Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Gastroenterology ; 162(1): 166-178, 2022 01.
Article in English | MEDLINE | ID: mdl-34606847

ABSTRACT

BACKGROUND & AIMS: Fecal microbiota transplantation (FMT) is an emerging treatment modality for ulcerative colitis (UC). Several randomized controlled trials have shown efficacy for FMT in the treatment of UC, but a better understanding of the transferable microbiota and their immune impact is needed to develop more efficient microbiome-based therapies for UC. METHODS: Metagenomic analysis and strain tracking was performed on 60 donor and recipient samples receiving FMT for active UC. Sorting and sequencing of immunoglobulin (Ig) A-coated microbiota (called IgA-seq) was used to define immune-reactive microbiota. Colonization of germ-free or genetically engineered mice with patient-derived strains was performed to determine the mechanism of microbial impact on intestinal immunity. RESULTS: Metagenomic analysis defined a core set of donor-derived transferable bacterial strains in UC subjects achieving clinical response, which predicted response in an independent trial of FMT for UC. IgA-seq of FMT recipient samples and gnotobiotic mice colonized with donor microbiota identified Odoribacter splanchnicus as a transferable strain shaping mucosal immunity, which correlated with clinical response and the induction of mucosal regulatory T cells. Colonization of mice with O splanchnicus led to an increase in Foxp3+/RORγt+ regulatory T cells, induction of interleukin (IL) 10, and production of short chain fatty acids, all of which were required for O splanchnicus to limit colitis in mouse models. CONCLUSIONS: This work provides the first evidence of transferable, donor-derived strains that correlate with clinical response to FMT in UC and reveals O splanchnicus as a key component promoting both metabolic and immune cell protection from colitis. These mechanistic features will help enable strategies to enhance the efficacy of microbial therapy for UC. Clinicaltrials.gov ID NCT02516384.


Subject(s)
Bacteroidetes/immunology , Colitis/therapy , Colon/microbiology , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Immunoglobulin A/immunology , Intestinal Mucosa/microbiology , Animals , Bacteroidetes/genetics , Bacteroidetes/metabolism , Clinical Trials as Topic , Colitis/immunology , Colitis/metabolism , Colitis/microbiology , Colitis, Ulcerative/diagnosis , Colitis, Ulcerative/immunology , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/microbiology , Colon/immunology , Colon/metabolism , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/immunology , Germ-Free Life , Humans , Immunity, Mucosal , Immunoglobulin A/genetics , Immunoglobulin A/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/metabolism , Intraepithelial Lymphocytes/microbiology , Metagenome , Metagenomics , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/microbiology , Treatment Outcome
2.
Cell Host Microbe ; 29(4): 607-619.e8, 2021 04 14.
Article in English | MEDLINE | ID: mdl-33539767

ABSTRACT

Adherent-invasive E. coli (AIEC) are enriched in the intestinal microbiota of patients with Crohn's disease (CD) and promote intestinal inflammation. Yet, how AIEC metabolism of nutrients impacts intestinal homeostasis is poorly defined. Here, we show that AIEC encoding the large subunit of propanediol dehydratase (PduC), which facilitates the utilization of fucose fermentation product 1,2-propanediol, are increased in the microbiome of CD patients and drive AIEC-induced intestinal T cell inflammation. In murine models, CX3CR1+ mononuclear phagocytes (MNP) are required for PduC-dependent induction of T helper 17 (Th17) cells and interleukin-1ß (IL-1ß) production that leads to AIEC-induced inflammatory colitis. Activation of this inflammatory cascade requires the catalytic activity of PduC to generate propionate, which synergizes with lipopolysaccharide (LPS) to induce IL-1ß by MNPs. Disrupting fucose availability limits AIEC-induced propionate production and intestinal inflammation. These findings identify MNPs as metabolic sensors linking AIEC metabolism with intestinal inflammation and identify microbial metabolism as a potential therapeutic target in Crohn's disease treatment.


Subject(s)
Crohn Disease/metabolism , Escherichia coli Infections/metabolism , Escherichia coli/metabolism , Inflammation/metabolism , Intestines/immunology , Phagocytes/metabolism , Propylene Glycols/metabolism , Animals , Bacterial Adhesion , Crohn Disease/microbiology , Escherichia coli Infections/microbiology , Female , Host-Pathogen Interactions , Humans , Immunity , Interleukin-1beta , Intestinal Mucosa/metabolism , Intestines/microbiology , Male , Mice , Phagocytes/immunology , Th17 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...