Cargando…

Effects of Oligosaccharides From Morinda officinalis on Gut Microbiota and Metabolome of APP/PS1 Transgenic Mice

Alzheimer's disease (AD), a progressive neurodegenerative disorder, lacks preclinical diagnostic biomarkers and therapeutic drugs. Thus, earlier intervention in AD is a top priority. Studies have shown that the gut microbiota influences central nervous system disorders and that prebiotics can i...

Descripción completa

Detalles Bibliográficos
Autores principales: Xin, Yang, Diling, Chen, Jian, Yang, Ting, Liu, Guoyan, Hu, Hualun, Liang, Xiaocui, Tang, Guoxiao, Lai, Ou, Shuai, Chaoqun, Zheng, Jun, Zhao, Yizhen, Xie
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6013575/
https://www.ncbi.nlm.nih.gov/pubmed/29962999
http://dx.doi.org/10.3389/fneur.2018.00412
_version_ 1783334042869956608
author Xin, Yang
Diling, Chen
Jian, Yang
Ting, Liu
Guoyan, Hu
Hualun, Liang
Xiaocui, Tang
Guoxiao, Lai
Ou, Shuai
Chaoqun, Zheng
Jun, Zhao
Yizhen, Xie
author_facet Xin, Yang
Diling, Chen
Jian, Yang
Ting, Liu
Guoyan, Hu
Hualun, Liang
Xiaocui, Tang
Guoxiao, Lai
Ou, Shuai
Chaoqun, Zheng
Jun, Zhao
Yizhen, Xie
author_sort Xin, Yang
collection PubMed
description Alzheimer's disease (AD), a progressive neurodegenerative disorder, lacks preclinical diagnostic biomarkers and therapeutic drugs. Thus, earlier intervention in AD is a top priority. Studies have shown that the gut microbiota influences central nervous system disorders and that prebiotics can improve the cognition of hosts with AD, but these effects are not well understood. Preliminary research has shown that oligosaccharides from Morinda officinalis (OMO) are a useful prebiotic and cause substantial memory improvements in animal models of AD; however, the mechanism is still unclear. Therefore, this study was conducted to investigate whether OMO are clinically effective in alleviating AD by improving gut microbiota. OMO were administered to APP/PS1 transgenic mice, and potential clinical biomarkers of AD were identified with metabolomics and bioinformatics. Behavioral experiments demonstrated that OMO significantly ameliorated the memory of the AD animal model. Histological changes indicated that OMO ameliorated brain tissue swelling and neuronal apoptosis and downregulated the expression of the intracellular AD marker Aβ(1−42). 16S rRNA sequencing analyses indicated that OMO maintained the diversity and stability of the microbial community. The data also indicated that OMO are an efficacious prebiotic in an animal model of AD, regulating the composition and metabolism of the gut microbiota. A serum metabolomics assay was performed using UHPLC-LTQ Orbitrap mass spectrometry to delineate the metabolic changes and potential early biomarkers in APP/PS1 transgenic mice. Multivariate statistical analysis showed that 14 metabolites were significantly upregulated, and 8 metabolites were downregulated in the model animals compared to the normal controls. Thus, key metabolites represent early indicators of the development of AD. Overall, we report a drug and signaling pathway with therapeutic potential, including proteins associated with cognitive deficits in normal mice or gene mutations that cause AD.
format Online
Article
Text
id pubmed-6013575
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-60135752018-06-29 Effects of Oligosaccharides From Morinda officinalis on Gut Microbiota and Metabolome of APP/PS1 Transgenic Mice Xin, Yang Diling, Chen Jian, Yang Ting, Liu Guoyan, Hu Hualun, Liang Xiaocui, Tang Guoxiao, Lai Ou, Shuai Chaoqun, Zheng Jun, Zhao Yizhen, Xie Front Neurol Neurology Alzheimer's disease (AD), a progressive neurodegenerative disorder, lacks preclinical diagnostic biomarkers and therapeutic drugs. Thus, earlier intervention in AD is a top priority. Studies have shown that the gut microbiota influences central nervous system disorders and that prebiotics can improve the cognition of hosts with AD, but these effects are not well understood. Preliminary research has shown that oligosaccharides from Morinda officinalis (OMO) are a useful prebiotic and cause substantial memory improvements in animal models of AD; however, the mechanism is still unclear. Therefore, this study was conducted to investigate whether OMO are clinically effective in alleviating AD by improving gut microbiota. OMO were administered to APP/PS1 transgenic mice, and potential clinical biomarkers of AD were identified with metabolomics and bioinformatics. Behavioral experiments demonstrated that OMO significantly ameliorated the memory of the AD animal model. Histological changes indicated that OMO ameliorated brain tissue swelling and neuronal apoptosis and downregulated the expression of the intracellular AD marker Aβ(1−42). 16S rRNA sequencing analyses indicated that OMO maintained the diversity and stability of the microbial community. The data also indicated that OMO are an efficacious prebiotic in an animal model of AD, regulating the composition and metabolism of the gut microbiota. A serum metabolomics assay was performed using UHPLC-LTQ Orbitrap mass spectrometry to delineate the metabolic changes and potential early biomarkers in APP/PS1 transgenic mice. Multivariate statistical analysis showed that 14 metabolites were significantly upregulated, and 8 metabolites were downregulated in the model animals compared to the normal controls. Thus, key metabolites represent early indicators of the development of AD. Overall, we report a drug and signaling pathway with therapeutic potential, including proteins associated with cognitive deficits in normal mice or gene mutations that cause AD. Frontiers Media S.A. 2018-06-15 /pmc/articles/PMC6013575/ /pubmed/29962999 http://dx.doi.org/10.3389/fneur.2018.00412 Text en Copyright © 2018 Xin, Diling, Jian, Ting, Guoyan, Hualun, Xiaocui, Guoxiao, Ou, Chaoqun, Jun and Yizhen. http://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 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 Neurology
Xin, Yang
Diling, Chen
Jian, Yang
Ting, Liu
Guoyan, Hu
Hualun, Liang
Xiaocui, Tang
Guoxiao, Lai
Ou, Shuai
Chaoqun, Zheng
Jun, Zhao
Yizhen, Xie
Effects of Oligosaccharides From Morinda officinalis on Gut Microbiota and Metabolome of APP/PS1 Transgenic Mice
title Effects of Oligosaccharides From Morinda officinalis on Gut Microbiota and Metabolome of APP/PS1 Transgenic Mice
title_full Effects of Oligosaccharides From Morinda officinalis on Gut Microbiota and Metabolome of APP/PS1 Transgenic Mice
title_fullStr Effects of Oligosaccharides From Morinda officinalis on Gut Microbiota and Metabolome of APP/PS1 Transgenic Mice
title_full_unstemmed Effects of Oligosaccharides From Morinda officinalis on Gut Microbiota and Metabolome of APP/PS1 Transgenic Mice
title_short Effects of Oligosaccharides From Morinda officinalis on Gut Microbiota and Metabolome of APP/PS1 Transgenic Mice
title_sort effects of oligosaccharides from morinda officinalis on gut microbiota and metabolome of app/ps1 transgenic mice
topic Neurology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6013575/
https://www.ncbi.nlm.nih.gov/pubmed/29962999
http://dx.doi.org/10.3389/fneur.2018.00412
work_keys_str_mv AT xinyang effectsofoligosaccharidesfrommorindaofficinalisongutmicrobiotaandmetabolomeofappps1transgenicmice
AT dilingchen effectsofoligosaccharidesfrommorindaofficinalisongutmicrobiotaandmetabolomeofappps1transgenicmice
AT jianyang effectsofoligosaccharidesfrommorindaofficinalisongutmicrobiotaandmetabolomeofappps1transgenicmice
AT tingliu effectsofoligosaccharidesfrommorindaofficinalisongutmicrobiotaandmetabolomeofappps1transgenicmice
AT guoyanhu effectsofoligosaccharidesfrommorindaofficinalisongutmicrobiotaandmetabolomeofappps1transgenicmice
AT hualunliang effectsofoligosaccharidesfrommorindaofficinalisongutmicrobiotaandmetabolomeofappps1transgenicmice
AT xiaocuitang effectsofoligosaccharidesfrommorindaofficinalisongutmicrobiotaandmetabolomeofappps1transgenicmice
AT guoxiaolai effectsofoligosaccharidesfrommorindaofficinalisongutmicrobiotaandmetabolomeofappps1transgenicmice
AT oushuai effectsofoligosaccharidesfrommorindaofficinalisongutmicrobiotaandmetabolomeofappps1transgenicmice
AT chaoqunzheng effectsofoligosaccharidesfrommorindaofficinalisongutmicrobiotaandmetabolomeofappps1transgenicmice
AT junzhao effectsofoligosaccharidesfrommorindaofficinalisongutmicrobiotaandmetabolomeofappps1transgenicmice
AT yizhenxie effectsofoligosaccharidesfrommorindaofficinalisongutmicrobiotaandmetabolomeofappps1transgenicmice