Gut microbiome promotes succinate-induced ulcerative colitis by enhancing glycolysis through SUCNR1/NF-κB signaling pathway.

IF 4.7 2区 生物学 Q2 CELL BIOLOGY
Long Huo, Qian Chen, Sailei Jia, Yuli Zhang, Lihui Wang, Xian Li, Zan Li, Boyun Sun, Jingyi Shan, Jiang Lin, Lili Yang, Hua Sui
{"title":"Gut microbiome promotes succinate-induced ulcerative colitis by enhancing glycolysis through SUCNR1/NF-κB signaling pathway.","authors":"Long Huo, Qian Chen, Sailei Jia, Yuli Zhang, Lihui Wang, Xian Li, Zan Li, Boyun Sun, Jingyi Shan, Jiang Lin, Lili Yang, Hua Sui","doi":"10.1152/ajpcell.00411.2025","DOIUrl":null,"url":null,"abstract":"<p><p>Ulcerative colitis (UC) is a chronic recurrent inflammatory disease. Previous studies demonstrate that excessive accumulation of gut microbial metabolites, especially succinate, increases the risk of disease progression. However, the role of succinate and its molecular mechanism have not been explored. We investigated the effects of succinate on colonic inflammation and intestinal microbiota and their association with succinate receptor (SUCNR1) signaling in 3% dextran sodium sulfate (DSS)-induced acute UC in C57BL/6J mice. After treatment, fecal bacteria from UC mice were evaluated by 16S rRNA sequencing. Colon tissues and cell lysates were collected and prepared for histological evaluation, immunohistochemistry, Western blotting, and inflammatory activity cytokine analysis. It was found that <i>Phascolarctobacterium</i> spp. (<i>Phascolarctobacterium faecium</i>), which consumed succinate, significantly decreased SUCNR1 expression, relieved colonic damage, reduced cytokine levels, and restored the integrity of the intestinal epithelial barrier in UC mice. In addition, the results of flow cytometry, quantitative real-time polymerase chain reaction, and enzyme-linked immunosorbent assay indicated that succinate deficiency markedly suppressed secretion of proinflammatory cytokines (e.g., interleukin-1β, interleukin-6, interleukin-10, and tumor necrosis factor-α). Moreover, the SUCNR1 inhibitor (NF-56-EJ40) inhibited glycolysis of intestinal epithelial cells (IECs) in the coculture system with Th17 cells, including downregulation of oxygen consumption rate and increased extracellular acidification rate reflecting overall glycolytic flux, and regulated the expression of glycolysis-related proteins, such as GLUT1, HK-II, and LDHA. Collectively, our findings indicate that microbiota consumption of succinate can ameliorate DSS-induced UC through suppressing Th17, reducing IEC glycolysis, lowering the secretion of proinflammatory cytokines, maintaining epithelial barrier function, and improving dysbiosis.<b>NEW & NOTEWORTHY</b> The gut microbiota contributes to host physiology through the production of a myriad of metabolites. Owing to the high degree of cross talk both within and between biological kingdoms, metabolite-focused research has identified multiple actionable microbial targets that are relevant for host health. In this study, we demonstrated that the microbiota consumption of succinate can alleviate DSS-induced UC in mice responses by modulating glycolytic metabolism through the SUCNR1/NF-κB signaling pathway.</p>","PeriodicalId":7585,"journal":{"name":"American journal of physiology. Cell physiology","volume":" ","pages":"C440-C454"},"PeriodicalIF":4.7000,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"American journal of physiology. Cell physiology","FirstCategoryId":"99","ListUrlMain":"https://doi.org/10.1152/ajpcell.00411.2025","RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2025/6/23 0:00:00","PubModel":"Epub","JCR":"Q2","JCRName":"CELL BIOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Ulcerative colitis (UC) is a chronic recurrent inflammatory disease. Previous studies demonstrate that excessive accumulation of gut microbial metabolites, especially succinate, increases the risk of disease progression. However, the role of succinate and its molecular mechanism have not been explored. We investigated the effects of succinate on colonic inflammation and intestinal microbiota and their association with succinate receptor (SUCNR1) signaling in 3% dextran sodium sulfate (DSS)-induced acute UC in C57BL/6J mice. After treatment, fecal bacteria from UC mice were evaluated by 16S rRNA sequencing. Colon tissues and cell lysates were collected and prepared for histological evaluation, immunohistochemistry, Western blotting, and inflammatory activity cytokine analysis. It was found that Phascolarctobacterium spp. (Phascolarctobacterium faecium), which consumed succinate, significantly decreased SUCNR1 expression, relieved colonic damage, reduced cytokine levels, and restored the integrity of the intestinal epithelial barrier in UC mice. In addition, the results of flow cytometry, quantitative real-time polymerase chain reaction, and enzyme-linked immunosorbent assay indicated that succinate deficiency markedly suppressed secretion of proinflammatory cytokines (e.g., interleukin-1β, interleukin-6, interleukin-10, and tumor necrosis factor-α). Moreover, the SUCNR1 inhibitor (NF-56-EJ40) inhibited glycolysis of intestinal epithelial cells (IECs) in the coculture system with Th17 cells, including downregulation of oxygen consumption rate and increased extracellular acidification rate reflecting overall glycolytic flux, and regulated the expression of glycolysis-related proteins, such as GLUT1, HK-II, and LDHA. Collectively, our findings indicate that microbiota consumption of succinate can ameliorate DSS-induced UC through suppressing Th17, reducing IEC glycolysis, lowering the secretion of proinflammatory cytokines, maintaining epithelial barrier function, and improving dysbiosis.NEW & NOTEWORTHY The gut microbiota contributes to host physiology through the production of a myriad of metabolites. Owing to the high degree of cross talk both within and between biological kingdoms, metabolite-focused research has identified multiple actionable microbial targets that are relevant for host health. In this study, we demonstrated that the microbiota consumption of succinate can alleviate DSS-induced UC in mice responses by modulating glycolytic metabolism through the SUCNR1/NF-κB signaling pathway.

肠道微生物组通过SUCNR1/NF-κB信号通路促进糖酵解,促进琥珀酸诱导的溃疡性结肠炎。
溃疡性结肠炎(UC)是一种慢性复发性炎性疾病。先前的研究表明,肠道微生物代谢物,特别是琥珀酸盐的过度积累会增加疾病进展的风险。然而,琥珀酸盐的作用及其分子机制尚未深入研究。我们研究了琥珀酸盐对3%葡聚糖硫酸钠(DSS)诱导的C57BL/6J小鼠急性UC的结肠炎症和肠道微生物群的影响,以及它们与琥珀酸盐受体(SUCNR1)信号传导的关系。治疗后,采用16S rRNA测序对UC小鼠粪便细菌进行评价。收集结肠组织和细胞裂解物并准备用于组织学评估、免疫组织化学、Western blotting和炎症活性细胞因子分析。研究发现,消耗琥珀酸盐的Phascolarctobacterium spp. (p.p urbacterium)显著降低UC小鼠的SUCNR1表达,减轻结肠损伤,降低细胞因子水平,恢复肠上皮屏障的完整性。此外,流式细胞术、实时定量聚合酶链反应和酶联免疫吸附实验结果显示,琥珀酸缺乏显著抑制促炎细胞因子(如白细胞介素-1β、白细胞介素-6、白细胞介素-10和肿瘤坏死因子-α)的分泌。此外,SUCNR1抑制剂(NF-56-EJ40)在与Th17细胞共培养体系中抑制肠上皮细胞(IECs)的糖酵解,包括下调耗氧率(OCR)和增加反映糖酵解总通量的细胞外酸化率(ECAR),并调节糖酵解相关蛋白如GLUT1、HK-II和LDHA的表达。总之,我们的研究结果表明,琥珀酸盐的微生物群消耗可以通过抑制Th17、减少IECs糖酵解、降低促炎细胞因子的分泌、维持上皮屏障功能和改善生态失调来改善dss诱导的UC。
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
CiteScore
9.10
自引率
1.80%
发文量
252
审稿时长
1 months
期刊介绍: The American Journal of Physiology-Cell Physiology is dedicated to innovative approaches to the study of cell and molecular physiology. Contributions that use cellular and molecular approaches to shed light on mechanisms of physiological control at higher levels of organization also appear regularly. Manuscripts dealing with the structure and function of cell membranes, contractile systems, cellular organelles, and membrane channels, transporters, and pumps are encouraged. Studies dealing with integrated regulation of cellular function, including mechanisms of signal transduction, development, gene expression, cell-to-cell interactions, and the cell physiology of pathophysiological states, are also eagerly sought. Interdisciplinary studies that apply the approaches of biochemistry, biophysics, molecular biology, morphology, and immunology to the determination of new principles in cell physiology are especially welcome.
×
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:604180095
Book学术官方微信