Cargando…

Mesoporous silica nanoparticle-encapsulated Bifidobacterium attenuates brain Aβ burden and improves olfactory dysfunction of APP/PS1 mice by nasal delivery

BACKGROUND: Dysbiosis or imbalance of gut microbiota in Alzheimer's disease (AD) affects the production of short-chain fatty acids (SCFAs), whereas exogenous SCFAs supplementation exacerbates brain Aβ burden in APP/PS1 mice. Bifidobacterium is the main producer of SCFAs in the gut flora, but or...

Descripción completa

Detalles Bibliográficos
Autores principales: Liu, Ni, Yang, Changwen, Liang, Xiaohan, Cao, Kai, Xie, Jun, Luo, Qingming, Luo, Haiming
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9547428/
https://www.ncbi.nlm.nih.gov/pubmed/36207740
http://dx.doi.org/10.1186/s12951-022-01642-z
_version_ 1784805262931001344
author Liu, Ni
Yang, Changwen
Liang, Xiaohan
Cao, Kai
Xie, Jun
Luo, Qingming
Luo, Haiming
author_facet Liu, Ni
Yang, Changwen
Liang, Xiaohan
Cao, Kai
Xie, Jun
Luo, Qingming
Luo, Haiming
author_sort Liu, Ni
collection PubMed
description BACKGROUND: Dysbiosis or imbalance of gut microbiota in Alzheimer's disease (AD) affects the production of short-chain fatty acids (SCFAs), whereas exogenous SCFAs supplementation exacerbates brain Aβ burden in APP/PS1 mice. Bifidobacterium is the main producer of SCFAs in the gut flora, but oral administration of Bifidobacterium is ineffective due to strong acids and bile salts in the gastrointestinal tract. Therefore, regulating the levels of SCFAs in the gut is of great significance for AD treatment. METHODS: We investigated the feasibility of intranasal delivery of MSNs-Bifidobacterium (MSNs-Bi) to the gut and their effect on behavior and brain pathology in APP/PS1 mice. RESULTS: Mesoporous silica nanospheres (MSNs) were efficiently immobilized on the surface of Bifidobacterium. After intranasal administration, fluorescence imaging of MSNs-Bi in the abdominal cavity and gastrointestinal tract revealed that intranasally delivered MSNs-Bi could be transported through the brain to the peripheral intestine. Intranasal administration of MSNs-Bi not only inhibited intestinal inflammation and reduced brain Aβ burden but also improved olfactory sensitivity in APP/PS1 mice. CONCLUSIONS: These findings suggested that restoring the balance of the gut microbiome contributes to ameliorating cognitive impairment in AD, and that intranasal administration of MSNs-Bi may be an effective therapeutic strategy for the prevention of AD and intestinal disease. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12951-022-01642-z.
format Online
Article
Text
id pubmed-9547428
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-95474282022-10-09 Mesoporous silica nanoparticle-encapsulated Bifidobacterium attenuates brain Aβ burden and improves olfactory dysfunction of APP/PS1 mice by nasal delivery Liu, Ni Yang, Changwen Liang, Xiaohan Cao, Kai Xie, Jun Luo, Qingming Luo, Haiming J Nanobiotechnology Research BACKGROUND: Dysbiosis or imbalance of gut microbiota in Alzheimer's disease (AD) affects the production of short-chain fatty acids (SCFAs), whereas exogenous SCFAs supplementation exacerbates brain Aβ burden in APP/PS1 mice. Bifidobacterium is the main producer of SCFAs in the gut flora, but oral administration of Bifidobacterium is ineffective due to strong acids and bile salts in the gastrointestinal tract. Therefore, regulating the levels of SCFAs in the gut is of great significance for AD treatment. METHODS: We investigated the feasibility of intranasal delivery of MSNs-Bifidobacterium (MSNs-Bi) to the gut and their effect on behavior and brain pathology in APP/PS1 mice. RESULTS: Mesoporous silica nanospheres (MSNs) were efficiently immobilized on the surface of Bifidobacterium. After intranasal administration, fluorescence imaging of MSNs-Bi in the abdominal cavity and gastrointestinal tract revealed that intranasally delivered MSNs-Bi could be transported through the brain to the peripheral intestine. Intranasal administration of MSNs-Bi not only inhibited intestinal inflammation and reduced brain Aβ burden but also improved olfactory sensitivity in APP/PS1 mice. CONCLUSIONS: These findings suggested that restoring the balance of the gut microbiome contributes to ameliorating cognitive impairment in AD, and that intranasal administration of MSNs-Bi may be an effective therapeutic strategy for the prevention of AD and intestinal disease. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12951-022-01642-z. BioMed Central 2022-10-07 /pmc/articles/PMC9547428/ /pubmed/36207740 http://dx.doi.org/10.1186/s12951-022-01642-z Text en © The Author(s) 2022 https://creativecommons.org/licenses/by/4.0/Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Liu, Ni
Yang, Changwen
Liang, Xiaohan
Cao, Kai
Xie, Jun
Luo, Qingming
Luo, Haiming
Mesoporous silica nanoparticle-encapsulated Bifidobacterium attenuates brain Aβ burden and improves olfactory dysfunction of APP/PS1 mice by nasal delivery
title Mesoporous silica nanoparticle-encapsulated Bifidobacterium attenuates brain Aβ burden and improves olfactory dysfunction of APP/PS1 mice by nasal delivery
title_full Mesoporous silica nanoparticle-encapsulated Bifidobacterium attenuates brain Aβ burden and improves olfactory dysfunction of APP/PS1 mice by nasal delivery
title_fullStr Mesoporous silica nanoparticle-encapsulated Bifidobacterium attenuates brain Aβ burden and improves olfactory dysfunction of APP/PS1 mice by nasal delivery
title_full_unstemmed Mesoporous silica nanoparticle-encapsulated Bifidobacterium attenuates brain Aβ burden and improves olfactory dysfunction of APP/PS1 mice by nasal delivery
title_short Mesoporous silica nanoparticle-encapsulated Bifidobacterium attenuates brain Aβ burden and improves olfactory dysfunction of APP/PS1 mice by nasal delivery
title_sort mesoporous silica nanoparticle-encapsulated bifidobacterium attenuates brain aβ burden and improves olfactory dysfunction of app/ps1 mice by nasal delivery
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9547428/
https://www.ncbi.nlm.nih.gov/pubmed/36207740
http://dx.doi.org/10.1186/s12951-022-01642-z
work_keys_str_mv AT liuni mesoporoussilicananoparticleencapsulatedbifidobacteriumattenuatesbrainabburdenandimprovesolfactorydysfunctionofappps1micebynasaldelivery
AT yangchangwen mesoporoussilicananoparticleencapsulatedbifidobacteriumattenuatesbrainabburdenandimprovesolfactorydysfunctionofappps1micebynasaldelivery
AT liangxiaohan mesoporoussilicananoparticleencapsulatedbifidobacteriumattenuatesbrainabburdenandimprovesolfactorydysfunctionofappps1micebynasaldelivery
AT caokai mesoporoussilicananoparticleencapsulatedbifidobacteriumattenuatesbrainabburdenandimprovesolfactorydysfunctionofappps1micebynasaldelivery
AT xiejun mesoporoussilicananoparticleencapsulatedbifidobacteriumattenuatesbrainabburdenandimprovesolfactorydysfunctionofappps1micebynasaldelivery
AT luoqingming mesoporoussilicananoparticleencapsulatedbifidobacteriumattenuatesbrainabburdenandimprovesolfactorydysfunctionofappps1micebynasaldelivery
AT luohaiming mesoporoussilicananoparticleencapsulatedbifidobacteriumattenuatesbrainabburdenandimprovesolfactorydysfunctionofappps1micebynasaldelivery