...
首页> 外文期刊>Cell death & disease. >ATG5-mediated autophagy suppresses NF-κB signaling to limit epithelial inflammatory response to kidney injury
【24h】

ATG5-mediated autophagy suppresses NF-κB signaling to limit epithelial inflammatory response to kidney injury

机译:ATG5介导的自噬抑制NF-κB信号,以限制对肾损伤的上皮炎症反应

获取原文
           

摘要

G2/M-arrested proximal tubular epithelial cells (TECs) after renal injury are linked to increased cytokines production. ATG5-mediated autophagy in proximal TECs has recently been shown to protect against G2/M cell cycle arrest and renal fibrosis. However, the impacts of autophagy in regulating inflammatorily response mounted by injured TECs remains largely unknown. In the present study, we investigated whether ATG5 acts as an innate immune suppressor in proximal TECs during kidney injury. Using the unilateral ureteric obstruction model in proximal tubule-specific autophagy-deficient mice, we demonstrated that ablation of epithelial ATG5 genes markedly impaired autophagy, resulting in enhanced nuclear factor κB (NF-κB) activation, macrophage and lymphocyte infiltration, and proinflammatory cytokines production in obstructed kidneys, as compared with wild-type mice. Following stimulation with angiotensin II (Ang II), siRNA silencing of ATG5 in cultured HK-2 cells or ATG5-deficient primary proximal TECs produced more cytokines, including IL-1β, IL-6, and TNF-α than did their control cells. Overexpressed ATG5, but not the autophagy-incompetent ATG5 mutant K130R in HK-2 cells, rendered resistant to Ang II-induced inflammatory response. Immunofluorescence assay indicated that ATG5 and p65 colocalized in the nucleus and cytoplasm, and their interaction was verified in immunoprecipitation assay from HEK-293T cell extracts. Genetic downregulation of endogenous ATG5 increased Ang II-induced phosphorylation and nuclear translocation of p65 and transcriptional activity of NF-κB, whereas the overexpressed ATG5, rather than ATG5 mutant K130R, hampered activation of NF-κB signaling, suggest an autophagy-dependent anti-inflammatory effect of ATG5. Further, pharmacological manipulation of autophagy yielded similar results both in vivo and in vitro. Additionally, JSH-23, a specific inhibitor of NF-κB nuclear translocation, rescued Ang II-driven IL-1β production in ATG5 siRNA-treated cells and decreased the proportion of cells in G2/M phase. In conclusion, ATG5-mediated autophagy in tubules targets NF-κB signaling to protect against renal inflammation.
机译:肾损伤后G2 / M污染的近端管状上皮细胞(TECS)与增加的细胞因子产生有关。近期TECS的ATG5介导的自噬已被证明可以防止G2 / M细胞周期停滞和肾纤维化。然而,自噬对调节受伤的TECS安装的炎症性反应的影响仍然很大程度上。在本研究中,我们研究了ATG5是否在肾脏损伤期间作为近端TECS的先天免疫抑制剂。使用单侧输尿管梗阻模型在近端小管特异性自噬缺乏小鼠中,我们证明了上皮ATG5基因的消融显着损害了自噬损害,导致核因子κB(NF-κB)活化,巨噬细胞和淋巴细胞浸润,以及促炎细胞因子的产生与野生型小鼠相比,肾脏受阻。随着血管紧张素II(Ang II)的刺激后,培养的HK-2细胞中的ATG5的siRNA沉默或ATG5缺乏初级近端TECS产生了更多细胞因子,包括IL-1β,IL-6和TNF-α比其对照细胞。过表达的ATG5,但不是HK-2细胞中的自噬 - 无能的ATG5突变K130R,使抗Ang II诱导的炎症反应具有抗性。免疫荧光测定表明,从HEK-293T细胞提取物中验证了在核和细胞质中粘接在细胞核和细胞质中的ATG5和P65及其相互作用。内源性ATG5的遗传下调增加了Ang II诱导的P65磷酸化和NF-κB的转录活性,而过表达ATG5,而不是ATG5突变K130R,阻碍了NF-κB信号传导的激活,表明了自噬依赖性的抗 - ATG5的炎症作用。此外,对自噬的药理操纵在体内和体外均产生类似的结果。另外,JSH-23是NF-κB核转移的特异性抑制剂,在ATG5 siRNA处理细胞中拯救了Ang II驱动的IL-1β产生,并降低了G2 / M相中细胞的比例。总之,小管中的ATG5介导的自噬靶向NF-κB信号,以防止肾炎。

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