首页> 外文期刊>Cell death and differentiation >Immunoproteasome dysfunction augments alternative polarization of alveolar macrophages
【24h】

Immunoproteasome dysfunction augments alternative polarization of alveolar macrophages

机译:免疫蛋白酶体功能障碍增强了肺泡巨噬细胞的替代极化

获取原文
获取原文并翻译 | 示例
       

摘要

The proteasome is a central regulatory hub for intracellular signaling by degrading numerous signaling mediators. Immunoproteasomes are specialized types of proteasomes involved in shaping adaptive immune responses, but their role in innate immune signaling is still elusive. Here, we analyzed immunoproteasome function for polarization of alveolar macrophages, highly specialized tissue macrophages of the alveolar lung surface. Classical activation (M1 polarization) of primary alveolar macrophages by LPS/IFN gamma transcriptionally induced all three immunoproteasome subunits, low molecular mass protein 2 (LMP2), LMP7 and multicatalytic endopeptidase complex-like 1, which was accompanied by increased immunoproteasome activity in M1 cells. Deficiency of LMP7 had no effect on the LPS/IFN gamma-triggered M1 profile indicating that immunoproteasome function is dispensable for classical alveolar macrophage activation. In contrast, IL-4 triggered alternative (M2) activation of primary alveolar macrophages was accompanied by a transcriptionally independent amplified expression of LMP2 and LMP7 and an increase in immunoproteasome activity. Alveolar macrophages from LMP7 knockout mice disclosed a distorted M2 profile upon IL-4 stimulation as characterized by increased M2 marker gene expression and CCL17 cytokine release. Comparative transcriptome analysis revealed enrichment of IL-4-responsive genes and of genes involved in cellular response to defense, wounding and inflammation in LMP7-deficient alveolar macrophages indicating a distinct M2 inflammation resolving phenotype. Moreover, augmented M2 polarization was accompanied by amplified AKT/STAT6 activation and increased RNA and protein expression of the M2 master transcription factor interferon regulatory factor 4 in LMP7(-/-) alveolar macrophages. IL-13 stimulation of LMP7-deficient macrophages induced a similar M2-skewed profile indicative for augmented signaling via the IL-4 receptor alpha (IL4R alpha). IL4R alpha expression was generally elevated only on protein but not RNA level in LMP7(-/-) alveolar macrophages. Importantly, specific catalytic inhibition with an LMP7-specific proteasome inhibitor confirmed augmented IL-4-mediated M2 polarization of alveolar macrophages. Our results thus suggest a novel role of immunoproteasome function for regulating alternative activation of macrophages by limiting IL4R alpha expression and signaling.
机译:蛋白酶体是通过降解许多信号传导介质而用于细胞内信号传导的中央调节中心。免疫蛋白酶体是参与形成适应性免疫应答的特殊类型的蛋白酶体,但是它们在先天免疫信号传导中的作用仍然难以捉摸。在这里,我们分析了肺泡巨噬细胞,肺泡肺表面高度专门化的组织巨噬细胞极化的免疫蛋白酶体功能。 LPS / IFNγ对肺泡巨噬细胞经典激活(M1极化)的转录诱导了所有三个免疫蛋白酶体亚基,低分子量蛋白2(LMP2),LMP7和多催化内肽酶复合物样1,并伴随着M1细胞免疫蛋白酶体活性的增加。 LMP7的缺乏对LPS / IFNγ触发的M1谱没有影响,表明免疫蛋白酶体功能对于经典的肺泡巨噬细胞激活是必不可少的。相反,IL-4触发原发性肺泡巨噬细胞的替代(M2)激活伴随有LMP2和LMP7的转录独立扩增表达以及免疫蛋白酶体活性的增加。 LMP7基因敲除小鼠的肺泡巨噬细胞显示IL-4刺激后M2分布畸变,其特征在于M2标记基因表达增加和CCL17细胞因子释放。对比转录组分析显示,LMP7缺陷型肺泡巨噬细胞中IL-4反应基因和参与细胞对防御,创伤和炎症反应的基因富集,表明存在独特的M2炎症表型。此外,增强的M2极化伴随着放大的AKT / STAT6激活以及LMP7(-/-)肺泡巨噬细胞中M2主转录因子干扰素调节因子4的RNA和蛋白质表达增加。 IL-13对LMP7缺陷型巨噬细胞的刺激诱导了相似的M2偏斜轮廓,表明经由IL-4受体α(IL4Rα)的信号增强。在LMP7(-/-)肺泡巨噬细胞中,IL4Rα表达通常仅在蛋白质上升高,而在RNA水平上则不升高。重要的是,用LMP7特异性蛋白酶体抑制剂的特异性催化抑制作用证实了肺泡巨噬细胞的IL-4介导的M2极化增强。因此,我们的结果表明免疫蛋白酶体功能通过限制IL4Rα表达和信号传导来调节巨噬细胞的选择性激活的新作用。

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