Rifaximin Alters Intestinal Microbiota and Prevents Progression of Ankylosing Spondylitis in Mice

Recently, accumulating evidence has suggested that gut microbiota may be involved in the occurrence and development of ankylosing spondylitis (AS). It has been suggested that rifaximin have the ability to modulate the gut bacterial communities, prevent inflammatory response, and modulate gut barrier...

Full description

Bibliographic Details
Main Authors: Lianjun Yang, Bin Liu, Junchi Zheng, Jincheng Huang, Qinghao Zhao, Jinshi Liu, Zhihai Su, Min Wang, Zhifei Cui, Tingxuan Wang, Weicong Zhang, Qingchu Li, Hai Lu
Format: Article
Language:English
Published: Frontiers Media S.A. 2019-03-01
Series:Frontiers in Cellular and Infection Microbiology
Subjects:
Online Access:https://www.frontiersin.org/article/10.3389/fcimb.2019.00044/full
id doaj-dbcc7c86d07340a1893d608f78a0769f
record_format Article
collection DOAJ
language English
format Article
sources DOAJ
author Lianjun Yang
Bin Liu
Junchi Zheng
Jincheng Huang
Qinghao Zhao
Jinshi Liu
Zhihai Su
Min Wang
Zhifei Cui
Tingxuan Wang
Weicong Zhang
Qingchu Li
Hai Lu
spellingShingle Lianjun Yang
Bin Liu
Junchi Zheng
Jincheng Huang
Qinghao Zhao
Jinshi Liu
Zhihai Su
Min Wang
Zhifei Cui
Tingxuan Wang
Weicong Zhang
Qingchu Li
Hai Lu
Rifaximin Alters Intestinal Microbiota and Prevents Progression of Ankylosing Spondylitis in Mice
Frontiers in Cellular and Infection Microbiology
ankylosing spondylitis
rifaximin
gut microbiota
inflammatory response
intestinal epithelial barrier
author_facet Lianjun Yang
Bin Liu
Junchi Zheng
Jincheng Huang
Qinghao Zhao
Jinshi Liu
Zhihai Su
Min Wang
Zhifei Cui
Tingxuan Wang
Weicong Zhang
Qingchu Li
Hai Lu
author_sort Lianjun Yang
title Rifaximin Alters Intestinal Microbiota and Prevents Progression of Ankylosing Spondylitis in Mice
title_short Rifaximin Alters Intestinal Microbiota and Prevents Progression of Ankylosing Spondylitis in Mice
title_full Rifaximin Alters Intestinal Microbiota and Prevents Progression of Ankylosing Spondylitis in Mice
title_fullStr Rifaximin Alters Intestinal Microbiota and Prevents Progression of Ankylosing Spondylitis in Mice
title_full_unstemmed Rifaximin Alters Intestinal Microbiota and Prevents Progression of Ankylosing Spondylitis in Mice
title_sort rifaximin alters intestinal microbiota and prevents progression of ankylosing spondylitis in mice
publisher Frontiers Media S.A.
series Frontiers in Cellular and Infection Microbiology
issn 2235-2988
publishDate 2019-03-01
description Recently, accumulating evidence has suggested that gut microbiota may be involved in the occurrence and development of ankylosing spondylitis (AS). It has been suggested that rifaximin have the ability to modulate the gut bacterial communities, prevent inflammatory response, and modulate gut barrier function. The goal of this work is to evaluate the protective effects of rifaximin in fighting AS and to elucidate the potential underlying mechanism. Rifaximin were administered to the proteoglycan (PG)-induced AS mice for 4 consecutive weeks. The disease severity was measured with the clinical and histological of arthritis and spondylitis. Intestinal histopathological, pro-inflammatory cytokine levels and the intestinal mucosal barrier were evaluated. Then, western blot was performed to explore the toll-like receptor 4 (TLR-4) signal transducer and NF-κB expression. Stool samples were collected to analyze the differences in the gut microbiota via next-generation sequencing of 16S rDNA. We found that rifaximin significantly reduced the severity of AS and resulted in down-regulation of inflammatory factors, such as TNF-α, IL-6, IL-17A, and IL-23. Meanwhile, rifaximin prevented ileum histological alterations, restored intestinal barrier function and inhibited TLR-4/NF-κB signaling pathway activation. Rifaximin also changed the gut microbiota composition with increased Bacteroidetes/Firmicutes phylum ratio, as well as selectively promoting some probiotic populations, including Lactobacillales. Our results suggest that rifaximin suppressed progression of AS and regulated gut microbiota in AS mice. Rifaximin might be useful as a novel treatment for AS.
topic ankylosing spondylitis
rifaximin
gut microbiota
inflammatory response
intestinal epithelial barrier
url https://www.frontiersin.org/article/10.3389/fcimb.2019.00044/full
work_keys_str_mv AT lianjunyang rifaximinaltersintestinalmicrobiotaandpreventsprogressionofankylosingspondylitisinmice
AT binliu rifaximinaltersintestinalmicrobiotaandpreventsprogressionofankylosingspondylitisinmice
AT junchizheng rifaximinaltersintestinalmicrobiotaandpreventsprogressionofankylosingspondylitisinmice
AT jinchenghuang rifaximinaltersintestinalmicrobiotaandpreventsprogressionofankylosingspondylitisinmice
AT qinghaozhao rifaximinaltersintestinalmicrobiotaandpreventsprogressionofankylosingspondylitisinmice
AT jinshiliu rifaximinaltersintestinalmicrobiotaandpreventsprogressionofankylosingspondylitisinmice
AT zhihaisu rifaximinaltersintestinalmicrobiotaandpreventsprogressionofankylosingspondylitisinmice
AT minwang rifaximinaltersintestinalmicrobiotaandpreventsprogressionofankylosingspondylitisinmice
AT zhifeicui rifaximinaltersintestinalmicrobiotaandpreventsprogressionofankylosingspondylitisinmice
AT tingxuanwang rifaximinaltersintestinalmicrobiotaandpreventsprogressionofankylosingspondylitisinmice
AT weicongzhang rifaximinaltersintestinalmicrobiotaandpreventsprogressionofankylosingspondylitisinmice
AT qingchuli rifaximinaltersintestinalmicrobiotaandpreventsprogressionofankylosingspondylitisinmice
AT hailu rifaximinaltersintestinalmicrobiotaandpreventsprogressionofankylosingspondylitisinmice
_version_ 1726013489128931328
spelling doaj-dbcc7c86d07340a1893d608f78a0769f2020-11-24T21:17:13ZengFrontiers Media S.A.Frontiers in Cellular and Infection Microbiology2235-29882019-03-01910.3389/fcimb.2019.00044432732Rifaximin Alters Intestinal Microbiota and Prevents Progression of Ankylosing Spondylitis in MiceLianjun Yang0Bin Liu1Junchi Zheng2Jincheng Huang3Qinghao Zhao4Jinshi Liu5Zhihai Su6Min Wang7Zhifei Cui8Tingxuan Wang9Weicong Zhang10Qingchu Li11Hai Lu12Department of Orthopedic Surgery, Orthopaedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, ChinaDepartment of Orthopedic Surgery, Orthopaedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, ChinaDepartment of Orthopedic Surgery, Orthopaedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, ChinaDepartment of Orthopedics, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, ChinaDepartment of Orthopedic Surgery, Orthopaedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, ChinaDepartment of Orthopedic Surgery, Orthopaedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, ChinaDepartment of Orthopedic Surgery, Orthopaedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, ChinaDepartment of Orthopedic Surgery, Orthopaedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, ChinaDepartment of Orthopedic Surgery, Orthopaedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, ChinaDepartment of Orthopedic Surgery, Orthopaedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, ChinaDepartment of Orthopedic Surgery, Orthopaedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, ChinaDepartment of Orthopedic Surgery, Orthopaedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, ChinaDepartment of Orthopedic Surgery, Orthopaedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, ChinaRecently, accumulating evidence has suggested that gut microbiota may be involved in the occurrence and development of ankylosing spondylitis (AS). It has been suggested that rifaximin have the ability to modulate the gut bacterial communities, prevent inflammatory response, and modulate gut barrier function. The goal of this work is to evaluate the protective effects of rifaximin in fighting AS and to elucidate the potential underlying mechanism. Rifaximin were administered to the proteoglycan (PG)-induced AS mice for 4 consecutive weeks. The disease severity was measured with the clinical and histological of arthritis and spondylitis. Intestinal histopathological, pro-inflammatory cytokine levels and the intestinal mucosal barrier were evaluated. Then, western blot was performed to explore the toll-like receptor 4 (TLR-4) signal transducer and NF-κB expression. Stool samples were collected to analyze the differences in the gut microbiota via next-generation sequencing of 16S rDNA. We found that rifaximin significantly reduced the severity of AS and resulted in down-regulation of inflammatory factors, such as TNF-α, IL-6, IL-17A, and IL-23. Meanwhile, rifaximin prevented ileum histological alterations, restored intestinal barrier function and inhibited TLR-4/NF-κB signaling pathway activation. Rifaximin also changed the gut microbiota composition with increased Bacteroidetes/Firmicutes phylum ratio, as well as selectively promoting some probiotic populations, including Lactobacillales. Our results suggest that rifaximin suppressed progression of AS and regulated gut microbiota in AS mice. Rifaximin might be useful as a novel treatment for AS.https://www.frontiersin.org/article/10.3389/fcimb.2019.00044/fullankylosing spondylitisrifaximingut microbiotainflammatory responseintestinal epithelial barrier