巨噬细胞 ATG16L1 的表达可通过促进噬脂作用抑制 MASH 的发展。

IF 14 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Clinical and Molecular Hepatology Pub Date : 2024-07-01 Epub Date: 2024-05-10 DOI:10.3350/cmh.2024.0107
Qi Wang, Qingfa Bu, Zibo Xu, Yuan Liang, Jinren Zhou, Yufeng Pan, Haoming Zhou, Ling Lu
{"title":"巨噬细胞 ATG16L1 的表达可通过促进噬脂作用抑制 MASH 的发展。","authors":"Qi Wang, Qingfa Bu, Zibo Xu, Yuan Liang, Jinren Zhou, Yufeng Pan, Haoming Zhou, Ling Lu","doi":"10.3350/cmh.2024.0107","DOIUrl":null,"url":null,"abstract":"<p><strong>Background/aims: </strong>Metabolic dysfunction-associated steatohepatitis (MASH) is an unmet clinical challenge due to the rapid increased occurrence but lacking approved drugs. Autophagy-related protein 16-like 1 (ATG16L1) plays an important role in the process of autophagy, which is indispensable for proper biogenesis of the autophagosome, but its role in modulating macrophage-related inflammation and metabolism during MASH has not been documented. Here, we aimed to elucidate the role of ATG16L1 in the progression of MASH.</p><p><strong>Methods: </strong>Expression analysis was performed with liver samples from human and mice. MASH models were induced in myeloid-specific Atg16l1-deficient and myeloid-specific Atg16l1-overexpressed mice by high-fat and high-cholesterol diet or methionine- and choline-deficient diet to explore the function and mechanism of macrophage ATG16L1 in MASH.</p><p><strong>Results: </strong>Macrophage-specific Atg16l1 knockout exacerbated MASH and inhibited energy expenditure, whereas macrophage-specific Atg16l1 transgenic overexpression attenuated MASH and promotes energy expenditure. Mechanistically, Atg16l1 knockout inhibited macrophage lipophagy, thereby suppressing macrophage β-oxidation and decreasing the production of 4-hydroxynonenal, which further inhibited stimulator of interferon genes(STING) carbonylation. STING palmitoylation was enhanced, STING trafficking from the endoplasmic reticulum to the Golgi was promoted, and downstream STING signaling was activated, promoting proinflammatory and profibrotic cytokines secretion, resulting in hepatic steatosis and hepatic stellate cells activation. Moreover, Atg16l1-deficiency enhanced macrophage phagosome ability but inhibited lysosome formation, engulfing mtDNA released by pyroptotic hepatocytes. Increased mtDNA promoted cGAS/STING signaling activation. Moreover, pharmacological promotion of ATG16L1 substantially blocked MASH progression.</p><p><strong>Conclusion: </strong>ATG16L1 suppresses MASH progression by maintaining macrophage lipophagy, restraining liver inflammation, and may be a promising therapeutic target for MASH management.</p>","PeriodicalId":10275,"journal":{"name":"Clinical and Molecular Hepatology","volume":null,"pages":null},"PeriodicalIF":14.0000,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11261221/pdf/","citationCount":"0","resultStr":"{\"title\":\"Macrophage ATG16L1 expression suppresses metabolic dysfunction-associated steatohepatitis progression by promoting lipophagy.\",\"authors\":\"Qi Wang, Qingfa Bu, Zibo Xu, Yuan Liang, Jinren Zhou, Yufeng Pan, Haoming Zhou, Ling Lu\",\"doi\":\"10.3350/cmh.2024.0107\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><strong>Background/aims: </strong>Metabolic dysfunction-associated steatohepatitis (MASH) is an unmet clinical challenge due to the rapid increased occurrence but lacking approved drugs. Autophagy-related protein 16-like 1 (ATG16L1) plays an important role in the process of autophagy, which is indispensable for proper biogenesis of the autophagosome, but its role in modulating macrophage-related inflammation and metabolism during MASH has not been documented. Here, we aimed to elucidate the role of ATG16L1 in the progression of MASH.</p><p><strong>Methods: </strong>Expression analysis was performed with liver samples from human and mice. MASH models were induced in myeloid-specific Atg16l1-deficient and myeloid-specific Atg16l1-overexpressed mice by high-fat and high-cholesterol diet or methionine- and choline-deficient diet to explore the function and mechanism of macrophage ATG16L1 in MASH.</p><p><strong>Results: </strong>Macrophage-specific Atg16l1 knockout exacerbated MASH and inhibited energy expenditure, whereas macrophage-specific Atg16l1 transgenic overexpression attenuated MASH and promotes energy expenditure. Mechanistically, Atg16l1 knockout inhibited macrophage lipophagy, thereby suppressing macrophage β-oxidation and decreasing the production of 4-hydroxynonenal, which further inhibited stimulator of interferon genes(STING) carbonylation. STING palmitoylation was enhanced, STING trafficking from the endoplasmic reticulum to the Golgi was promoted, and downstream STING signaling was activated, promoting proinflammatory and profibrotic cytokines secretion, resulting in hepatic steatosis and hepatic stellate cells activation. Moreover, Atg16l1-deficiency enhanced macrophage phagosome ability but inhibited lysosome formation, engulfing mtDNA released by pyroptotic hepatocytes. Increased mtDNA promoted cGAS/STING signaling activation. Moreover, pharmacological promotion of ATG16L1 substantially blocked MASH progression.</p><p><strong>Conclusion: </strong>ATG16L1 suppresses MASH progression by maintaining macrophage lipophagy, restraining liver inflammation, and may be a promising therapeutic target for MASH management.</p>\",\"PeriodicalId\":10275,\"journal\":{\"name\":\"Clinical and Molecular Hepatology\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":14.0000,\"publicationDate\":\"2024-07-01\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11261221/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Clinical and Molecular Hepatology\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.3350/cmh.2024.0107\",\"RegionNum\":1,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"2024/5/10 0:00:00\",\"PubModel\":\"Epub\",\"JCR\":\"Q1\",\"JCRName\":\"GASTROENTEROLOGY & HEPATOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Clinical and Molecular Hepatology","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.3350/cmh.2024.0107","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2024/5/10 0:00:00","PubModel":"Epub","JCR":"Q1","JCRName":"GASTROENTEROLOGY & HEPATOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

背景/目的:代谢功能障碍相关性脂肪性肝炎(MASH)的发病率迅速上升,但却缺乏已获批准的药物,因此是一项尚未解决的临床难题。自噬相关蛋白 16-like 1(ATG16L1)在自噬过程中发挥重要作用,自噬体的正常生物生成离不开自噬相关蛋白 16-like 1,但其在 MASH 期间调节巨噬细胞相关炎症和代谢的作用尚未见文献报道。在此,我们旨在阐明 ATG16L1 在 MASH 进展过程中的作用:方法:对人和小鼠的肝脏样本进行表达分析。通过高脂高胆固醇饮食或蛋氨酸和胆碱缺乏饮食诱导髓系特异性Atg16l1缺陷小鼠和髓系特异性Atg16l1缺失小鼠建立MASH模型,探讨巨噬细胞ATG16L1在MASH中的功能和机制:结果:巨噬细胞特异性Atg16l1敲除可加重MASH并抑制能量消耗,而巨噬细胞特异性Atg16l1转基因过表达可减轻MASH并促进能量消耗。从机理上讲,Atg16l1基因敲除抑制了巨噬细胞的噬脂性,从而抑制了巨噬细胞的β-氧化,减少了4-羟基壬烯醛(4-HNE)的产生,这进一步抑制了干扰素基因刺激因子(STING)的羰基化。STING 棕榈酰化作用增强,促进了 STING 从 ER 向高尔基体的迁移,激活了 STING 的下游信号传导,促进了促炎和促组织坏死细胞因子的分泌,导致肝脏脂肪变性和造血干细胞活化。此外,Atg16l1缺陷增强了巨噬细胞吞噬能力,但抑制了溶酶体的形成,从而吞噬了脓毒性肝细胞释放的mtDNA。增加的mtDNA促进了cGAS/STING信号的激活。此外,ATG16L1的药理促进作用大大阻止了MASH的进展:结论:ATG16L1通过维持巨噬细胞噬脂功能抑制MASH进展,抑制肝脏炎症,可能是治疗MASH的一个有前途的靶点。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
Macrophage ATG16L1 expression suppresses metabolic dysfunction-associated steatohepatitis progression by promoting lipophagy.

Background/aims: Metabolic dysfunction-associated steatohepatitis (MASH) is an unmet clinical challenge due to the rapid increased occurrence but lacking approved drugs. Autophagy-related protein 16-like 1 (ATG16L1) plays an important role in the process of autophagy, which is indispensable for proper biogenesis of the autophagosome, but its role in modulating macrophage-related inflammation and metabolism during MASH has not been documented. Here, we aimed to elucidate the role of ATG16L1 in the progression of MASH.

Methods: Expression analysis was performed with liver samples from human and mice. MASH models were induced in myeloid-specific Atg16l1-deficient and myeloid-specific Atg16l1-overexpressed mice by high-fat and high-cholesterol diet or methionine- and choline-deficient diet to explore the function and mechanism of macrophage ATG16L1 in MASH.

Results: Macrophage-specific Atg16l1 knockout exacerbated MASH and inhibited energy expenditure, whereas macrophage-specific Atg16l1 transgenic overexpression attenuated MASH and promotes energy expenditure. Mechanistically, Atg16l1 knockout inhibited macrophage lipophagy, thereby suppressing macrophage β-oxidation and decreasing the production of 4-hydroxynonenal, which further inhibited stimulator of interferon genes(STING) carbonylation. STING palmitoylation was enhanced, STING trafficking from the endoplasmic reticulum to the Golgi was promoted, and downstream STING signaling was activated, promoting proinflammatory and profibrotic cytokines secretion, resulting in hepatic steatosis and hepatic stellate cells activation. Moreover, Atg16l1-deficiency enhanced macrophage phagosome ability but inhibited lysosome formation, engulfing mtDNA released by pyroptotic hepatocytes. Increased mtDNA promoted cGAS/STING signaling activation. Moreover, pharmacological promotion of ATG16L1 substantially blocked MASH progression.

Conclusion: ATG16L1 suppresses MASH progression by maintaining macrophage lipophagy, restraining liver inflammation, and may be a promising therapeutic target for MASH management.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
Clinical and Molecular Hepatology
Clinical and Molecular Hepatology Medicine-Hepatology
CiteScore
15.60
自引率
9.00%
发文量
89
审稿时长
10 weeks
期刊介绍: Clinical and Molecular Hepatology is an internationally recognized, peer-reviewed, open-access journal published quarterly in English. Its mission is to disseminate cutting-edge knowledge, trends, and insights into hepatobiliary diseases, fostering an inclusive academic platform for robust debate and discussion among clinical practitioners, translational researchers, and basic scientists. With a multidisciplinary approach, the journal strives to enhance public health, particularly in the resource-limited Asia-Pacific region, which faces significant challenges such as high prevalence of B viral infection and hepatocellular carcinoma. Furthermore, Clinical and Molecular Hepatology prioritizes epidemiological studies of hepatobiliary diseases across diverse regions including East Asia, North Asia, Southeast Asia, Central Asia, South Asia, Southwest Asia, Pacific, Africa, Central Europe, Eastern Europe, Central America, and South America. The journal publishes a wide range of content, including original research papers, meta-analyses, letters to the editor, case reports, reviews, guidelines, editorials, and liver images and pathology, encompassing all facets of hepatology.
期刊最新文献
Letter regarding "Bariatric intervention improves metabolic dysfunction-associated steatohepatitis in patients with obesity: A systematic review and meta-analysis". Correspondence on Editorial regarding "Spotting Undiagnosed Significant Liver Fibrosis in the General Population: Impact on Subsequent Clinical Care". Correspondence on Letter regarding "A complement to epigenetics in MASLD". PNPLA3 is one of the bridges between TM6SF2 E167K variant and MASLD. Correspondence on Editorial regarding "KCTD17 as a RAS stabilizer in hepatocellular carcinoma progression".
×
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
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1