Cargando…

Longitudinal development of the gut microbiome and metabolome in preterm neonates with late onset sepsis and healthy controls

BACKGROUND: Late onset sepsis (LOS) in preterm infants is associated with considerable morbidity and mortality. While studies have implicated gut bacteria in the aetiology of the disease, functional analysis and mechanistic insights are generally lacking. We performed temporal bacterial (n = 613) an...

Descripción completa

Detalles Bibliográficos
Autores principales: Stewart, Christopher J., Embleton, Nicholas D., Marrs, Emma C. L., Smith, Daniel P., Fofanova, Tatiana, Nelson, Andrew, Skeath, Tom, Perry, John D., Petrosino, Joseph F., Berrington, Janet E., Cummings, Stephen P.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5508794/
https://www.ncbi.nlm.nih.gov/pubmed/28701177
http://dx.doi.org/10.1186/s40168-017-0295-1
_version_ 1783249935415640064
author Stewart, Christopher J.
Embleton, Nicholas D.
Marrs, Emma C. L.
Smith, Daniel P.
Fofanova, Tatiana
Nelson, Andrew
Skeath, Tom
Perry, John D.
Petrosino, Joseph F.
Berrington, Janet E.
Cummings, Stephen P.
author_facet Stewart, Christopher J.
Embleton, Nicholas D.
Marrs, Emma C. L.
Smith, Daniel P.
Fofanova, Tatiana
Nelson, Andrew
Skeath, Tom
Perry, John D.
Petrosino, Joseph F.
Berrington, Janet E.
Cummings, Stephen P.
author_sort Stewart, Christopher J.
collection PubMed
description BACKGROUND: Late onset sepsis (LOS) in preterm infants is associated with considerable morbidity and mortality. While studies have implicated gut bacteria in the aetiology of the disease, functional analysis and mechanistic insights are generally lacking. We performed temporal bacterial (n = 613) and metabolomic (n = 63) profiling on extensively sampled stool from 7 infants with LOS and 28 matched healthy (no LOS or NEC) controls. RESULTS: The bacteria isolated in diagnostic blood culture usually corresponded to the dominant bacterial genera in the gut microbiome. Longitudinal changes were monitored based on preterm gut community types (PGCTs), where control infants had an increased number of PGCTs compared to LOS infants (P = 0.011). PGCT 6, characterised by Bifidobacteria dominance, was only present in control infants. Metabolite profiles differed between LOS and control infants at diagnosis and 7 days later, but not 7 days prior to diagnosis. Bifidobacteria was positively correlated with control metabolites, including raffinose, sucrose, and acetic acid. CONCLUSIONS: Using multi-omic analysis, we show that the gut microbiome is involved in the pathogenesis of LOS. While the causative agent of LOS varies, it is usually abundant in the gut. Bifidobacteria dominance was associated with control infants, and the presence of this organism may directly protect, or act as a marker for protection, against gut epithelial translocation. While the metabolomic data is preliminary, the findings support that gut development and protection in preterm infants is associated with increased in prebiotic oligosaccharides (e.g. raffinose) and the growth of beneficial bacteria (e.g. Bifidobacterium). ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/s40168-017-0295-1) contains supplementary material, which is available to authorized users.
format Online
Article
Text
id pubmed-5508794
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-55087942017-07-17 Longitudinal development of the gut microbiome and metabolome in preterm neonates with late onset sepsis and healthy controls Stewart, Christopher J. Embleton, Nicholas D. Marrs, Emma C. L. Smith, Daniel P. Fofanova, Tatiana Nelson, Andrew Skeath, Tom Perry, John D. Petrosino, Joseph F. Berrington, Janet E. Cummings, Stephen P. Microbiome Research BACKGROUND: Late onset sepsis (LOS) in preterm infants is associated with considerable morbidity and mortality. While studies have implicated gut bacteria in the aetiology of the disease, functional analysis and mechanistic insights are generally lacking. We performed temporal bacterial (n = 613) and metabolomic (n = 63) profiling on extensively sampled stool from 7 infants with LOS and 28 matched healthy (no LOS or NEC) controls. RESULTS: The bacteria isolated in diagnostic blood culture usually corresponded to the dominant bacterial genera in the gut microbiome. Longitudinal changes were monitored based on preterm gut community types (PGCTs), where control infants had an increased number of PGCTs compared to LOS infants (P = 0.011). PGCT 6, characterised by Bifidobacteria dominance, was only present in control infants. Metabolite profiles differed between LOS and control infants at diagnosis and 7 days later, but not 7 days prior to diagnosis. Bifidobacteria was positively correlated with control metabolites, including raffinose, sucrose, and acetic acid. CONCLUSIONS: Using multi-omic analysis, we show that the gut microbiome is involved in the pathogenesis of LOS. While the causative agent of LOS varies, it is usually abundant in the gut. Bifidobacteria dominance was associated with control infants, and the presence of this organism may directly protect, or act as a marker for protection, against gut epithelial translocation. While the metabolomic data is preliminary, the findings support that gut development and protection in preterm infants is associated with increased in prebiotic oligosaccharides (e.g. raffinose) and the growth of beneficial bacteria (e.g. Bifidobacterium). ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/s40168-017-0295-1) contains supplementary material, which is available to authorized users. BioMed Central 2017-07-12 /pmc/articles/PMC5508794/ /pubmed/28701177 http://dx.doi.org/10.1186/s40168-017-0295-1 Text en © The Author(s). 2017 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
Stewart, Christopher J.
Embleton, Nicholas D.
Marrs, Emma C. L.
Smith, Daniel P.
Fofanova, Tatiana
Nelson, Andrew
Skeath, Tom
Perry, John D.
Petrosino, Joseph F.
Berrington, Janet E.
Cummings, Stephen P.
Longitudinal development of the gut microbiome and metabolome in preterm neonates with late onset sepsis and healthy controls
title Longitudinal development of the gut microbiome and metabolome in preterm neonates with late onset sepsis and healthy controls
title_full Longitudinal development of the gut microbiome and metabolome in preterm neonates with late onset sepsis and healthy controls
title_fullStr Longitudinal development of the gut microbiome and metabolome in preterm neonates with late onset sepsis and healthy controls
title_full_unstemmed Longitudinal development of the gut microbiome and metabolome in preterm neonates with late onset sepsis and healthy controls
title_short Longitudinal development of the gut microbiome and metabolome in preterm neonates with late onset sepsis and healthy controls
title_sort longitudinal development of the gut microbiome and metabolome in preterm neonates with late onset sepsis and healthy controls
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5508794/
https://www.ncbi.nlm.nih.gov/pubmed/28701177
http://dx.doi.org/10.1186/s40168-017-0295-1
work_keys_str_mv AT stewartchristopherj longitudinaldevelopmentofthegutmicrobiomeandmetabolomeinpretermneonateswithlateonsetsepsisandhealthycontrols
AT embletonnicholasd longitudinaldevelopmentofthegutmicrobiomeandmetabolomeinpretermneonateswithlateonsetsepsisandhealthycontrols
AT marrsemmacl longitudinaldevelopmentofthegutmicrobiomeandmetabolomeinpretermneonateswithlateonsetsepsisandhealthycontrols
AT smithdanielp longitudinaldevelopmentofthegutmicrobiomeandmetabolomeinpretermneonateswithlateonsetsepsisandhealthycontrols
AT fofanovatatiana longitudinaldevelopmentofthegutmicrobiomeandmetabolomeinpretermneonateswithlateonsetsepsisandhealthycontrols
AT nelsonandrew longitudinaldevelopmentofthegutmicrobiomeandmetabolomeinpretermneonateswithlateonsetsepsisandhealthycontrols
AT skeathtom longitudinaldevelopmentofthegutmicrobiomeandmetabolomeinpretermneonateswithlateonsetsepsisandhealthycontrols
AT perryjohnd longitudinaldevelopmentofthegutmicrobiomeandmetabolomeinpretermneonateswithlateonsetsepsisandhealthycontrols
AT petrosinojosephf longitudinaldevelopmentofthegutmicrobiomeandmetabolomeinpretermneonateswithlateonsetsepsisandhealthycontrols
AT berringtonjanete longitudinaldevelopmentofthegutmicrobiomeandmetabolomeinpretermneonateswithlateonsetsepsisandhealthycontrols
AT cummingsstephenp longitudinaldevelopmentofthegutmicrobiomeandmetabolomeinpretermneonateswithlateonsetsepsisandhealthycontrols