TBK1-Zyxin signaling controls tumor-associated macrophage recruitment to mitigate antitumor immunity.

Ruyuan Zhou,Mengqiu Wang,Xiao Li,Yutong Liu,Yihan Yao,Ailian Wang,Chen Chen,Qian Zhang,Qirou Wu,Qi Zhang,Dante Neculai,Bing Xia,Jian-Zhong Shao,Xin-Hua Feng,Tingbo Liang,Jian Zou,Xiaojian Wang,Pinglong Xu
{"title":"TBK1-Zyxin signaling controls tumor-associated macrophage recruitment to mitigate antitumor immunity.","authors":"Ruyuan Zhou,Mengqiu Wang,Xiao Li,Yutong Liu,Yihan Yao,Ailian Wang,Chen Chen,Qian Zhang,Qirou Wu,Qi Zhang,Dante Neculai,Bing Xia,Jian-Zhong Shao,Xin-Hua Feng,Tingbo Liang,Jian Zou,Xiaojian Wang,Pinglong Xu","doi":"10.1038/s44318-024-00244-9","DOIUrl":null,"url":null,"abstract":"Mechanical control is fundamental for cellular localization within a tissue, including for tumor-associated macrophages (TAMs). While the innate immune sensing pathways cGAS-STING and RLR-MAVS impact the pathogenesis and therapeutics of malignant diseases, their effects on cell residency and motility remain incompletely understood. Here, we uncovered that TBK1 kinase, activated by cGAS-STING or RLR-MAVS signaling in macrophages, directly phosphorylates and mobilizes Zyxin, a key regulator of actin dynamics. Under pathological conditions and in STING or MAVS signalosomes, TBK1-mediated Zyxin phosphorylation at S143 facilitates rapid recruitment of phospho-Zyxin to focal adhesions, leading to subsequent F-actin reorganization and reduced macrophage migration. Intratumoral STING-TBK1-Zyxin signaling was evident in TAMs and critical in antitumor immunity. Furthermore, myeloid-specific or global disruption of this signaling decreased the population of CD11b+ F4/80+ TAMs and promoted PD-1-mediated antitumor immunotherapy. Thus, our findings identify a new biological function of innate immune sensing pathways by regulating macrophage tissue localization, thus providing insights into context-dependent mitigation of antitumor immunity.","PeriodicalId":501009,"journal":{"name":"The EMBO Journal","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2024-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"The EMBO Journal","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1038/s44318-024-00244-9","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

Abstract

Mechanical control is fundamental for cellular localization within a tissue, including for tumor-associated macrophages (TAMs). While the innate immune sensing pathways cGAS-STING and RLR-MAVS impact the pathogenesis and therapeutics of malignant diseases, their effects on cell residency and motility remain incompletely understood. Here, we uncovered that TBK1 kinase, activated by cGAS-STING or RLR-MAVS signaling in macrophages, directly phosphorylates and mobilizes Zyxin, a key regulator of actin dynamics. Under pathological conditions and in STING or MAVS signalosomes, TBK1-mediated Zyxin phosphorylation at S143 facilitates rapid recruitment of phospho-Zyxin to focal adhesions, leading to subsequent F-actin reorganization and reduced macrophage migration. Intratumoral STING-TBK1-Zyxin signaling was evident in TAMs and critical in antitumor immunity. Furthermore, myeloid-specific or global disruption of this signaling decreased the population of CD11b+ F4/80+ TAMs and promoted PD-1-mediated antitumor immunotherapy. Thus, our findings identify a new biological function of innate immune sensing pathways by regulating macrophage tissue localization, thus providing insights into context-dependent mitigation of antitumor immunity.
TBK1-Zyxin信号控制肿瘤相关巨噬细胞的招募,从而减轻抗肿瘤免疫。
机械控制是细胞(包括肿瘤相关巨噬细胞(TAMs))在组织内定位的基础。先天性免疫传感通路 cGAS-STING 和 RLR-MAVS 影响着恶性疾病的发病机制和治疗方法,但它们对细胞驻留和运动的影响仍不完全清楚。在这里,我们发现 TBK1 激酶在巨噬细胞中被 cGAS-STING 或 RLR-MAVS 信号激活后,会直接磷酸化并调动肌动蛋白动态的关键调节因子 Zyxin。在病理条件下,以及在 STING 或 MAVS 信号体中,TBK1 介导的 Zyxin 在 S143 处磷酸化可促进磷酸化 Zyxin 快速招募到焦点粘附处,从而导致随后的 F-肌动蛋白重组并减少巨噬细胞的迁移。瘤内 STING-TBK1-Zyxin 信号在 TAMs 中很明显,在抗肿瘤免疫中至关重要。此外,髓系特异性或全局性破坏这种信号传导会减少 CD11b+ F4/80+ TAMs 的数量,促进 PD-1 介导的抗肿瘤免疫疗法。因此,我们的研究结果发现了先天性免疫传感通路通过调节巨噬细胞组织定位的一种新的生物学功能,从而为抗肿瘤免疫的情境依赖性缓解提供了见解。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
自引率
0.00%
发文量
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信