细菌鞘磷脂通过抑制 ILC3 衍生的 IL-22 的产生加剧结肠炎

IF 7.1 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY
Bin Bao , Youyuan Wang , Pavl Boudreau , Xinyang Song , Meng Wu , Xi Chen , Izabel Patik , Ying Tang , Jodie Ouahed , Amit Ringel , Jared Barends , Chuan Wu , Emily Balskus , Jay Thiagarajah , Jian Liu , Michael R. Wessels , Wayne Isaac Lencer , Dennis L. Kasper , Dingding An , Bruce Harold Horwitz , Scott B. Snapper
{"title":"细菌鞘磷脂通过抑制 ILC3 衍生的 IL-22 的产生加剧结肠炎","authors":"Bin Bao ,&nbsp;Youyuan Wang ,&nbsp;Pavl Boudreau ,&nbsp;Xinyang Song ,&nbsp;Meng Wu ,&nbsp;Xi Chen ,&nbsp;Izabel Patik ,&nbsp;Ying Tang ,&nbsp;Jodie Ouahed ,&nbsp;Amit Ringel ,&nbsp;Jared Barends ,&nbsp;Chuan Wu ,&nbsp;Emily Balskus ,&nbsp;Jay Thiagarajah ,&nbsp;Jian Liu ,&nbsp;Michael R. Wessels ,&nbsp;Wayne Isaac Lencer ,&nbsp;Dennis L. Kasper ,&nbsp;Dingding An ,&nbsp;Bruce Harold Horwitz ,&nbsp;Scott B. Snapper","doi":"10.1016/j.jcmgh.2024.04.007","DOIUrl":null,"url":null,"abstract":"<div><h3>Background &amp; Aims</h3><p>Gut bacterial sphingolipids, primarily produced by Bacteroidetes, have dual roles as bacterial virulence factors and regulators of the host mucosal immune system, including regulatory T cells and invariant natural killer T cells. Patients with inflammatory bowel disease display altered sphingolipids profiles in fecal samples. However, how bacterial sphingolipids modulate mucosal homeostasis and regulate intestinal inflammation remains unclear.</p></div><div><h3>Methods</h3><p>We used dextran sodium sulfate (DSS)-induced colitis in mice monocolonized with <em>Bacteroides fragilis</em> strains expressing or lacking sphingolipids to assess the influence of bacterial sphingolipids on intestinal inflammation using transcriptional, protein, and cellular analyses. Colonic explant and organoid were used to study the function of bacterial sphingolipids. Host mucosal immune cells and cytokines were profiled and characterized using flow cytometry, enzyme-linked immunosorbent assay, and Western blot, and cytokine function in vivo was investigated by monoclonal antibody injection.</p></div><div><h3>Results</h3><p><em>B fragilis</em> sphingolipids exacerbated intestinal inflammation. Mice monocolonized with <em>B fragilis</em> lacking sphingolipids exhibited less severe DSS-induced colitis. This amelioration of colitis was associated with increased production of interleukin (IL)-22 by ILC3. Mice colonized with <em>B fragilis</em> lacking sphingolipids following DSS treatment showed enhanced epithelial STAT3 activity, intestinal cell proliferation, and antimicrobial peptide production. Protection against DSS colitis associated with <em>B fragilis</em> lacking sphingolipids was reversed on IL22 blockade. Furthermore, bacterial sphingolipids restricted epithelial IL18 production following DSS treatment and interfered with IL22 production by a subset of ILC3 cells expressing both IL18R and major histocompatibility complex class II.</p></div><div><h3>Conclusions</h3><p><em>B fragilis</em>–derived sphingolipids exacerbate mucosal inflammation by impeding epithelial IL18 expression and concomitantly suppressing the production of IL22 by ILC3 cells.</p></div>","PeriodicalId":55974,"journal":{"name":"Cellular and Molecular Gastroenterology and Hepatology","volume":null,"pages":null},"PeriodicalIF":7.1000,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2352345X24001048/pdfft?md5=59a34194ef86160c5f3532ad4c9dd497&pid=1-s2.0-S2352345X24001048-main.pdf","citationCount":"0","resultStr":"{\"title\":\"Bacterial Sphingolipids Exacerbate Colitis by Inhibiting ILC3-derived IL-22 Production\",\"authors\":\"Bin Bao ,&nbsp;Youyuan Wang ,&nbsp;Pavl Boudreau ,&nbsp;Xinyang Song ,&nbsp;Meng Wu ,&nbsp;Xi Chen ,&nbsp;Izabel Patik ,&nbsp;Ying Tang ,&nbsp;Jodie Ouahed ,&nbsp;Amit Ringel ,&nbsp;Jared Barends ,&nbsp;Chuan Wu ,&nbsp;Emily Balskus ,&nbsp;Jay Thiagarajah ,&nbsp;Jian Liu ,&nbsp;Michael R. Wessels ,&nbsp;Wayne Isaac Lencer ,&nbsp;Dennis L. Kasper ,&nbsp;Dingding An ,&nbsp;Bruce Harold Horwitz ,&nbsp;Scott B. Snapper\",\"doi\":\"10.1016/j.jcmgh.2024.04.007\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<div><h3>Background &amp; Aims</h3><p>Gut bacterial sphingolipids, primarily produced by Bacteroidetes, have dual roles as bacterial virulence factors and regulators of the host mucosal immune system, including regulatory T cells and invariant natural killer T cells. Patients with inflammatory bowel disease display altered sphingolipids profiles in fecal samples. However, how bacterial sphingolipids modulate mucosal homeostasis and regulate intestinal inflammation remains unclear.</p></div><div><h3>Methods</h3><p>We used dextran sodium sulfate (DSS)-induced colitis in mice monocolonized with <em>Bacteroides fragilis</em> strains expressing or lacking sphingolipids to assess the influence of bacterial sphingolipids on intestinal inflammation using transcriptional, protein, and cellular analyses. Colonic explant and organoid were used to study the function of bacterial sphingolipids. Host mucosal immune cells and cytokines were profiled and characterized using flow cytometry, enzyme-linked immunosorbent assay, and Western blot, and cytokine function in vivo was investigated by monoclonal antibody injection.</p></div><div><h3>Results</h3><p><em>B fragilis</em> sphingolipids exacerbated intestinal inflammation. Mice monocolonized with <em>B fragilis</em> lacking sphingolipids exhibited less severe DSS-induced colitis. This amelioration of colitis was associated with increased production of interleukin (IL)-22 by ILC3. Mice colonized with <em>B fragilis</em> lacking sphingolipids following DSS treatment showed enhanced epithelial STAT3 activity, intestinal cell proliferation, and antimicrobial peptide production. Protection against DSS colitis associated with <em>B fragilis</em> lacking sphingolipids was reversed on IL22 blockade. Furthermore, bacterial sphingolipids restricted epithelial IL18 production following DSS treatment and interfered with IL22 production by a subset of ILC3 cells expressing both IL18R and major histocompatibility complex class II.</p></div><div><h3>Conclusions</h3><p><em>B fragilis</em>–derived sphingolipids exacerbate mucosal inflammation by impeding epithelial IL18 expression and concomitantly suppressing the production of IL22 by ILC3 cells.</p></div>\",\"PeriodicalId\":55974,\"journal\":{\"name\":\"Cellular and Molecular Gastroenterology and Hepatology\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":7.1000,\"publicationDate\":\"2024-01-01\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.sciencedirect.com/science/article/pii/S2352345X24001048/pdfft?md5=59a34194ef86160c5f3532ad4c9dd497&pid=1-s2.0-S2352345X24001048-main.pdf\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Cellular and Molecular Gastroenterology and Hepatology\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://www.sciencedirect.com/science/article/pii/S2352345X24001048\",\"RegionNum\":1,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"GASTROENTEROLOGY & HEPATOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cellular and Molecular Gastroenterology and Hepatology","FirstCategoryId":"3","ListUrlMain":"https://www.sciencedirect.com/science/article/pii/S2352345X24001048","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"GASTROENTEROLOGY & HEPATOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

背景与目的:肠道细菌鞘磷脂主要由类杆菌产生,具有双重作用,既是细菌毒力因子,也是宿主粘膜免疫系统(包括调节性 T 细胞和 iNKT 细胞)的调节因子。IBD 患者粪便样本中的鞘磷脂谱发生了改变。然而,细菌鞘磷脂如何调节粘膜稳态并调节肠道炎症仍不清楚:方法:我们利用表达或缺乏鞘磷脂的脆弱拟杆菌菌株单菌落小鼠 DSS 诱导的结肠炎,通过转录、蛋白质和细胞分析来评估细菌鞘磷脂对肠道炎症的影响。结肠外植体和类器官用于研究细菌鞘磷脂的功能。使用流式细胞术、酶联免疫吸附试验(ELISA)和免疫印迹(Western Blot)对宿主粘膜免疫细胞和细胞因子进行了分析和表征,并通过注射单克隆抗体研究了细胞因子在体内的功能:结果:脆弱拟杆菌鞘磷脂加剧了肠道炎症。用缺乏鞘磷脂的脆弱拟杆菌对小鼠进行单克隆化处理后,DSS诱导的结肠炎症状较轻。结肠炎的改善与 ILC3 产生的白细胞介素-22 增加有关。用缺乏鞘磷脂的脆弱拟杆菌对小鼠进行定植后,小鼠的上皮细胞 STAT3 活性、肠道细胞增殖和抗菌肽的产生均有所增强。缺乏鞘磷脂的脆弱拟杆菌对DSS结肠炎的保护作用在IL-22阻断后被逆转。此外,细菌鞘磷脂限制了DSS处理后上皮细胞IL-18的产生,并干扰了同时表达IL-18R和MHC II的ILC3细胞亚群产生IL-22:结论:来源于脆弱拟杆菌的鞘脂通过阻碍上皮细胞IL-18的表达并同时抑制ILC3细胞产生IL-22而加剧粘膜炎症。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

Bacterial Sphingolipids Exacerbate Colitis by Inhibiting ILC3-derived IL-22 Production

Bacterial Sphingolipids Exacerbate Colitis by Inhibiting ILC3-derived IL-22 Production

Bacterial Sphingolipids Exacerbate Colitis by Inhibiting ILC3-derived IL-22 Production

Background & Aims

Gut bacterial sphingolipids, primarily produced by Bacteroidetes, have dual roles as bacterial virulence factors and regulators of the host mucosal immune system, including regulatory T cells and invariant natural killer T cells. Patients with inflammatory bowel disease display altered sphingolipids profiles in fecal samples. However, how bacterial sphingolipids modulate mucosal homeostasis and regulate intestinal inflammation remains unclear.

Methods

We used dextran sodium sulfate (DSS)-induced colitis in mice monocolonized with Bacteroides fragilis strains expressing or lacking sphingolipids to assess the influence of bacterial sphingolipids on intestinal inflammation using transcriptional, protein, and cellular analyses. Colonic explant and organoid were used to study the function of bacterial sphingolipids. Host mucosal immune cells and cytokines were profiled and characterized using flow cytometry, enzyme-linked immunosorbent assay, and Western blot, and cytokine function in vivo was investigated by monoclonal antibody injection.

Results

B fragilis sphingolipids exacerbated intestinal inflammation. Mice monocolonized with B fragilis lacking sphingolipids exhibited less severe DSS-induced colitis. This amelioration of colitis was associated with increased production of interleukin (IL)-22 by ILC3. Mice colonized with B fragilis lacking sphingolipids following DSS treatment showed enhanced epithelial STAT3 activity, intestinal cell proliferation, and antimicrobial peptide production. Protection against DSS colitis associated with B fragilis lacking sphingolipids was reversed on IL22 blockade. Furthermore, bacterial sphingolipids restricted epithelial IL18 production following DSS treatment and interfered with IL22 production by a subset of ILC3 cells expressing both IL18R and major histocompatibility complex class II.

Conclusions

B fragilis–derived sphingolipids exacerbate mucosal inflammation by impeding epithelial IL18 expression and concomitantly suppressing the production of IL22 by ILC3 cells.

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
CiteScore
13.00
自引率
2.80%
发文量
246
审稿时长
42 days
期刊介绍: "Cell and Molecular Gastroenterology and Hepatology (CMGH)" is a journal dedicated to advancing the understanding of digestive biology through impactful research that spans the spectrum of normal gastrointestinal, hepatic, and pancreatic functions, as well as their pathologies. The journal's mission is to publish high-quality, hypothesis-driven studies that offer mechanistic novelty and are methodologically robust, covering a wide range of themes in gastroenterology, hepatology, and pancreatology. CMGH reports on the latest scientific advances in cell biology, immunology, physiology, microbiology, genetics, and neurobiology related to gastrointestinal, hepatobiliary, and pancreatic health and disease. The research published in CMGH is designed to address significant questions in the field, utilizing a variety of experimental approaches, including in vitro models, patient-derived tissues or cells, and animal models. This multifaceted approach enables the journal to contribute to both fundamental discoveries and their translation into clinical applications, ultimately aiming to improve patient care and treatment outcomes in digestive health.
×
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