Cargando…

Muc2 mucin o-glycosylation interacts with enteropathogenic Escherichia coli to influence the development of ulcerative colitis based on the NF-kB signaling pathway

BACKGROUND: Ulcerative colitis (UC) is a chronic inflammatory disease of the intestine characterized by a compromised intestinal epithelial barrier. Mucin glycans are crucial in preserving barrier function during bacterial infections, although the underlying mechanisms remain largely unexplored. MET...

Descripción completa

Detalles Bibliográficos
Autores principales: Wei, Juan, Chen, Chunyan, Feng, Jing, Zhou, Shuping, Feng, Xiaoyue, Yang, Zhao, Lu, Heng, Tao, Hui, Li, Liuying, Xv, Huabing, Xuan, Ji, Wang, Fangyu
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10631195/
https://www.ncbi.nlm.nih.gov/pubmed/37940996
http://dx.doi.org/10.1186/s12967-023-04687-2
_version_ 1785132321144307712
author Wei, Juan
Chen, Chunyan
Feng, Jing
Zhou, Shuping
Feng, Xiaoyue
Yang, Zhao
Lu, Heng
Tao, Hui
Li, Liuying
Xv, Huabing
Xuan, Ji
Wang, Fangyu
author_facet Wei, Juan
Chen, Chunyan
Feng, Jing
Zhou, Shuping
Feng, Xiaoyue
Yang, Zhao
Lu, Heng
Tao, Hui
Li, Liuying
Xv, Huabing
Xuan, Ji
Wang, Fangyu
author_sort Wei, Juan
collection PubMed
description BACKGROUND: Ulcerative colitis (UC) is a chronic inflammatory disease of the intestine characterized by a compromised intestinal epithelial barrier. Mucin glycans are crucial in preserving barrier function during bacterial infections, although the underlying mechanisms remain largely unexplored. METHODS: A cohort comprising 15 patients diagnosed with UC and 15 healthy individuals was recruited. Stool samples were collected to perform 16S rRNA gene sequencing, while biopsy samples were subjected to nanocapillary liquid chromatography-tandem mass spectrometry (nanoLC-MS/MS) to assess O-glycosylation. Gene expression was evaluated through qPCR analysis and Western blotting. Furthermore, animal experiments were conducted to investigate the effects of Escherichia coli and/or O-glycan inhibitor benzyl-α-GalNAc on the development of colitis in mice. RESULTS: Our findings revealed that the mucus barrier was disrupted during the early stages of UC, while the MUC2 protein content remained unaltered. Additionally, a noteworthy reduction in the o-glycosylation of MUC2 was observed, along with significant changes in the intestinal microbiota during the early stages of UC. These changes included a decrease in intestinal species richness and an increase in the abundance of Escherichia coli (E. coli). Moreover, subsequent to the administration of galactose or o-glycan inhibitor to intestinal epithelial cells, it was observed that the cell culture supernatant had the ability to modify the proliferation and adhesive capacity of E. coli. Furthermore, when pathogenic E. coli or commensal E. coli were cocultured with intestinal epithelium, both strains elicited activation of the NF-KB signaling pathway in epithelial cells and facilitated the expression of serine protease in comparison to the untreated control. Consistently, the inhibition of o-glycans has been observed to enhance the pathogenicity of E. coli in vivo. Furthermore, a correlation has been established between the level of o-glycans and the development of ulcerative colitis. Specifically, a reduction in the O-glycan content of MUC2 cells has been found to increase the virulence of E. coli, thereby compromising the integrity of the intestinal epithelial barrier. CONCLUSIONS: Together, there exist complex interactions between the intestinal epithelium, o-glycans, and the intestinal microbiota, which may inform the development of novel therapeutic strategies for the treatment of ulcerative colitis. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12967-023-04687-2.
format Online
Article
Text
id pubmed-10631195
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-106311952023-11-08 Muc2 mucin o-glycosylation interacts with enteropathogenic Escherichia coli to influence the development of ulcerative colitis based on the NF-kB signaling pathway Wei, Juan Chen, Chunyan Feng, Jing Zhou, Shuping Feng, Xiaoyue Yang, Zhao Lu, Heng Tao, Hui Li, Liuying Xv, Huabing Xuan, Ji Wang, Fangyu J Transl Med Research BACKGROUND: Ulcerative colitis (UC) is a chronic inflammatory disease of the intestine characterized by a compromised intestinal epithelial barrier. Mucin glycans are crucial in preserving barrier function during bacterial infections, although the underlying mechanisms remain largely unexplored. METHODS: A cohort comprising 15 patients diagnosed with UC and 15 healthy individuals was recruited. Stool samples were collected to perform 16S rRNA gene sequencing, while biopsy samples were subjected to nanocapillary liquid chromatography-tandem mass spectrometry (nanoLC-MS/MS) to assess O-glycosylation. Gene expression was evaluated through qPCR analysis and Western blotting. Furthermore, animal experiments were conducted to investigate the effects of Escherichia coli and/or O-glycan inhibitor benzyl-α-GalNAc on the development of colitis in mice. RESULTS: Our findings revealed that the mucus barrier was disrupted during the early stages of UC, while the MUC2 protein content remained unaltered. Additionally, a noteworthy reduction in the o-glycosylation of MUC2 was observed, along with significant changes in the intestinal microbiota during the early stages of UC. These changes included a decrease in intestinal species richness and an increase in the abundance of Escherichia coli (E. coli). Moreover, subsequent to the administration of galactose or o-glycan inhibitor to intestinal epithelial cells, it was observed that the cell culture supernatant had the ability to modify the proliferation and adhesive capacity of E. coli. Furthermore, when pathogenic E. coli or commensal E. coli were cocultured with intestinal epithelium, both strains elicited activation of the NF-KB signaling pathway in epithelial cells and facilitated the expression of serine protease in comparison to the untreated control. Consistently, the inhibition of o-glycans has been observed to enhance the pathogenicity of E. coli in vivo. Furthermore, a correlation has been established between the level of o-glycans and the development of ulcerative colitis. Specifically, a reduction in the O-glycan content of MUC2 cells has been found to increase the virulence of E. coli, thereby compromising the integrity of the intestinal epithelial barrier. CONCLUSIONS: Together, there exist complex interactions between the intestinal epithelium, o-glycans, and the intestinal microbiota, which may inform the development of novel therapeutic strategies for the treatment of ulcerative colitis. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12967-023-04687-2. BioMed Central 2023-11-08 /pmc/articles/PMC10631195/ /pubmed/37940996 http://dx.doi.org/10.1186/s12967-023-04687-2 Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Wei, Juan
Chen, Chunyan
Feng, Jing
Zhou, Shuping
Feng, Xiaoyue
Yang, Zhao
Lu, Heng
Tao, Hui
Li, Liuying
Xv, Huabing
Xuan, Ji
Wang, Fangyu
Muc2 mucin o-glycosylation interacts with enteropathogenic Escherichia coli to influence the development of ulcerative colitis based on the NF-kB signaling pathway
title Muc2 mucin o-glycosylation interacts with enteropathogenic Escherichia coli to influence the development of ulcerative colitis based on the NF-kB signaling pathway
title_full Muc2 mucin o-glycosylation interacts with enteropathogenic Escherichia coli to influence the development of ulcerative colitis based on the NF-kB signaling pathway
title_fullStr Muc2 mucin o-glycosylation interacts with enteropathogenic Escherichia coli to influence the development of ulcerative colitis based on the NF-kB signaling pathway
title_full_unstemmed Muc2 mucin o-glycosylation interacts with enteropathogenic Escherichia coli to influence the development of ulcerative colitis based on the NF-kB signaling pathway
title_short Muc2 mucin o-glycosylation interacts with enteropathogenic Escherichia coli to influence the development of ulcerative colitis based on the NF-kB signaling pathway
title_sort muc2 mucin o-glycosylation interacts with enteropathogenic escherichia coli to influence the development of ulcerative colitis based on the nf-kb signaling pathway
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10631195/
https://www.ncbi.nlm.nih.gov/pubmed/37940996
http://dx.doi.org/10.1186/s12967-023-04687-2
work_keys_str_mv AT weijuan muc2mucinoglycosylationinteractswithenteropathogenicescherichiacolitoinfluencethedevelopmentofulcerativecolitisbasedonthenfkbsignalingpathway
AT chenchunyan muc2mucinoglycosylationinteractswithenteropathogenicescherichiacolitoinfluencethedevelopmentofulcerativecolitisbasedonthenfkbsignalingpathway
AT fengjing muc2mucinoglycosylationinteractswithenteropathogenicescherichiacolitoinfluencethedevelopmentofulcerativecolitisbasedonthenfkbsignalingpathway
AT zhoushuping muc2mucinoglycosylationinteractswithenteropathogenicescherichiacolitoinfluencethedevelopmentofulcerativecolitisbasedonthenfkbsignalingpathway
AT fengxiaoyue muc2mucinoglycosylationinteractswithenteropathogenicescherichiacolitoinfluencethedevelopmentofulcerativecolitisbasedonthenfkbsignalingpathway
AT yangzhao muc2mucinoglycosylationinteractswithenteropathogenicescherichiacolitoinfluencethedevelopmentofulcerativecolitisbasedonthenfkbsignalingpathway
AT luheng muc2mucinoglycosylationinteractswithenteropathogenicescherichiacolitoinfluencethedevelopmentofulcerativecolitisbasedonthenfkbsignalingpathway
AT taohui muc2mucinoglycosylationinteractswithenteropathogenicescherichiacolitoinfluencethedevelopmentofulcerativecolitisbasedonthenfkbsignalingpathway
AT liliuying muc2mucinoglycosylationinteractswithenteropathogenicescherichiacolitoinfluencethedevelopmentofulcerativecolitisbasedonthenfkbsignalingpathway
AT xvhuabing muc2mucinoglycosylationinteractswithenteropathogenicescherichiacolitoinfluencethedevelopmentofulcerativecolitisbasedonthenfkbsignalingpathway
AT xuanji muc2mucinoglycosylationinteractswithenteropathogenicescherichiacolitoinfluencethedevelopmentofulcerativecolitisbasedonthenfkbsignalingpathway
AT wangfangyu muc2mucinoglycosylationinteractswithenteropathogenicescherichiacolitoinfluencethedevelopmentofulcerativecolitisbasedonthenfkbsignalingpathway