Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS Biol ; 15(2): e2000689, 2017 02.
Article in English | MEDLINE | ID: mdl-28207737

ABSTRACT

Sustaining a balanced intestinal microbial community is critical for maintaining intestinal health and preventing chronic inflammation. The gut is a highly dynamic environment, subject to periodic waves of peristaltic activity. We hypothesized that this dynamic environment is a prerequisite for a balanced microbial community and that the enteric nervous system (ENS), a chief regulator of physiological processes within the gut, profoundly influences gut microbiota composition. We found that zebrafish lacking an ENS due to a mutation in the Hirschsprung disease gene, sox10, develop microbiota-dependent inflammation that is transmissible between hosts. Profiling microbial communities across a spectrum of inflammatory phenotypes revealed that increased levels of inflammation were linked to an overabundance of pro-inflammatory bacterial lineages and a lack of anti-inflammatory bacterial lineages. Moreover, either administering a representative anti-inflammatory strain or restoring ENS function corrected the pathology. Thus, we demonstrate that the ENS modulates gut microbiota community membership to maintain intestinal health.


Subject(s)
Enteric Nervous System/physiology , Gastrointestinal Microbiome , Intestines/microbiology , Animals , Bacteria/growth & development , Bacteria/isolation & purification , Cell Count , Colony Count, Microbial , Dysbiosis/genetics , Dysbiosis/microbiology , Dysbiosis/pathology , Enteric Nervous System/cytology , Gene Expression Regulation , Inflammation/genetics , Inflammation/pathology , Intestines/pathology , Leukocyte Count , Models, Biological , Mutation/genetics , Neutrophils/metabolism , Phylogeny , SOXE Transcription Factors/metabolism , Stem Cell Transplantation , Zebrafish , Zebrafish Proteins/metabolism
2.
Dis Model Mech ; 9(2): 187-98, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26681746

ABSTRACT

Animal hosts must co-exist with beneficial microbes while simultaneously being able to mount rapid, non-specific, innate immune responses to pathogenic microbes. How this balance is achieved is not fully understood, and disruption of this relationship can lead to disease. Excessive inflammatory responses to resident microbes are characteristic of certain gastrointestinal pathologies such as inflammatory bowel disease (IBD). The immune dysregulation of IBD has complex genetic underpinnings that cannot be fully recapitulated with single-gene-knockout models. A deeper understanding of the genetic regulation of innate immune responses to resident microbes requires the ability to measure immune responses in the presence and absence of the microbiota using vertebrate models with complex genetic variation. Here, we describe a new gnotobiotic vertebrate model to explore the natural genetic variation that contributes to differences in innate immune responses to microbiota. Threespine stickleback, Gasterosteus aculeatus, has been used to study the developmental genetics of complex traits during the repeated evolution from ancestral oceanic to derived freshwater forms. We established methods to rear germ-free stickleback larvae and gnotobiotic animals monoassociated with single bacterial isolates. We characterized the innate immune response of these fish to resident gut microbes by quantifying the neutrophil cells in conventionally reared monoassociated or germ-free stickleback from both oceanic and freshwater populations grown in a common intermediate salinity environment. We found that oceanic and freshwater fish in the wild and in the laboratory share many intestinal microbial community members. However, oceanic fish mount a strong immune response to residential microbiota, whereas freshwater fish frequently do not. A strong innate immune response was uniformly observed across oceanic families, but this response varied among families of freshwater fish. The gnotobiotic stickleback model that we have developed therefore provides a platform for future studies mapping the natural genetic basis of the variation in immune response to microbes.


Subject(s)
Fishes/microbiology , Immunity, Innate , Intestines/microbiology , Microbiota , Animals , Fresh Water , Seawater
3.
ISME J ; 5(10): 1595-608, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21472014

ABSTRACT

Experimental analysis of gut microbial communities and their interactions with vertebrate hosts is conducted predominantly in domesticated animals that have been maintained in laboratory facilities for many generations. These animal models are useful for studying coevolved relationships between host and microbiota only if the microbial communities that occur in animals in lab facilities are representative of those that occur in nature. We performed 16S rRNA gene sequence-based comparisons of gut bacterial communities in zebrafish collected recently from their natural habitat and those reared for generations in lab facilities in different geographic locations. Patterns of gut microbiota structure in domesticated zebrafish varied across different lab facilities in correlation with historical connections between those facilities. However, gut microbiota membership in domesticated and recently caught zebrafish was strikingly similar, with a shared core gut microbiota. The zebrafish intestinal habitat therefore selects for specific bacterial taxa despite radical differences in host provenance and domestication status.


Subject(s)
Bacteria/classification , Gastrointestinal Tract/microbiology , Metagenome , Zebrafish/microbiology , Animals , Bacteria/genetics , Bacteria/isolation & purification , Intestines/microbiology , Molecular Sequence Data , RNA, Bacterial/genetics , RNA, Ribosomal, 16S/genetics , Zebrafish/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...