Cargando…

Fetal Exposure to Maternal Inflammation Does Not Affect Postnatal Development of Genetically-Driven Ileitis and Colitis

Background: Chronic inflammatory disorders have been increasing in incidence over the past decades following geographical patterns of industrialization. Fetal exposure to maternal inflammation may alter organ functions and the offspring's disease risk. We studied the development of genetically-...

Descripción completa

Detalles Bibliográficos
Autores principales: Hemmerling, Jana, Heller, Katharina, Hörmannsperger, Gabriele, Bazanella, Monika, Clavel, Thomas, Kollias, George, Haller, Dirk
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2014
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4029898/
https://www.ncbi.nlm.nih.gov/pubmed/24849654
http://dx.doi.org/10.1371/journal.pone.0098237
_version_ 1782317295265120256
author Hemmerling, Jana
Heller, Katharina
Hörmannsperger, Gabriele
Bazanella, Monika
Clavel, Thomas
Kollias, George
Haller, Dirk
author_facet Hemmerling, Jana
Heller, Katharina
Hörmannsperger, Gabriele
Bazanella, Monika
Clavel, Thomas
Kollias, George
Haller, Dirk
author_sort Hemmerling, Jana
collection PubMed
description Background: Chronic inflammatory disorders have been increasing in incidence over the past decades following geographical patterns of industrialization. Fetal exposure to maternal inflammation may alter organ functions and the offspring's disease risk. We studied the development of genetically-driven ileitis and colitis in response to maternal inflammation using mouse models. Methods: Disease susceptible (Tnf (ΔARE/+) and IL10(−/−)) and disease-free (Tnf (+/+) and IL10(−/+)) offspring were raised in inflamed and non-inflamed dams. Ileal, caecal and colonic pathology was evaluated in the offspring at 8 or 12 weeks of age. Ly6G-positive cells in inflamed sections from the distal ileum and distal colon were analysed by immunofluorescence microscopy. Gene expression of pro-inflammatory cytokines was measured in whole tissue specimens by quantitative PCR. Microarray analyses were performed on laser microdissected intestinal epithelium. Caecal bacterial communities were assessed by Illumina sequencing of 16S rRNA amplicons. Results: Disease severity, the number of infiltrated neutrophils as well as Tnf and Il12p40 mRNA expression were independent of maternal inflammation in the offspring of mouse models for ileitis (Tnf (ΔARE/+)) and colitis (IL10(−/−)). Although TNF-driven maternal inflammation regulated 2,174 (wild type) and 3,345 (Tnf (ΔARE/+)) genes in the fetal epithelium, prenatal gene expression patterns were completely overwritten after birth. In addition, co-housing experiments revealed no change in phylogenetic diversity of the offspring's caecal microbiota in response to maternal inflammation. This is independent of the offspring's genotype before and after the onset of tissue pathology. Conclusions: Disease risk and activity in mouse models of chronic ileitis and colitis was independent of the fetal exposure to maternal inflammation. Likewise, maternal inflammation did not alter the diversity and composition of offspring's caecal microbiota, clearly demonstrating that changes of the gene expression program in the fetal gut epithelium were not relevant for the development of chronic inflammatory disorders in the gut.
format Online
Article
Text
id pubmed-4029898
institution National Center for Biotechnology Information
language English
publishDate 2014
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-40298982014-05-28 Fetal Exposure to Maternal Inflammation Does Not Affect Postnatal Development of Genetically-Driven Ileitis and Colitis Hemmerling, Jana Heller, Katharina Hörmannsperger, Gabriele Bazanella, Monika Clavel, Thomas Kollias, George Haller, Dirk PLoS One Research Article Background: Chronic inflammatory disorders have been increasing in incidence over the past decades following geographical patterns of industrialization. Fetal exposure to maternal inflammation may alter organ functions and the offspring's disease risk. We studied the development of genetically-driven ileitis and colitis in response to maternal inflammation using mouse models. Methods: Disease susceptible (Tnf (ΔARE/+) and IL10(−/−)) and disease-free (Tnf (+/+) and IL10(−/+)) offspring were raised in inflamed and non-inflamed dams. Ileal, caecal and colonic pathology was evaluated in the offspring at 8 or 12 weeks of age. Ly6G-positive cells in inflamed sections from the distal ileum and distal colon were analysed by immunofluorescence microscopy. Gene expression of pro-inflammatory cytokines was measured in whole tissue specimens by quantitative PCR. Microarray analyses were performed on laser microdissected intestinal epithelium. Caecal bacterial communities were assessed by Illumina sequencing of 16S rRNA amplicons. Results: Disease severity, the number of infiltrated neutrophils as well as Tnf and Il12p40 mRNA expression were independent of maternal inflammation in the offspring of mouse models for ileitis (Tnf (ΔARE/+)) and colitis (IL10(−/−)). Although TNF-driven maternal inflammation regulated 2,174 (wild type) and 3,345 (Tnf (ΔARE/+)) genes in the fetal epithelium, prenatal gene expression patterns were completely overwritten after birth. In addition, co-housing experiments revealed no change in phylogenetic diversity of the offspring's caecal microbiota in response to maternal inflammation. This is independent of the offspring's genotype before and after the onset of tissue pathology. Conclusions: Disease risk and activity in mouse models of chronic ileitis and colitis was independent of the fetal exposure to maternal inflammation. Likewise, maternal inflammation did not alter the diversity and composition of offspring's caecal microbiota, clearly demonstrating that changes of the gene expression program in the fetal gut epithelium were not relevant for the development of chronic inflammatory disorders in the gut. Public Library of Science 2014-05-21 /pmc/articles/PMC4029898/ /pubmed/24849654 http://dx.doi.org/10.1371/journal.pone.0098237 Text en © 2014 Hemmerling et al http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are properly credited.
spellingShingle Research Article
Hemmerling, Jana
Heller, Katharina
Hörmannsperger, Gabriele
Bazanella, Monika
Clavel, Thomas
Kollias, George
Haller, Dirk
Fetal Exposure to Maternal Inflammation Does Not Affect Postnatal Development of Genetically-Driven Ileitis and Colitis
title Fetal Exposure to Maternal Inflammation Does Not Affect Postnatal Development of Genetically-Driven Ileitis and Colitis
title_full Fetal Exposure to Maternal Inflammation Does Not Affect Postnatal Development of Genetically-Driven Ileitis and Colitis
title_fullStr Fetal Exposure to Maternal Inflammation Does Not Affect Postnatal Development of Genetically-Driven Ileitis and Colitis
title_full_unstemmed Fetal Exposure to Maternal Inflammation Does Not Affect Postnatal Development of Genetically-Driven Ileitis and Colitis
title_short Fetal Exposure to Maternal Inflammation Does Not Affect Postnatal Development of Genetically-Driven Ileitis and Colitis
title_sort fetal exposure to maternal inflammation does not affect postnatal development of genetically-driven ileitis and colitis
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4029898/
https://www.ncbi.nlm.nih.gov/pubmed/24849654
http://dx.doi.org/10.1371/journal.pone.0098237
work_keys_str_mv AT hemmerlingjana fetalexposuretomaternalinflammationdoesnotaffectpostnataldevelopmentofgeneticallydrivenileitisandcolitis
AT hellerkatharina fetalexposuretomaternalinflammationdoesnotaffectpostnataldevelopmentofgeneticallydrivenileitisandcolitis
AT hormannspergergabriele fetalexposuretomaternalinflammationdoesnotaffectpostnataldevelopmentofgeneticallydrivenileitisandcolitis
AT bazanellamonika fetalexposuretomaternalinflammationdoesnotaffectpostnataldevelopmentofgeneticallydrivenileitisandcolitis
AT clavelthomas fetalexposuretomaternalinflammationdoesnotaffectpostnataldevelopmentofgeneticallydrivenileitisandcolitis
AT kolliasgeorge fetalexposuretomaternalinflammationdoesnotaffectpostnataldevelopmentofgeneticallydrivenileitisandcolitis
AT hallerdirk fetalexposuretomaternalinflammationdoesnotaffectpostnataldevelopmentofgeneticallydrivenileitisandcolitis