Cargando…

Disruption of maternal gut microbiota during gestation alters offspring microbiota and immunity

BACKGROUND: Early life microbiota is an important determinant of immune and metabolic development and may have lasting consequences. The maternal gut microbiota during pregnancy or breastfeeding is important for defining infant gut microbiota. We hypothesized that maternal gut microbiota during preg...

Descripción completa

Detalles Bibliográficos
Autores principales: Nyangahu, Donald D., Lennard, Katie S., Brown, Bryan P., Darby, Matthew G., Wendoh, Jerome M., Havyarimana, Enock, Smith, Peter, Butcher, James, Stintzi, Alain, Mulder, Nicola, Horsnell, William, Jaspan, Heather B.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6035804/
https://www.ncbi.nlm.nih.gov/pubmed/29981583
http://dx.doi.org/10.1186/s40168-018-0511-7
_version_ 1783338079590809600
author Nyangahu, Donald D.
Lennard, Katie S.
Brown, Bryan P.
Darby, Matthew G.
Wendoh, Jerome M.
Havyarimana, Enock
Smith, Peter
Butcher, James
Stintzi, Alain
Mulder, Nicola
Horsnell, William
Jaspan, Heather B.
author_facet Nyangahu, Donald D.
Lennard, Katie S.
Brown, Bryan P.
Darby, Matthew G.
Wendoh, Jerome M.
Havyarimana, Enock
Smith, Peter
Butcher, James
Stintzi, Alain
Mulder, Nicola
Horsnell, William
Jaspan, Heather B.
author_sort Nyangahu, Donald D.
collection PubMed
description BACKGROUND: Early life microbiota is an important determinant of immune and metabolic development and may have lasting consequences. The maternal gut microbiota during pregnancy or breastfeeding is important for defining infant gut microbiota. We hypothesized that maternal gut microbiota during pregnancy and breastfeeding is a critical determinant of infant immunity. To test this, pregnant BALB/c dams were fed vancomycin for 5 days prior to delivery (gestation; Mg), 14 days postpartum during nursing (Mn), or during gestation and nursing (Mgn), or no vancomycin (Mc). We analyzed adaptive immunity and gut microbiota in dams and pups at various times after delivery. RESULTS: In addition to direct alterations to maternal gut microbial composition, pup gut microbiota displayed lower α-diversity and distinct community clusters according to timing of maternal vancomycin. Vancomycin was undetectable in maternal and offspring sera, therefore the observed changes in the microbiota of stomach contents (as a proxy for breastmilk) and pup gut signify an indirect mechanism through which maternal intestinal microbiota influences extra-intestinal and neonatal commensal colonization. These effects on microbiota influenced both maternal and offspring immunity. Maternal immunity was altered, as demonstrated by significantly higher levels of both total IgG and IgM in Mgn and Mn breastmilk when compared to Mc. In pups, lymphocyte numbers in the spleens of Pg and Pn were significantly increased compared to Pc. This increase in cellularity was in part attributable to elevated numbers of both CD4+ T cells and B cells, most notable Follicular B cells. CONCLUSION: Our results indicate that perturbations to maternal gut microbiota dictate neonatal adaptive immunity. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1186/s40168-018-0511-7) contains supplementary material, which is available to authorized users.
format Online
Article
Text
id pubmed-6035804
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-60358042018-07-12 Disruption of maternal gut microbiota during gestation alters offspring microbiota and immunity Nyangahu, Donald D. Lennard, Katie S. Brown, Bryan P. Darby, Matthew G. Wendoh, Jerome M. Havyarimana, Enock Smith, Peter Butcher, James Stintzi, Alain Mulder, Nicola Horsnell, William Jaspan, Heather B. Microbiome Research BACKGROUND: Early life microbiota is an important determinant of immune and metabolic development and may have lasting consequences. The maternal gut microbiota during pregnancy or breastfeeding is important for defining infant gut microbiota. We hypothesized that maternal gut microbiota during pregnancy and breastfeeding is a critical determinant of infant immunity. To test this, pregnant BALB/c dams were fed vancomycin for 5 days prior to delivery (gestation; Mg), 14 days postpartum during nursing (Mn), or during gestation and nursing (Mgn), or no vancomycin (Mc). We analyzed adaptive immunity and gut microbiota in dams and pups at various times after delivery. RESULTS: In addition to direct alterations to maternal gut microbial composition, pup gut microbiota displayed lower α-diversity and distinct community clusters according to timing of maternal vancomycin. Vancomycin was undetectable in maternal and offspring sera, therefore the observed changes in the microbiota of stomach contents (as a proxy for breastmilk) and pup gut signify an indirect mechanism through which maternal intestinal microbiota influences extra-intestinal and neonatal commensal colonization. These effects on microbiota influenced both maternal and offspring immunity. Maternal immunity was altered, as demonstrated by significantly higher levels of both total IgG and IgM in Mgn and Mn breastmilk when compared to Mc. In pups, lymphocyte numbers in the spleens of Pg and Pn were significantly increased compared to Pc. This increase in cellularity was in part attributable to elevated numbers of both CD4+ T cells and B cells, most notable Follicular B cells. CONCLUSION: Our results indicate that perturbations to maternal gut microbiota dictate neonatal adaptive immunity. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1186/s40168-018-0511-7) contains supplementary material, which is available to authorized users. BioMed Central 2018-07-07 /pmc/articles/PMC6035804/ /pubmed/29981583 http://dx.doi.org/10.1186/s40168-018-0511-7 Text en © The Author(s). 2018 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research
Nyangahu, Donald D.
Lennard, Katie S.
Brown, Bryan P.
Darby, Matthew G.
Wendoh, Jerome M.
Havyarimana, Enock
Smith, Peter
Butcher, James
Stintzi, Alain
Mulder, Nicola
Horsnell, William
Jaspan, Heather B.
Disruption of maternal gut microbiota during gestation alters offspring microbiota and immunity
title Disruption of maternal gut microbiota during gestation alters offspring microbiota and immunity
title_full Disruption of maternal gut microbiota during gestation alters offspring microbiota and immunity
title_fullStr Disruption of maternal gut microbiota during gestation alters offspring microbiota and immunity
title_full_unstemmed Disruption of maternal gut microbiota during gestation alters offspring microbiota and immunity
title_short Disruption of maternal gut microbiota during gestation alters offspring microbiota and immunity
title_sort disruption of maternal gut microbiota during gestation alters offspring microbiota and immunity
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6035804/
https://www.ncbi.nlm.nih.gov/pubmed/29981583
http://dx.doi.org/10.1186/s40168-018-0511-7
work_keys_str_mv AT nyangahudonaldd disruptionofmaternalgutmicrobiotaduringgestationaltersoffspringmicrobiotaandimmunity
AT lennardkaties disruptionofmaternalgutmicrobiotaduringgestationaltersoffspringmicrobiotaandimmunity
AT brownbryanp disruptionofmaternalgutmicrobiotaduringgestationaltersoffspringmicrobiotaandimmunity
AT darbymatthewg disruptionofmaternalgutmicrobiotaduringgestationaltersoffspringmicrobiotaandimmunity
AT wendohjeromem disruptionofmaternalgutmicrobiotaduringgestationaltersoffspringmicrobiotaandimmunity
AT havyarimanaenock disruptionofmaternalgutmicrobiotaduringgestationaltersoffspringmicrobiotaandimmunity
AT smithpeter disruptionofmaternalgutmicrobiotaduringgestationaltersoffspringmicrobiotaandimmunity
AT butcherjames disruptionofmaternalgutmicrobiotaduringgestationaltersoffspringmicrobiotaandimmunity
AT stintzialain disruptionofmaternalgutmicrobiotaduringgestationaltersoffspringmicrobiotaandimmunity
AT muldernicola disruptionofmaternalgutmicrobiotaduringgestationaltersoffspringmicrobiotaandimmunity
AT horsnellwilliam disruptionofmaternalgutmicrobiotaduringgestationaltersoffspringmicrobiotaandimmunity
AT jaspanheatherb disruptionofmaternalgutmicrobiotaduringgestationaltersoffspringmicrobiotaandimmunity