hsup+/sup-gated cation channel that promotes the progression of RA, but the role of ASIC1a in synovial inflammation is unclear. This study aimed to investigate whether ASIC1a is involved in the synovial inflammation and explore the underlying mechanisms in vitro and in vivo . Methods: The expression of ASIC1a and nuclear factor of activated T cells (NFATs) were analyzed by Western blotting, immunofluorescence, and immunohistochemistry both in vitro and in vivo . The Casup2+/sup influx mediated by ASIC1a was detected by calcium imaging and flow cytometry. The role of ASIC1a in inflammation was studied in rats with adjuvant-induced arthritis (AA). Inflammatory cytokine profile was analyzed by protein chip in RA synovial fibroblasts (RASF) and verified by a magnetic multi-cytokine assay and ELISA. The NFATc3-regulated RANTES (Regulated upon activation, normal T cell expressed and secreted) gene transcription was investigated by ChIP-qPCR and dual-luciferase reporter assay. Results: The expression of ASIC1a was significantly increased in human RA synovial tissues and primary human RASF as well as in ankle synovium of AA rats. Activated ASIC1a mediated Casup2+/sup influx to increase [Casup2+/sup]i in RASF. The activation/overexpression of ASIC1a in RASF up-regulated the expression of inflammatory cytokines RANTES, sTNF RI, MIP-1a, IL-8, sTNF RII, and ICAM-1 among which RANTES was increased most remarkably. In vivo , ASIC1a promoted inflammation, synovial hyperplasia, articular cartilage, and bone destruction, leading to the progression of AA. Furthermore, activation of ASIC1a upregulated the nuclear translocation of NFATc3, which bound to RANTES promoter and directly regulated gene transcription to enhance RANTES expression. Conclusion: ASIC1a induces synovial inflammation, which leads to the progression of RA. Our study reveals a novel RA inflammation regulatory mechanism and indicates that ASIC1a might be a potential therapeutic target for RA.
机译:理由:滑膜炎症是类风湿性关节炎(RA)的主要病理特征之一,是导致RA进展的关键因素。了解滑膜炎症的调节机制对于RA治疗至关重要。酸感测离子通道1a(ASIC1a)是促进RA进展的H + / sup> - 阳离子通道,但ASIC1a在滑膜炎症中的作用尚不清楚。本研究旨在调查ASIC1A是否参与了滑膜炎症,并在体外和体内探讨了潜在机制。方法:通过蛋白质印迹,免疫荧光和免疫组织化学分析活性T细胞(NFAT)的ASIC1a和核因子的表达,体外和体内。通过钙成像和流式细胞术检测由ASIC1A介导的Ca 2 + / sup> inofum。在佐剂诱导的关节炎(AA)的大鼠中研究了ASIC1A在炎症中的作用。通过蛋白质芯片在Ra滑膜成纤维细胞(RASF)中分析炎症细胞因子谱,并通过磁性多细胞因子测定和ELISA验证。通过CHIP-QPCR和双荧光素酶报告结果研究了NFATC3调节的RANT(调节的激活,正常的T细胞表达和分泌的)基因转录。结果:人RA滑膜组织和原发性人RASF中的ASIC1a的表达显着增加,以及AA大鼠的踝臂。活化ASIC1a介导的Ca 2 + 流入,以增加Rasf中的[Ca 2 + ] i。 RASF中ASIC1a的激活/过度表达上调炎性细胞因子咆哮,STNF RI,MIP-1A,IL-8,STNF RII和ICAM-1中的表达,其中咆哮最显着增加。体内,ASIC1A促进炎症,滑膜增生,关节软骨和骨破坏,导致AA的进展。此外,ASIC1a的激活上调了NFATC3的核易位,其结合Rantes启动子并直接调节基因转录以增强咆哮的表达。结论:ASIC1A诱导滑膜炎症,从而导致RA的进展。我们的研究揭示了一种新的RA炎症调节机制,表明ASIC1A可能是RA的潜在治疗目标。

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
获取原文

客服邮箱:kefu@zhangqiaokeyan.com

京公网安备:11010802029741号 ICP备案号:京ICP备15016152号-6 六维联合信息科技 (北京) 有限公司©版权所有
  • 客服微信

  • 服务号