4.7 Article

Laminar shear stress inhibits inflammation by activating autophagy in human aortic endothelial cells through HMGB1 nuclear translocation

期刊

COMMUNICATIONS BIOLOGY
卷 5, 期 1, 页码 -

出版社

NATURE PORTFOLIO
DOI: 10.1038/s42003-022-03392-y

关键词

-

资金

  1. National Natural Science Foundation of China [82171553]
  2. Chinese Ministry of Foreign Affairs and Chinese Ministry of Education [105213000000200012]
  3. Fundamental Research Funds for the Central Universities

向作者/读者索取更多资源

The study reveals that laminar shear stress inhibits inflammatory response in human aortic endothelial cells through autophagy induction. EPHB2 and the LOC107986345/miR-128-3p/EPHB2 axis play crucial roles in laminar shear stress-mediated anti-atherosclerotic development.
The transcriptional response to laminar sheer stress in human aortic endothelial cells reveals a pathway leading to inhibition of endothelial cell inflammation by autophagy induction. Prevention and treatment of atherosclerosis (AS) by targeting the inflammatory response in vascular endothelial cells has attracted much attention in recent years. Laminar shear stress (LSS) has well-recognized anti-AS properties, however, the exact molecular mechanism remains unclear. In this study, we found that LSS could inhibit the increased expression of intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), cyclooxygenase-2 (COX-2), and matrix metallopeptidase-9 (MMP-9) caused by TNF-alpha in an autophagy-dependent pathway in human aortic endothelial cells (HAECs) and human umbilical vein endothelial cells (HUVECs). Whole-transcriptome sequencing analysis revealed that erythropoietin-producing hepatocyte receptor B2 (EPHB2) was a key gene in response to LSS. Moreover, co-immunoprecipitation assay indicated that LSS could enhance the EPHB2-mediated nuclear translocation of high mobility group box-1 (HMGB1), which interacts with Beclin-1 (BECN1) and finally leads to autophagy. Simultaneously, we identified an LSS-sensitive long non-coding RNA (lncRNA), LOC10798635, and constructed an LSS-related LOC107986345/miR-128-3p/EPHB2 regulatory axis. Further research revealed the anti-inflammatory effect of LSS depends on autophagy activation resulting from the nuclear translocation of HMGB1 via the LOC107986345/miR-128-3p/EPHB2 axis. Our study demonstrates that LSS could regulate the expression of EPHB2 in HAECs, and the LOC107986345/miR-128-3p/EPHB2 axis plays a vital role in AS development.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据