The role of HDAC6 in enhancing macrophage autophagy via the autophagolysosomal pathway to alleviate legionella pneumophila-induced pneumonia.

IF 5.5 1区 农林科学 Q1 IMMUNOLOGY
Virulence Pub Date : 2024-12-01 Epub Date: 2024-03-11 DOI:10.1080/21505594.2024.2327096
Minjia Chen, Xiuqin Cao, Ronghui Zheng, Haixia Chen, Ruixia He, Hao Zhou, Zhiwei Yang
{"title":"The role of HDAC6 in enhancing macrophage autophagy via the autophagolysosomal pathway to alleviate legionella pneumophila-induced pneumonia.","authors":"Minjia Chen, Xiuqin Cao, Ronghui Zheng, Haixia Chen, Ruixia He, Hao Zhou, Zhiwei Yang","doi":"10.1080/21505594.2024.2327096","DOIUrl":null,"url":null,"abstract":"<p><p><i>Legionella pneumophila</i> (<i>L. pneumophila</i>) is a prevalent pathogenic bacterium responsible for significant global health concerns. Nonetheless, the precise pathogenic mechanisms of <i>L. pneumophila</i> have still remained elusive. Autophagy, a direct cellular response to <i>L. pneumophila</i> infection and other pathogens, involves the recognition and degradation of these invaders in lysosomes. Histone deacetylase 6 (HDAC6), a distinctive member of the histone deacetylase family, plays a multifaceted role in autophagy regulation. This study aimed to investigate the role of HDAC6 in macrophage autophagy via the autophagolysosomal pathway, leading to alleviate <i>L. pneumophila</i>-induced pneumonia. The results revealed a substantial upregulation of HDAC6 expression level in murine lung tissues infected by <i>L. pneumophila</i>. Notably, mice lacking HDAC6 exhibited a protective response against <i>L. pneumophila</i>-induced pulmonary tissue inflammation, which was characterized by the reduced bacterial load and diminished release of pro-inflammatory cytokines. Transcriptomic analysis has shed light on the regulatory role of HDAC6 in <i>L. pneumophila</i> infection in mice, particularly through the autophagy pathway of macrophages. Validation using <i>L. pneumophila</i>-induced macrophages from mice with HDAC6 gene knockout demonstrated a decrease in cellular bacterial load, activation of the autophagolysosomal pathway, and enhancement of cellular autophagic flux. In summary, the findings indicated that HDAC6 knockout could lead to the upregulation of p-ULK1 expression level, promoting the autophagy-lysosomal pathway, increasing autophagic flux, and ultimately strengthening the bactericidal capacity of macrophages. This contributes to the alleviation of <i>L. pneumophila</i>-induced pneumonia.</p>","PeriodicalId":23747,"journal":{"name":"Virulence","volume":null,"pages":null},"PeriodicalIF":5.5000,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10936600/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Virulence","FirstCategoryId":"99","ListUrlMain":"https://doi.org/10.1080/21505594.2024.2327096","RegionNum":1,"RegionCategory":"农林科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2024/3/11 0:00:00","PubModel":"Epub","JCR":"Q1","JCRName":"IMMUNOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Legionella pneumophila (L. pneumophila) is a prevalent pathogenic bacterium responsible for significant global health concerns. Nonetheless, the precise pathogenic mechanisms of L. pneumophila have still remained elusive. Autophagy, a direct cellular response to L. pneumophila infection and other pathogens, involves the recognition and degradation of these invaders in lysosomes. Histone deacetylase 6 (HDAC6), a distinctive member of the histone deacetylase family, plays a multifaceted role in autophagy regulation. This study aimed to investigate the role of HDAC6 in macrophage autophagy via the autophagolysosomal pathway, leading to alleviate L. pneumophila-induced pneumonia. The results revealed a substantial upregulation of HDAC6 expression level in murine lung tissues infected by L. pneumophila. Notably, mice lacking HDAC6 exhibited a protective response against L. pneumophila-induced pulmonary tissue inflammation, which was characterized by the reduced bacterial load and diminished release of pro-inflammatory cytokines. Transcriptomic analysis has shed light on the regulatory role of HDAC6 in L. pneumophila infection in mice, particularly through the autophagy pathway of macrophages. Validation using L. pneumophila-induced macrophages from mice with HDAC6 gene knockout demonstrated a decrease in cellular bacterial load, activation of the autophagolysosomal pathway, and enhancement of cellular autophagic flux. In summary, the findings indicated that HDAC6 knockout could lead to the upregulation of p-ULK1 expression level, promoting the autophagy-lysosomal pathway, increasing autophagic flux, and ultimately strengthening the bactericidal capacity of macrophages. This contributes to the alleviation of L. pneumophila-induced pneumonia.

HDAC6 在通过自噬溶酶体途径增强巨噬细胞自噬以缓解嗜肺军团菌诱发的肺炎中的作用
嗜肺军团菌(L. pneumophila)是一种普遍存在的致病细菌,对全球健康造成重大影响。然而,嗜肺军团菌的确切致病机制仍然难以捉摸。自噬是细胞对嗜肺肺孢子菌感染和其他病原体的直接反应,包括在溶酶体中识别和降解这些入侵者。组蛋白去乙酰化酶 6(HDAC6)是组蛋白去乙酰化酶家族的一个独特成员,在自噬调控中发挥着多方面的作用。本研究旨在探讨HDAC6通过自噬溶酶体途径在巨噬细胞自噬中的作用,从而缓解嗜肺菌诱发的肺炎。结果发现,在受嗜肺病毒感染的小鼠肺组织中,HDAC6的表达水平大幅上调。值得注意的是,缺乏 HDAC6 的小鼠对嗜肺蝇菌诱发的肺组织炎症表现出保护性反应,其特点是细菌负荷减少,促炎细胞因子释放减少。转录组分析揭示了 HDAC6 在小鼠嗜肺病毒感染中的调控作用,特别是通过巨噬细胞的自噬途径。利用 HDAC6 基因敲除小鼠的嗜肺病毒诱导的巨噬细胞进行验证,结果表明细胞细菌负荷减少,自噬溶酶体途径激活,细胞自噬通量增强。综上所述,研究结果表明,HDAC6 基因敲除可导致 p-ULK1 表达水平上调,促进自噬溶酶体通路,增加自噬通量,最终增强巨噬细胞的杀菌能力。这有助于缓解嗜肺菌诱发的肺炎。
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
Virulence
Virulence IMMUNOLOGY-MICROBIOLOGY
CiteScore
9.20
自引率
1.90%
发文量
123
审稿时长
6-12 weeks
期刊介绍: Virulence is a fully open access peer-reviewed journal. All articles will (if accepted) be available for anyone to read anywhere, at any time immediately on publication. Virulence is the first international peer-reviewed journal of its kind to focus exclusively on microbial pathogenicity, the infection process and host-pathogen interactions. To address the new infectious challenges, emerging infectious agents and antimicrobial resistance, there is a clear need for interdisciplinary research.
文献相关原料
公司名称 产品信息 采购帮参考价格
×
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