High salt diet alleviates disease severity in native experimental autoimmune uveitis.

Frontiers in ophthalmology Pub Date : 2024-05-31 eCollection Date: 2024-01-01 DOI:10.3389/fopht.2024.1370374
Naomi Derluyn, Vincent Foucart, Marko Verce, Rami Abdo, Louis Vaudoisey, Deborah Lipski, Véronique Flamand, Amandine Everard, Catherine Bruyns, François Willermain
{"title":"High salt diet alleviates disease severity in native experimental autoimmune uveitis.","authors":"Naomi Derluyn, Vincent Foucart, Marko Verce, Rami Abdo, Louis Vaudoisey, Deborah Lipski, Véronique Flamand, Amandine Everard, Catherine Bruyns, François Willermain","doi":"10.3389/fopht.2024.1370374","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Recent studies reported a link between high salt diet (HSD) and clinical exacerbation in mouse models of autoimmune diseases, mainly through the induction of pathogenic Th17 cells and/or HSD-induced dysbiosis. However, the topic remains controversial and not fully understood.</p><p><strong>Purpose: </strong>In this study, we investigated the effects of HSD on the development of experimental autoimmune uveitis (EAU) in C57BL/6J mice.</p><p><strong>Methods and results: </strong>Unexpectedly, our data showed a significant attenuating effect of HSD on disease severity of native EAU, induced by direct immunization with IRBP peptide. That said, HSD had no effect on EAU disease severity induced by adoptive transfer of semi-purified auto-reactive IRBP-specific T lymphocytes. Accordingly, HSD did not affect IRBP-specific systemic afferent immune response as attested by no HSD-linked changes in T lymphocytes proliferation, cytokine production and Treg proportion. Gut microbiota analysis from cecal samples in naïve and EAU mice demonstrated that HSD affected differentially α-diversity between groups, whereas β-diversity was significantly modified in all groups. Unknown <i>Tannerellaceae</i> was the only taxon associated to HSD exposure in all treatment groups. Interestingly, a significantly higher abundance of unknown <i>Gastranaerophilales</i>, with potential anti-inflammatory properties, appeared in HSD-fed native EAU mice, only.</p><p><strong>Discussion: </strong>In conclusion, our study suggests a possible impact of HSD on gut microbiota composition and consequently on development and clinical severity of EAU. Further studies are required to investigate the potential beneficial role of <i>Gastranaerophilales</i> in EAU.</p>","PeriodicalId":73096,"journal":{"name":"Frontiers in ophthalmology","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2024-05-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11182228/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Frontiers in ophthalmology","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.3389/fopht.2024.1370374","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2024/1/1 0:00:00","PubModel":"eCollection","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Recent studies reported a link between high salt diet (HSD) and clinical exacerbation in mouse models of autoimmune diseases, mainly through the induction of pathogenic Th17 cells and/or HSD-induced dysbiosis. However, the topic remains controversial and not fully understood.

Purpose: In this study, we investigated the effects of HSD on the development of experimental autoimmune uveitis (EAU) in C57BL/6J mice.

Methods and results: Unexpectedly, our data showed a significant attenuating effect of HSD on disease severity of native EAU, induced by direct immunization with IRBP peptide. That said, HSD had no effect on EAU disease severity induced by adoptive transfer of semi-purified auto-reactive IRBP-specific T lymphocytes. Accordingly, HSD did not affect IRBP-specific systemic afferent immune response as attested by no HSD-linked changes in T lymphocytes proliferation, cytokine production and Treg proportion. Gut microbiota analysis from cecal samples in naïve and EAU mice demonstrated that HSD affected differentially α-diversity between groups, whereas β-diversity was significantly modified in all groups. Unknown Tannerellaceae was the only taxon associated to HSD exposure in all treatment groups. Interestingly, a significantly higher abundance of unknown Gastranaerophilales, with potential anti-inflammatory properties, appeared in HSD-fed native EAU mice, only.

Discussion: In conclusion, our study suggests a possible impact of HSD on gut microbiota composition and consequently on development and clinical severity of EAU. Further studies are required to investigate the potential beneficial role of Gastranaerophilales in EAU.

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
高盐饮食可减轻原生实验性自身免疫性葡萄膜炎的病情严重程度。
背景:最近的研究报道了高盐饮食(HSD)与自身免疫性疾病小鼠模型临床恶化之间的联系,主要是通过诱导致病性Th17细胞和/或HSD诱导的菌群失调。目的:在这项研究中,我们调查了 HSD 对 C57BL/6J 小鼠实验性自身免疫性葡萄膜炎(EAU)发病的影响:出乎意料的是,我们的数据显示,HSD对IRBP肽直接免疫诱导的原发性EAU的疾病严重程度有显著的抑制作用。尽管如此,HSD对通过收养性转移半纯化的自身反应性IRBP特异性T淋巴细胞诱导的EAU疾病严重程度没有影响。因此,HSD不会影响IRBP特异性的全身传入性免疫反应,T淋巴细胞的增殖、细胞因子的产生和Treg比例没有发生与HSD相关的变化就证明了这一点。对天真小鼠和 EAU 小鼠盲肠样本进行的肠道微生物群分析表明,HSD 对不同组间的α-多样性有不同影响,而β-多样性在所有组中都有显著变化。在所有处理组中,未知丹顶鹤科是唯一与接触 HSD 相关的分类群。有趣的是,仅在喂食 HSD 的本地 EAU 小鼠中,具有潜在抗炎特性的未知嗜胃肠菌的丰度明显更高:总之,我们的研究表明 HSD 可能会影响肠道微生物群的组成,进而影响 EAU 的发展和临床严重程度。还需要进一步研究嗜胃肠球菌对 EAU 的潜在有益作用。
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 去求助
来源期刊
CiteScore
0.50
自引率
0.00%
发文量
0
期刊最新文献
Comparison of the PlusoptiX A16 and vision screener V100. UV light and the ocular lens: a review of exposure models and resulting biomolecular changes. Diagnostic accuracy of a modularized, virtual-reality-based automated pupillometer for detection of relative afferent pupillary defect in unilateral optic neuropathies. Advances in the management of intraocular foreign bodies. Large animal model species in pluripotent stem cell therapy research and development for retinal diseases: a systematic review.
×
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