Cargando…

Molecular and Microbial Signatures Predictive of Prebiotic Action of Neoagarotetraose in a Dextran Sulfate Sodium-Induced Murine Colitis Model

Neoagarotetraose (NT), a hydrolytic product of agar by β-agarase, is known to possess bioactive properties. However, the mechanisms via which NT alleviates intestinal inflammation remain unknown. In this study, a dextran sulfate sodium (DSS)-induced murine model was developed to evaluate the effect...

Descripción completa

Detalles Bibliográficos
Autores principales: Liu, Fang, Liu, Jianan, Wang, Thomas T.Y., Liu, Zhen, Xue, Changhu, Mao, Xiangzhao, Tang, Qingjuan, Li, Robert W.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7409226/
https://www.ncbi.nlm.nih.gov/pubmed/32635315
http://dx.doi.org/10.3390/microorganisms8070995
_version_ 1783568016793927680
author Liu, Fang
Liu, Jianan
Wang, Thomas T.Y.
Liu, Zhen
Xue, Changhu
Mao, Xiangzhao
Tang, Qingjuan
Li, Robert W.
author_facet Liu, Fang
Liu, Jianan
Wang, Thomas T.Y.
Liu, Zhen
Xue, Changhu
Mao, Xiangzhao
Tang, Qingjuan
Li, Robert W.
author_sort Liu, Fang
collection PubMed
description Neoagarotetraose (NT), a hydrolytic product of agar by β-agarase, is known to possess bioactive properties. However, the mechanisms via which NT alleviates intestinal inflammation remain unknown. In this study, a dextran sulfate sodium (DSS)-induced murine model was developed to evaluate the effect of NT on gut microbiome and microbial metabolism using 16S rRNA gene sequencing and untargeted metabolomics. Our data demonstrate that NT ingestion improved gut integrity and inflammation scores. NT reversed the abundance of Proteobacteria from an elevated level induced by DSS and significantly increased the abundance of Verrucomicrobia. Further, NT significantly increased the abundance of Akkermansia and Lactobacillus and concomitantly decreased that of Sutterella, which were among the important features identified by random forests analysis contributing to classification accuracy for NT supplementation. A microbial signature consisting of Adlercreutzia (denominator) and Turicibacter (numerator) predicted the NT supplementation status. Moreover, NT significantly modulated multiple gut metabolites, particularly those related to histidine, polyamine and tocopherol metabolism. Together, our findings provided novel insights into the mechanisms by which NT modulated the gut microbiome and metabolome and should facilitate the development of NT as a potent prebiotic for colitis management.
format Online
Article
Text
id pubmed-7409226
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-74092262020-08-26 Molecular and Microbial Signatures Predictive of Prebiotic Action of Neoagarotetraose in a Dextran Sulfate Sodium-Induced Murine Colitis Model Liu, Fang Liu, Jianan Wang, Thomas T.Y. Liu, Zhen Xue, Changhu Mao, Xiangzhao Tang, Qingjuan Li, Robert W. Microorganisms Article Neoagarotetraose (NT), a hydrolytic product of agar by β-agarase, is known to possess bioactive properties. However, the mechanisms via which NT alleviates intestinal inflammation remain unknown. In this study, a dextran sulfate sodium (DSS)-induced murine model was developed to evaluate the effect of NT on gut microbiome and microbial metabolism using 16S rRNA gene sequencing and untargeted metabolomics. Our data demonstrate that NT ingestion improved gut integrity and inflammation scores. NT reversed the abundance of Proteobacteria from an elevated level induced by DSS and significantly increased the abundance of Verrucomicrobia. Further, NT significantly increased the abundance of Akkermansia and Lactobacillus and concomitantly decreased that of Sutterella, which were among the important features identified by random forests analysis contributing to classification accuracy for NT supplementation. A microbial signature consisting of Adlercreutzia (denominator) and Turicibacter (numerator) predicted the NT supplementation status. Moreover, NT significantly modulated multiple gut metabolites, particularly those related to histidine, polyamine and tocopherol metabolism. Together, our findings provided novel insights into the mechanisms by which NT modulated the gut microbiome and metabolome and should facilitate the development of NT as a potent prebiotic for colitis management. MDPI 2020-07-03 /pmc/articles/PMC7409226/ /pubmed/32635315 http://dx.doi.org/10.3390/microorganisms8070995 Text en © 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Liu, Fang
Liu, Jianan
Wang, Thomas T.Y.
Liu, Zhen
Xue, Changhu
Mao, Xiangzhao
Tang, Qingjuan
Li, Robert W.
Molecular and Microbial Signatures Predictive of Prebiotic Action of Neoagarotetraose in a Dextran Sulfate Sodium-Induced Murine Colitis Model
title Molecular and Microbial Signatures Predictive of Prebiotic Action of Neoagarotetraose in a Dextran Sulfate Sodium-Induced Murine Colitis Model
title_full Molecular and Microbial Signatures Predictive of Prebiotic Action of Neoagarotetraose in a Dextran Sulfate Sodium-Induced Murine Colitis Model
title_fullStr Molecular and Microbial Signatures Predictive of Prebiotic Action of Neoagarotetraose in a Dextran Sulfate Sodium-Induced Murine Colitis Model
title_full_unstemmed Molecular and Microbial Signatures Predictive of Prebiotic Action of Neoagarotetraose in a Dextran Sulfate Sodium-Induced Murine Colitis Model
title_short Molecular and Microbial Signatures Predictive of Prebiotic Action of Neoagarotetraose in a Dextran Sulfate Sodium-Induced Murine Colitis Model
title_sort molecular and microbial signatures predictive of prebiotic action of neoagarotetraose in a dextran sulfate sodium-induced murine colitis model
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7409226/
https://www.ncbi.nlm.nih.gov/pubmed/32635315
http://dx.doi.org/10.3390/microorganisms8070995
work_keys_str_mv AT liufang molecularandmicrobialsignaturespredictiveofprebioticactionofneoagarotetraoseinadextransulfatesodiuminducedmurinecolitismodel
AT liujianan molecularandmicrobialsignaturespredictiveofprebioticactionofneoagarotetraoseinadextransulfatesodiuminducedmurinecolitismodel
AT wangthomasty molecularandmicrobialsignaturespredictiveofprebioticactionofneoagarotetraoseinadextransulfatesodiuminducedmurinecolitismodel
AT liuzhen molecularandmicrobialsignaturespredictiveofprebioticactionofneoagarotetraoseinadextransulfatesodiuminducedmurinecolitismodel
AT xuechanghu molecularandmicrobialsignaturespredictiveofprebioticactionofneoagarotetraoseinadextransulfatesodiuminducedmurinecolitismodel
AT maoxiangzhao molecularandmicrobialsignaturespredictiveofprebioticactionofneoagarotetraoseinadextransulfatesodiuminducedmurinecolitismodel
AT tangqingjuan molecularandmicrobialsignaturespredictiveofprebioticactionofneoagarotetraoseinadextransulfatesodiuminducedmurinecolitismodel
AT lirobertw molecularandmicrobialsignaturespredictiveofprebioticactionofneoagarotetraoseinadextransulfatesodiuminducedmurinecolitismodel