Protective effect of zinc gluconate on intestinal mucosal barrier injury in antibiotics and LPS-induced mice

Yongcai Wang, Juan Xiao, Sumei Wei, Ying Su, Xia Yang, Shiqi Su, Liancheng Lan, Xiuqi Chen, Ting Huang, Qing-wen Shan
{"title":"Protective effect of zinc gluconate on intestinal mucosal barrier injury in antibiotics and LPS-induced mice","authors":"Yongcai Wang, Juan Xiao, Sumei Wei, Ying Su, Xia Yang, Shiqi Su, Liancheng Lan, Xiuqi Chen, Ting Huang, Qing-wen Shan","doi":"10.3389/fmicb.2024.1407091","DOIUrl":null,"url":null,"abstract":"The aim of the study is to investigate the function and mechanism of Zinc Gluconate (ZG) on intestinal mucosal barrier damage in antibiotics and Lipopolysaccharide (LPS)-induced mice.We established a composite mouse model by inducing intestinal mucosal barrier damage using antibiotics and LPS. The animals were divided into five groups: Control (normal and model) and experimental (low, medium, and high-dose ZG treatments). We evaluated the intestinal mucosal barrier using various methods, including monitoring body weight and fecal changes, assessing pathological damage and ultrastructure of the mouse ileum, analyzing expression levels of tight junction (TJ)-related proteins and genes, confirming the TLR4/NF-κB signaling pathway, and examining the structure of the intestinal flora.In mice, the dual induction of antibiotics and LPS led to weight loss, fecal abnormalities, disruption of ileocecal mucosal structure, increased intestinal barrier permeability, and disorganization of the microbiota structure. ZG restored body weight, alleviated diarrheal symptoms and pathological damage, and maintained the structural integrity of intestinal epithelial cells (IECs). Additionally, ZG reduced intestinal mucosal permeability by upregulating TJ-associated proteins (ZO-1, Occludin, Claudin-1, and JAM-A) and downregulating MLCK, thereby repairing intestinal mucosal barrier damage induced by dual induction of antibiotics and LPS. Moreover, ZG suppressed the TLR4/NF-κB signaling pathway, demonstrating anti-inflammatory properties and preserving barrier integrity. Furthermore, ZG restored gut microbiota diversity and richness, evidenced by increased Shannon and Observed features indices, and decreased Simpson’s index. ZG also modulated the relative abundance of beneficial human gut bacteria (Bacteroidetes, Firmicutes, Verrucomicrobia, Parabacteroides, Lactobacillus, and Akkermansia) and harmful bacteria (Proteobacteria and Enterobacter), repairing the damage induced by dual administration of antibiotics and LPS.ZG attenuates the dual induction of antibiotics and LPS-induced intestinal barrier damage and also protects the intestinal barrier function in mice.","PeriodicalId":509565,"journal":{"name":"Frontiers in Microbiology","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2024-05-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Frontiers in Microbiology","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.3389/fmicb.2024.1407091","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

Abstract

The aim of the study is to investigate the function and mechanism of Zinc Gluconate (ZG) on intestinal mucosal barrier damage in antibiotics and Lipopolysaccharide (LPS)-induced mice.We established a composite mouse model by inducing intestinal mucosal barrier damage using antibiotics and LPS. The animals were divided into five groups: Control (normal and model) and experimental (low, medium, and high-dose ZG treatments). We evaluated the intestinal mucosal barrier using various methods, including monitoring body weight and fecal changes, assessing pathological damage and ultrastructure of the mouse ileum, analyzing expression levels of tight junction (TJ)-related proteins and genes, confirming the TLR4/NF-κB signaling pathway, and examining the structure of the intestinal flora.In mice, the dual induction of antibiotics and LPS led to weight loss, fecal abnormalities, disruption of ileocecal mucosal structure, increased intestinal barrier permeability, and disorganization of the microbiota structure. ZG restored body weight, alleviated diarrheal symptoms and pathological damage, and maintained the structural integrity of intestinal epithelial cells (IECs). Additionally, ZG reduced intestinal mucosal permeability by upregulating TJ-associated proteins (ZO-1, Occludin, Claudin-1, and JAM-A) and downregulating MLCK, thereby repairing intestinal mucosal barrier damage induced by dual induction of antibiotics and LPS. Moreover, ZG suppressed the TLR4/NF-κB signaling pathway, demonstrating anti-inflammatory properties and preserving barrier integrity. Furthermore, ZG restored gut microbiota diversity and richness, evidenced by increased Shannon and Observed features indices, and decreased Simpson’s index. ZG also modulated the relative abundance of beneficial human gut bacteria (Bacteroidetes, Firmicutes, Verrucomicrobia, Parabacteroides, Lactobacillus, and Akkermansia) and harmful bacteria (Proteobacteria and Enterobacter), repairing the damage induced by dual administration of antibiotics and LPS.ZG attenuates the dual induction of antibiotics and LPS-induced intestinal barrier damage and also protects the intestinal barrier function in mice.
葡萄糖酸锌对抗生素和 LPS 诱导的小鼠肠粘膜屏障损伤的保护作用
本研究旨在探讨葡萄糖酸锌(ZG)对抗生素和脂多糖(LPS)诱导的小鼠肠粘膜屏障损伤的作用和机制。动物被分为五组:对照组(正常组和模型组)和实验组(低、中、高剂量 ZG 处理组)。我们采用多种方法评估肠粘膜屏障,包括监测体重和粪便变化、评估小鼠回肠的病理损伤和超微结构、分析紧密连接(TJ)相关蛋白和基因的表达水平、确认 TLR4/NF-κB 信号通路以及检查肠道菌群结构。在小鼠体内,抗生素和 LPS 的双重诱导导致体重下降、粪便异常、回盲部粘膜结构破坏、肠屏障通透性增加以及微生物群结构紊乱。ZG 可恢复体重,减轻腹泻症状和病理损伤,并保持肠上皮细胞(IECs)结构的完整性。此外,ZG 还通过上调 TJ 相关蛋白(ZO-1、Occludin、Claudin-1 和 JAM-A)和下调 MLCK 降低了肠粘膜通透性,从而修复了抗生素和 LPS 双重诱导引起的肠粘膜屏障损伤。此外,ZG 还能抑制 TLR4/NF-κB 信号通路,显示出抗炎特性并保护屏障完整性。此外,ZG 还恢复了肠道微生物群的多样性和丰富度,这体现在香农指数和观察特征指数的增加以及辛普森指数的降低。ZG 还调节了人类肠道有益菌(类杆菌、固执菌、疣状芽胞杆菌、副乳杆菌、乳酸杆菌和 Akkermansia)和有害菌(变形杆菌和肠杆菌)的相对丰度,修复了抗生素和 LPS 双重给药诱导的损伤。
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
自引率
0.00%
发文量
0
×
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