Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Gut Microbes ; 3(5): 426-33, 2012.
Article in English | MEDLINE | ID: mdl-22713270

ABSTRACT

Epidemiologic data suggest that early nutritional exposures may inflict persistent changes in the developing mammalian "super-organism" (i.e., the host and its residing microbiota). Such persistent modifications could predispose young adults to inflammatory bowel diseases (IBD). We recently observed that the dietary supplementation of four micronutrients to dams augmented colitis susceptibility in murine offspring in association with mucosal microbiota composition changes. In this study the effects of the four micronutrients on the microbiota of dams and female mice was examined. Additionally, age dependent microbiota composition shifts during pediatric development were delineated from the previous offspring data sets. Maternal and adult female microbiota did not separate secondary to the nutritional intervention. Significant microbiota composition changes occurred from postnatal day 30 (P30) to P90 at the level of 1 phylum and 15 genera. Most of these changes were absent or opposite in the maternally supplemented offspring. Nutritionally induced alterations in mucosal microbiota maturation may be contributors to colitis susceptibility in mammals.


Subject(s)
Bacteria/classification , Bacteria/drug effects , Biota , Colon/microbiology , Micronutrients/metabolism , Animals , Bacteria/genetics , Feces/microbiology , Female , Metagenome , Mice , Mice, Inbred C57BL
2.
Hum Mol Genet ; 20(9): 1687-96, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21296867

ABSTRACT

Developmental epigenetic changes, such as DNA methylation, have been recognized as potential pathogenic factors in inflammatory bowel diseases, the hallmark of which is an exaggerated immune response against luminal microbes. A methyl-donor (MD) diet can modify DNA methylation at select murine genomic loci during early development. The components of the MDs are routinely incorporated into prenatal human supplements. Therefore, we studied the effects of maternal MD supplementation on offspring colitis susceptibility and colonic mucosal DNA methylation and gene expression changes in mice as a model. Additionally, we investigated the offspring mucosal microbiomic response to the maternal dietary supplementation. Colitis was induced by dextran sulfate sodium. Colonic mucosa from offspring of MD-supplemented mothers following reversal to control diet at weaning was interrogated by methylation-specific microarrays and pyrosequencing at postnatal days 30 (P30) and P90. Transcriptomic changes were analyzed by microarray profiling and real-time reverse transcription polymerase chain reaction. The mucosal microbiome was studied by high throughput pyrosequencing of 16S rRNA. Maternal MD supplementation induced a striking susceptibility to colitis in offspring. This phenotype was associated with colonic mucosal DNA methylation and expression changes. Metagenomic analyses did not reveal consistent bacteriomic differences between P30 and P90, but showed a prolonged effect of the diet on the offspring mucosal microbiome. In conclusion, maternal MD supplementation increases offspring colitis susceptibility that associates with persistent epigenetic and prolonged microbiomic changes. These findings underscore that epigenomic reprogramming relevant to mammalian colitis can occur during early development in response to maternal dietary modifications.


Subject(s)
Colitis/metabolism , Dietary Supplements/adverse effects , Disease Susceptibility , Epigenesis, Genetic , Intestinal Mucosa/microbiology , Maternal Nutritional Physiological Phenomena , Metagenome , Prenatal Exposure Delayed Effects/metabolism , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Betaine/administration & dosage , Betaine/adverse effects , Choline/administration & dosage , Choline/adverse effects , Colitis/etiology , Colitis/genetics , Colitis/microbiology , DNA Methylation , Disease Susceptibility/metabolism , Female , Folic Acid/administration & dosage , Folic Acid/adverse effects , Humans , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Pedigree , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/microbiology , Vitamin B 12/administration & dosage , Vitamin B 12/adverse effects
3.
FASEB J ; 25(5): 1449-60, 2011 May.
Article in English | MEDLINE | ID: mdl-21228220

ABSTRACT

The connection between intestinal microbiota and host physiology is increasingly becoming recognized. The details of this dynamic interaction, however, remain to be explored. Toll-like receptor 2 (Tlr2) is important for its role in bacterial recognition, intestinal inflammation, and obesity-related metabolic changes. Therefore, we sought to determine the epigenomic and metagenomic consequences of Tlr2 deficiency in the colonic mucosa of mice to gain insights into biological pathways that shape the interface between the gut microbiota and the mammalian host. Colonic mucosa from wild type (WT) and Tlr2(-/-) C57BL/6 mice was interrogated by microarrays specific for DNA methylation and gene expression. The mucosal microbiome was studied by next-generation pyrosequencing of bacterial 16S rRNA. The expression of genes involved in immune processes was significantly modified by the absence of Tlr2, a number of which correlated with DNA methylation changes. The epigenomic and transcriptomic modifications associated with alteration in mucosal microbial composition. Several bacterial species, including members of the Firmicutes were significantly different in abundance between WT and Tlr2(-/-) animals. This manuscript highlights the intimate interrelationships between expression of immune-related genes and immunity pathways in the host with compositional and functional differences of the mammalian microbiome.


Subject(s)
Colon/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Toll-Like Receptor 2/metabolism , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Computational Biology , DNA Methylation/genetics , Epigenomics , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/immunology , Male , Metabolic Syndrome/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , RNA, Ribosomal, 16S/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Toll-Like Receptor 2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...