Cargando…

Role of the Gut Microbiome in Modulating Arthritis Progression in Mice

Genetics alone cannot explain most cases of rheumatoid arthritis (RA). Thus, investigating environmental factors such as the gut microbiota may provide new insights into the initiation and progression of RA. In this study, we performed 16S rRNA sequencing to characterise the gut microbiota of DBA1 m...

Descripción completa

Detalles Bibliográficos
Autores principales: Liu, Xiaofei, Zeng, Benhua, Zhang, Juan, Li, Wenxia, Mou, Fangxiang, Wang, Heng, Zou, Qinghua, Zhong, Bing, Wu, Like, Wei, Hong, Fang, Yongfei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4969881/
https://www.ncbi.nlm.nih.gov/pubmed/27481047
http://dx.doi.org/10.1038/srep30594
_version_ 1782445862879756288
author Liu, Xiaofei
Zeng, Benhua
Zhang, Juan
Li, Wenxia
Mou, Fangxiang
Wang, Heng
Zou, Qinghua
Zhong, Bing
Wu, Like
Wei, Hong
Fang, Yongfei
author_facet Liu, Xiaofei
Zeng, Benhua
Zhang, Juan
Li, Wenxia
Mou, Fangxiang
Wang, Heng
Zou, Qinghua
Zhong, Bing
Wu, Like
Wei, Hong
Fang, Yongfei
author_sort Liu, Xiaofei
collection PubMed
description Genetics alone cannot explain most cases of rheumatoid arthritis (RA). Thus, investigating environmental factors such as the gut microbiota may provide new insights into the initiation and progression of RA. In this study, we performed 16S rRNA sequencing to characterise the gut microbiota of DBA1 mice that did or did not develop arthritis after induction with collagen. We found that divergence in the distribution of microbiota after induction was pronounced and significant. Mice susceptible to collagen-induced arthritis (CIA) showed enriched operational taxonomic units (OTUs) affiliated with the genus Lactobacillus as the dominant genus prior to arthritis onset. With disease development, the abundance of OTUs affiliated with the families Bacteroidaceae, Lachnospiraceae, and S24-7 increased significantly in CIA-susceptible mice. Notably, germ-free mice conventionalized with the microbiota from CIA-susceptible mice showed a higher frequency of arthritis induction than those conventionalized with the microbiota from CIA-resistant mice. Consistently, the concentration of the cytokine interleukin-17 in serum and the proportions of CD8+T cells and Th17 lymphocytes in the spleen were significantly higher in the former group, whereas the abundances of dendritic cells, B cells, and Treg cells in the spleen were significantly lower. Our results suggest that the gut microbiome influences arthritis susceptibility.
format Online
Article
Text
id pubmed-4969881
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher Nature Publishing Group
record_format MEDLINE/PubMed
spelling pubmed-49698812016-08-11 Role of the Gut Microbiome in Modulating Arthritis Progression in Mice Liu, Xiaofei Zeng, Benhua Zhang, Juan Li, Wenxia Mou, Fangxiang Wang, Heng Zou, Qinghua Zhong, Bing Wu, Like Wei, Hong Fang, Yongfei Sci Rep Article Genetics alone cannot explain most cases of rheumatoid arthritis (RA). Thus, investigating environmental factors such as the gut microbiota may provide new insights into the initiation and progression of RA. In this study, we performed 16S rRNA sequencing to characterise the gut microbiota of DBA1 mice that did or did not develop arthritis after induction with collagen. We found that divergence in the distribution of microbiota after induction was pronounced and significant. Mice susceptible to collagen-induced arthritis (CIA) showed enriched operational taxonomic units (OTUs) affiliated with the genus Lactobacillus as the dominant genus prior to arthritis onset. With disease development, the abundance of OTUs affiliated with the families Bacteroidaceae, Lachnospiraceae, and S24-7 increased significantly in CIA-susceptible mice. Notably, germ-free mice conventionalized with the microbiota from CIA-susceptible mice showed a higher frequency of arthritis induction than those conventionalized with the microbiota from CIA-resistant mice. Consistently, the concentration of the cytokine interleukin-17 in serum and the proportions of CD8+T cells and Th17 lymphocytes in the spleen were significantly higher in the former group, whereas the abundances of dendritic cells, B cells, and Treg cells in the spleen were significantly lower. Our results suggest that the gut microbiome influences arthritis susceptibility. Nature Publishing Group 2016-08-02 /pmc/articles/PMC4969881/ /pubmed/27481047 http://dx.doi.org/10.1038/srep30594 Text en Copyright © 2016, The Author(s) http://creativecommons.org/licenses/by/4.0/ This work is licensed under a Creative Commons Attribution 4.0 International License. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in the credit line; if the material is not included under the Creative Commons license, users will need to obtain permission from the license holder to reproduce the material. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/
spellingShingle Article
Liu, Xiaofei
Zeng, Benhua
Zhang, Juan
Li, Wenxia
Mou, Fangxiang
Wang, Heng
Zou, Qinghua
Zhong, Bing
Wu, Like
Wei, Hong
Fang, Yongfei
Role of the Gut Microbiome in Modulating Arthritis Progression in Mice
title Role of the Gut Microbiome in Modulating Arthritis Progression in Mice
title_full Role of the Gut Microbiome in Modulating Arthritis Progression in Mice
title_fullStr Role of the Gut Microbiome in Modulating Arthritis Progression in Mice
title_full_unstemmed Role of the Gut Microbiome in Modulating Arthritis Progression in Mice
title_short Role of the Gut Microbiome in Modulating Arthritis Progression in Mice
title_sort role of the gut microbiome in modulating arthritis progression in mice
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4969881/
https://www.ncbi.nlm.nih.gov/pubmed/27481047
http://dx.doi.org/10.1038/srep30594
work_keys_str_mv AT liuxiaofei roleofthegutmicrobiomeinmodulatingarthritisprogressioninmice
AT zengbenhua roleofthegutmicrobiomeinmodulatingarthritisprogressioninmice
AT zhangjuan roleofthegutmicrobiomeinmodulatingarthritisprogressioninmice
AT liwenxia roleofthegutmicrobiomeinmodulatingarthritisprogressioninmice
AT moufangxiang roleofthegutmicrobiomeinmodulatingarthritisprogressioninmice
AT wangheng roleofthegutmicrobiomeinmodulatingarthritisprogressioninmice
AT zouqinghua roleofthegutmicrobiomeinmodulatingarthritisprogressioninmice
AT zhongbing roleofthegutmicrobiomeinmodulatingarthritisprogressioninmice
AT wulike roleofthegutmicrobiomeinmodulatingarthritisprogressioninmice
AT weihong roleofthegutmicrobiomeinmodulatingarthritisprogressioninmice
AT fangyongfei roleofthegutmicrobiomeinmodulatingarthritisprogressioninmice