Cargando…

Oral administration of live combined Bacillus subtilis and Enterococcus faecium alleviates colonic oxidative stress and inflammation in osteoarthritic rats by improving fecal microbiome metabolism and enhancing the colonic barrier

Osteoarthritis (OA) causes intestinal damage. The protective effect of probiotics on the intestine is indeed effective; however, the mechanism of protection against intestinal damage in OA is not clear. In this study, we used meniscal/ligamentous injury (MLI) to mimic OA in rats and explored the col...

Descripción completa

Detalles Bibliográficos
Autores principales: Tang, Jilang, Song, Xiaopeng, Zhao, Mingchao, Chen, Hong, Wang, Yingying, Zhao, Binger, Yu, Shiming, Ma, Tianwen, Gao, Li
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9686382/
https://www.ncbi.nlm.nih.gov/pubmed/36439850
http://dx.doi.org/10.3389/fmicb.2022.1005842
_version_ 1784835734101819392
author Tang, Jilang
Song, Xiaopeng
Zhao, Mingchao
Chen, Hong
Wang, Yingying
Zhao, Binger
Yu, Shiming
Ma, Tianwen
Gao, Li
author_facet Tang, Jilang
Song, Xiaopeng
Zhao, Mingchao
Chen, Hong
Wang, Yingying
Zhao, Binger
Yu, Shiming
Ma, Tianwen
Gao, Li
author_sort Tang, Jilang
collection PubMed
description Osteoarthritis (OA) causes intestinal damage. The protective effect of probiotics on the intestine is indeed effective; however, the mechanism of protection against intestinal damage in OA is not clear. In this study, we used meniscal/ligamentous injury (MLI) to mimic OA in rats and explored the colonic protective effects of Bacillus subtilis and Enterococcus faecium on OA. Our study showed that treatment with B. subtilis and E. faecium attenuated colonic injury and reduced inflammatory and oxidative stress factors in the serum of osteoarthritic rats. α- and ß diversity of the fecal flora were not different among groups; no significant differences were observed in the abundances of taxa at the phylum and genus levels. We observed the presence of the depression-related genera Alistipes and Paraprevotella. Analysis of fecal untargeted metabolism revealed that histamine level was significantly reduced in the colon of OA rats, affecting intestinal function. Compared to that in the control group, the enriched metabolic pathways in the OA group were primarily for energy metabolisms, such as pantothenate and CoA biosynthesis, and beta-alanine metabolism. The treatment group had enriched linoleic acid metabolism, fatty acid biosynthesis, and primary bile acid biosynthesis, which were different from those in the control group. The differences in the metabolic pathways between the treatment and OA groups were more evident, primarily in symptom-related metabolic pathways such as Huntington's disease, spinocerebellar ataxia, energy-related central carbon metabolism in cancer, pantothenate and CoA biosynthesis metabolic pathways, as well as some neurotransmission and amino acid transport, and uptake- and synthesis-related metabolic pathways. On further investigation, we found that B. subtilis and E. faecium treatment enhanced the colonic barrier of OA rats, with elevated expressions of tight junction proteins occludin and Zonula occludens 1 and MUC2 mRNA. Intestinal permeability was reduced, and serum LPS levels were downregulated in the treatment group. B. subtilis and E. faecium also regulated the oxidative stress pathway Keap1/Nrf2, promoted the expression of the downstream protective proteins HO-1 and Gpx4, and reduced intestinal apoptosis. Hence, B. subtilis and E. faecium alleviate colonic oxidative stress and inflammation in OA rats by improving fecal metabolism and enhancing the colonic barrier.
format Online
Article
Text
id pubmed-9686382
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-96863822022-11-25 Oral administration of live combined Bacillus subtilis and Enterococcus faecium alleviates colonic oxidative stress and inflammation in osteoarthritic rats by improving fecal microbiome metabolism and enhancing the colonic barrier Tang, Jilang Song, Xiaopeng Zhao, Mingchao Chen, Hong Wang, Yingying Zhao, Binger Yu, Shiming Ma, Tianwen Gao, Li Front Microbiol Microbiology Osteoarthritis (OA) causes intestinal damage. The protective effect of probiotics on the intestine is indeed effective; however, the mechanism of protection against intestinal damage in OA is not clear. In this study, we used meniscal/ligamentous injury (MLI) to mimic OA in rats and explored the colonic protective effects of Bacillus subtilis and Enterococcus faecium on OA. Our study showed that treatment with B. subtilis and E. faecium attenuated colonic injury and reduced inflammatory and oxidative stress factors in the serum of osteoarthritic rats. α- and ß diversity of the fecal flora were not different among groups; no significant differences were observed in the abundances of taxa at the phylum and genus levels. We observed the presence of the depression-related genera Alistipes and Paraprevotella. Analysis of fecal untargeted metabolism revealed that histamine level was significantly reduced in the colon of OA rats, affecting intestinal function. Compared to that in the control group, the enriched metabolic pathways in the OA group were primarily for energy metabolisms, such as pantothenate and CoA biosynthesis, and beta-alanine metabolism. The treatment group had enriched linoleic acid metabolism, fatty acid biosynthesis, and primary bile acid biosynthesis, which were different from those in the control group. The differences in the metabolic pathways between the treatment and OA groups were more evident, primarily in symptom-related metabolic pathways such as Huntington's disease, spinocerebellar ataxia, energy-related central carbon metabolism in cancer, pantothenate and CoA biosynthesis metabolic pathways, as well as some neurotransmission and amino acid transport, and uptake- and synthesis-related metabolic pathways. On further investigation, we found that B. subtilis and E. faecium treatment enhanced the colonic barrier of OA rats, with elevated expressions of tight junction proteins occludin and Zonula occludens 1 and MUC2 mRNA. Intestinal permeability was reduced, and serum LPS levels were downregulated in the treatment group. B. subtilis and E. faecium also regulated the oxidative stress pathway Keap1/Nrf2, promoted the expression of the downstream protective proteins HO-1 and Gpx4, and reduced intestinal apoptosis. Hence, B. subtilis and E. faecium alleviate colonic oxidative stress and inflammation in OA rats by improving fecal metabolism and enhancing the colonic barrier. Frontiers Media S.A. 2022-11-10 /pmc/articles/PMC9686382/ /pubmed/36439850 http://dx.doi.org/10.3389/fmicb.2022.1005842 Text en Copyright © 2022 Tang, Song, Zhao, Chen, Wang, Zhao, Yu, Ma and Gao. https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Microbiology
Tang, Jilang
Song, Xiaopeng
Zhao, Mingchao
Chen, Hong
Wang, Yingying
Zhao, Binger
Yu, Shiming
Ma, Tianwen
Gao, Li
Oral administration of live combined Bacillus subtilis and Enterococcus faecium alleviates colonic oxidative stress and inflammation in osteoarthritic rats by improving fecal microbiome metabolism and enhancing the colonic barrier
title Oral administration of live combined Bacillus subtilis and Enterococcus faecium alleviates colonic oxidative stress and inflammation in osteoarthritic rats by improving fecal microbiome metabolism and enhancing the colonic barrier
title_full Oral administration of live combined Bacillus subtilis and Enterococcus faecium alleviates colonic oxidative stress and inflammation in osteoarthritic rats by improving fecal microbiome metabolism and enhancing the colonic barrier
title_fullStr Oral administration of live combined Bacillus subtilis and Enterococcus faecium alleviates colonic oxidative stress and inflammation in osteoarthritic rats by improving fecal microbiome metabolism and enhancing the colonic barrier
title_full_unstemmed Oral administration of live combined Bacillus subtilis and Enterococcus faecium alleviates colonic oxidative stress and inflammation in osteoarthritic rats by improving fecal microbiome metabolism and enhancing the colonic barrier
title_short Oral administration of live combined Bacillus subtilis and Enterococcus faecium alleviates colonic oxidative stress and inflammation in osteoarthritic rats by improving fecal microbiome metabolism and enhancing the colonic barrier
title_sort oral administration of live combined bacillus subtilis and enterococcus faecium alleviates colonic oxidative stress and inflammation in osteoarthritic rats by improving fecal microbiome metabolism and enhancing the colonic barrier
topic Microbiology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9686382/
https://www.ncbi.nlm.nih.gov/pubmed/36439850
http://dx.doi.org/10.3389/fmicb.2022.1005842
work_keys_str_mv AT tangjilang oraladministrationoflivecombinedbacillussubtilisandenterococcusfaeciumalleviatescolonicoxidativestressandinflammationinosteoarthriticratsbyimprovingfecalmicrobiomemetabolismandenhancingthecolonicbarrier
AT songxiaopeng oraladministrationoflivecombinedbacillussubtilisandenterococcusfaeciumalleviatescolonicoxidativestressandinflammationinosteoarthriticratsbyimprovingfecalmicrobiomemetabolismandenhancingthecolonicbarrier
AT zhaomingchao oraladministrationoflivecombinedbacillussubtilisandenterococcusfaeciumalleviatescolonicoxidativestressandinflammationinosteoarthriticratsbyimprovingfecalmicrobiomemetabolismandenhancingthecolonicbarrier
AT chenhong oraladministrationoflivecombinedbacillussubtilisandenterococcusfaeciumalleviatescolonicoxidativestressandinflammationinosteoarthriticratsbyimprovingfecalmicrobiomemetabolismandenhancingthecolonicbarrier
AT wangyingying oraladministrationoflivecombinedbacillussubtilisandenterococcusfaeciumalleviatescolonicoxidativestressandinflammationinosteoarthriticratsbyimprovingfecalmicrobiomemetabolismandenhancingthecolonicbarrier
AT zhaobinger oraladministrationoflivecombinedbacillussubtilisandenterococcusfaeciumalleviatescolonicoxidativestressandinflammationinosteoarthriticratsbyimprovingfecalmicrobiomemetabolismandenhancingthecolonicbarrier
AT yushiming oraladministrationoflivecombinedbacillussubtilisandenterococcusfaeciumalleviatescolonicoxidativestressandinflammationinosteoarthriticratsbyimprovingfecalmicrobiomemetabolismandenhancingthecolonicbarrier
AT matianwen oraladministrationoflivecombinedbacillussubtilisandenterococcusfaeciumalleviatescolonicoxidativestressandinflammationinosteoarthriticratsbyimprovingfecalmicrobiomemetabolismandenhancingthecolonicbarrier
AT gaoli oraladministrationoflivecombinedbacillussubtilisandenterococcusfaeciumalleviatescolonicoxidativestressandinflammationinosteoarthriticratsbyimprovingfecalmicrobiomemetabolismandenhancingthecolonicbarrier