Infant gut microbiota modulation by human milk disaccharides in humanized microbiome mice

Human milk glycans present a unique diversity of structures that suggest different mechanisms by which they may affect the infant microbiome development. A humanized mouse model generated by infant fecal transplantation was utilized here to evaluate the impact of fucosyl-α1,3-GlcNAc (3FN), fucosyl-α1,6-GlcNAc, lacto-N-biose (LNB) and galacto-N-biose on the fecal microbiota and host–microbiota interactions. 16S rRNA amplicon sequencing showed that certain bacterial genera significantly increased (Ruminococcus and Oscillospira) or decreased (Eubacterium and Clostridium) in all disaccharide-supplemented groups. Interestingly, cluster analysis differentiates the consumption of fucosyl-oligosaccharides from galactosyl-oligosaccharides, highlighting the disappearance of Akkermansia genus in both fucosyl-oligosaccharides. An increment of the relative abundance of Coprococcus genus was only observed with 3FN. As well, LNB significantly increased the relative abundance of Bifidobacterium, whereas the absolute levels of this genus, as measured by quantitative real-time PCR, did not significantly increase. OTUs corresponding to the species Bifidobacterium longum, Bifidobacterium adolescentis and Ruminococcus gnavus were not present in the control after the 3-week intervention, but were shared among the donor and specific disaccharide groups, indicating that their survival is dependent on disaccharide supplementation. The 3FN-feeding group showed increased levels of butyrate and acetate in the colon, and decreased levels of serum HDL-cholesterol. 3FN also down-regulated the pro-inflammatory cytokine TNF-α and up-regulated the anti-inflammatory cytokines IL-10 and IL-13, and the Toll-like receptor 2 in the large intestine tissue. The present study revealed that the four disaccharides show efficacy in producing beneficial compositional shifts of the gut microbiota and in addition, the 3FN demonstrated physiological and immunomodulatory roles.

Saved in:
Bibliographic Details
Main Authors: Rubio-Del-Campo, Antonio, Gozalbo-Rovira, Roberto, Moya Gonzálvez, Eva M., Alberola, Juan, Rodríguez-Díaz, Jesús, Yebra, María Jesús
Other Authors: Ministerio de Economía y Competitividad (España)
Format: artículo biblioteca
Language:English
Published: Taylor & Francis 2021-05-03
Subjects:fucosyl-α-13-N-acetylglucosamine, fucosyl-α-16-N-acetylglucosamine, lacto-N-biosegalacto-N–biose, human milk oligosaccharides, infant fecal microbiota, humanized mouse model, short-chain fatty acidscytokines,
Online Access:http://hdl.handle.net/10261/244406
http://dx.doi.org/10.13039/501100000780
http://dx.doi.org/10.13039/501100003359
http://dx.doi.org/10.13039/501100003329
Tags: Add Tag
No Tags, Be the first to tag this record!
id dig-iata-es-10261-244406
record_format koha
spelling dig-iata-es-10261-2444062022-06-24T09:40:11Z Infant gut microbiota modulation by human milk disaccharides in humanized microbiome mice Rubio-Del-Campo, Antonio Gozalbo-Rovira, Roberto Moya Gonzálvez, Eva M. Alberola, Juan Rodríguez-Díaz, Jesús Yebra, María Jesús Ministerio de Economía y Competitividad (España) Generalitat Valenciana European Commission fucosyl-α-13-N-acetylglucosamine fucosyl-α-16-N-acetylglucosamine lacto-N-biosegalacto-N–biose human milk oligosaccharides infant fecal microbiota humanized mouse model short-chain fatty acidscytokines Human milk glycans present a unique diversity of structures that suggest different mechanisms by which they may affect the infant microbiome development. A humanized mouse model generated by infant fecal transplantation was utilized here to evaluate the impact of fucosyl-α1,3-GlcNAc (3FN), fucosyl-α1,6-GlcNAc, lacto-N-biose (LNB) and galacto-N-biose on the fecal microbiota and host–microbiota interactions. 16S rRNA amplicon sequencing showed that certain bacterial genera significantly increased (Ruminococcus and Oscillospira) or decreased (Eubacterium and Clostridium) in all disaccharide-supplemented groups. Interestingly, cluster analysis differentiates the consumption of fucosyl-oligosaccharides from galactosyl-oligosaccharides, highlighting the disappearance of Akkermansia genus in both fucosyl-oligosaccharides. An increment of the relative abundance of Coprococcus genus was only observed with 3FN. As well, LNB significantly increased the relative abundance of Bifidobacterium, whereas the absolute levels of this genus, as measured by quantitative real-time PCR, did not significantly increase. OTUs corresponding to the species Bifidobacterium longum, Bifidobacterium adolescentis and Ruminococcus gnavus were not present in the control after the 3-week intervention, but were shared among the donor and specific disaccharide groups, indicating that their survival is dependent on disaccharide supplementation. The 3FN-feeding group showed increased levels of butyrate and acetate in the colon, and decreased levels of serum HDL-cholesterol. 3FN also down-regulated the pro-inflammatory cytokine TNF-α and up-regulated the anti-inflammatory cytokines IL-10 and IL-13, and the Toll-like receptor 2 in the large intestine tissue. The present study revealed that the four disaccharides show efficacy in producing beneficial compositional shifts of the gut microbiota and in addition, the 3FN demonstrated physiological and immunomodulatory roles. This work was supported by the Spanish Ministry for Economy and Competitiveness (MINECO)/FEDER under Grant AGL2017-84165-C2 (1-R and 2-R). JRD was supported by a Ramon y Cajal Contract RYC-2013-12442 by the Spanish Ministry for Economy and Competitiveness and by Valencian Government/FEDER grant IDIFEDER/2018/056. EMMG was supported by a pre-doctoral Contract PRE2018-085768 by the Spanish Ministry of Science, Innovation and Universities. Peer reviewed 2021-06-23T05:59:48Z 2021-06-23T05:59:48Z 2021-05-03 artículo http://purl.org/coar/resource_type/c_6501 Gut microbes 13 (1): 1914377 (2021) 1949-0976 http://hdl.handle.net/10261/244406 10.1080/19490976.2021.1914377 http://dx.doi.org/10.13039/501100000780 http://dx.doi.org/10.13039/501100003359 http://dx.doi.org/10.13039/501100003329 33938391 en #PLACEHOLDER_PARENT_METADATA_VALUE# info:eu-repo/grantAgreement/MINECO/Plan Estatal de Investigación Científica y Técnica y de Innovación 2013-2016/AGL2017-84165-C2 Publisher's version https://doi.org/10.1080/19490976.2021.1914377 Sí open Taylor & Francis
institution IATA ES
collection DSpace
country España
countrycode ES
component Bibliográfico
access En linea
databasecode dig-iata-es
tag biblioteca
region Europa del Sur
libraryname Biblioteca del IATA España
language English
topic fucosyl-α-13-N-acetylglucosamine
fucosyl-α-16-N-acetylglucosamine
lacto-N-biosegalacto-N–biose
human milk oligosaccharides
infant fecal microbiota
humanized mouse model
short-chain fatty acidscytokines
fucosyl-α-13-N-acetylglucosamine
fucosyl-α-16-N-acetylglucosamine
lacto-N-biosegalacto-N–biose
human milk oligosaccharides
infant fecal microbiota
humanized mouse model
short-chain fatty acidscytokines
spellingShingle fucosyl-α-13-N-acetylglucosamine
fucosyl-α-16-N-acetylglucosamine
lacto-N-biosegalacto-N–biose
human milk oligosaccharides
infant fecal microbiota
humanized mouse model
short-chain fatty acidscytokines
fucosyl-α-13-N-acetylglucosamine
fucosyl-α-16-N-acetylglucosamine
lacto-N-biosegalacto-N–biose
human milk oligosaccharides
infant fecal microbiota
humanized mouse model
short-chain fatty acidscytokines
Rubio-Del-Campo, Antonio
Gozalbo-Rovira, Roberto
Moya Gonzálvez, Eva M.
Alberola, Juan
Rodríguez-Díaz, Jesús
Yebra, María Jesús
Infant gut microbiota modulation by human milk disaccharides in humanized microbiome mice
description Human milk glycans present a unique diversity of structures that suggest different mechanisms by which they may affect the infant microbiome development. A humanized mouse model generated by infant fecal transplantation was utilized here to evaluate the impact of fucosyl-α1,3-GlcNAc (3FN), fucosyl-α1,6-GlcNAc, lacto-N-biose (LNB) and galacto-N-biose on the fecal microbiota and host–microbiota interactions. 16S rRNA amplicon sequencing showed that certain bacterial genera significantly increased (Ruminococcus and Oscillospira) or decreased (Eubacterium and Clostridium) in all disaccharide-supplemented groups. Interestingly, cluster analysis differentiates the consumption of fucosyl-oligosaccharides from galactosyl-oligosaccharides, highlighting the disappearance of Akkermansia genus in both fucosyl-oligosaccharides. An increment of the relative abundance of Coprococcus genus was only observed with 3FN. As well, LNB significantly increased the relative abundance of Bifidobacterium, whereas the absolute levels of this genus, as measured by quantitative real-time PCR, did not significantly increase. OTUs corresponding to the species Bifidobacterium longum, Bifidobacterium adolescentis and Ruminococcus gnavus were not present in the control after the 3-week intervention, but were shared among the donor and specific disaccharide groups, indicating that their survival is dependent on disaccharide supplementation. The 3FN-feeding group showed increased levels of butyrate and acetate in the colon, and decreased levels of serum HDL-cholesterol. 3FN also down-regulated the pro-inflammatory cytokine TNF-α and up-regulated the anti-inflammatory cytokines IL-10 and IL-13, and the Toll-like receptor 2 in the large intestine tissue. The present study revealed that the four disaccharides show efficacy in producing beneficial compositional shifts of the gut microbiota and in addition, the 3FN demonstrated physiological and immunomodulatory roles.
author2 Ministerio de Economía y Competitividad (España)
author_facet Ministerio de Economía y Competitividad (España)
Rubio-Del-Campo, Antonio
Gozalbo-Rovira, Roberto
Moya Gonzálvez, Eva M.
Alberola, Juan
Rodríguez-Díaz, Jesús
Yebra, María Jesús
format artículo
topic_facet fucosyl-α-13-N-acetylglucosamine
fucosyl-α-16-N-acetylglucosamine
lacto-N-biosegalacto-N–biose
human milk oligosaccharides
infant fecal microbiota
humanized mouse model
short-chain fatty acidscytokines
author Rubio-Del-Campo, Antonio
Gozalbo-Rovira, Roberto
Moya Gonzálvez, Eva M.
Alberola, Juan
Rodríguez-Díaz, Jesús
Yebra, María Jesús
author_sort Rubio-Del-Campo, Antonio
title Infant gut microbiota modulation by human milk disaccharides in humanized microbiome mice
title_short Infant gut microbiota modulation by human milk disaccharides in humanized microbiome mice
title_full Infant gut microbiota modulation by human milk disaccharides in humanized microbiome mice
title_fullStr Infant gut microbiota modulation by human milk disaccharides in humanized microbiome mice
title_full_unstemmed Infant gut microbiota modulation by human milk disaccharides in humanized microbiome mice
title_sort infant gut microbiota modulation by human milk disaccharides in humanized microbiome mice
publisher Taylor & Francis
publishDate 2021-05-03
url http://hdl.handle.net/10261/244406
http://dx.doi.org/10.13039/501100000780
http://dx.doi.org/10.13039/501100003359
http://dx.doi.org/10.13039/501100003329
work_keys_str_mv AT rubiodelcampoantonio infantgutmicrobiotamodulationbyhumanmilkdisaccharidesinhumanizedmicrobiomemice
AT gozalboroviraroberto infantgutmicrobiotamodulationbyhumanmilkdisaccharidesinhumanizedmicrobiomemice
AT moyagonzalvezevam infantgutmicrobiotamodulationbyhumanmilkdisaccharidesinhumanizedmicrobiomemice
AT alberolajuan infantgutmicrobiotamodulationbyhumanmilkdisaccharidesinhumanizedmicrobiomemice
AT rodriguezdiazjesus infantgutmicrobiotamodulationbyhumanmilkdisaccharidesinhumanizedmicrobiomemice
AT yebramariajesus infantgutmicrobiotamodulationbyhumanmilkdisaccharidesinhumanizedmicrobiomemice
_version_ 1777670183216742400