Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Mol Gastroenterol Hepatol ; 18(3): 101357, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38750900

ABSTRACT

BACKGROUND & AIMS: Crohn's disease is associated with alterations in the gut microbiome and metabolome described as dysbiosis. We characterized the microbial and metabolic consequences of ileal resection, the most common Crohn's disease surgery. METHODS: Patients with and without intestinal resection were identified from the Diet to Induce Remission in Crohn's Disease and Study of a Prospective Adult Research Cohort with Inflammatory Bowel Disease studies. Stool samples were analyzed with shotgun metagenomics sequencing. Fecal butyrate was measured with 1H nuclear magnetic resonance spectroscopy. Fecal bile acids and plasma 7α-hydroxy-4-cholesten-3-one (C4) was measured with mass spectrometry. RESULTS: Intestinal resection was associated with reduced alpha diversity and altered beta diversity with increased Proteobacteria and reduced Bacteroidetes and Firmicutes. Surgery was associated with higher representation of genes in the KEGG pathway for ABC transporters and reduction in genes related to bacterial metabolism. Surgery was associated with reduced concentration of the But gene but this did not translate to reduced fecal butyrate concentration. Surgery was associated with decreased abundance of bai operon genes, with increased plasma C4 concentration, increased primary bile acids and reduced secondary bile acids, including isoLCA. Additionally, Egerthella lenta, Adlercreutzia equalofaciens, and Gordonibacter pamelaeae were lower in abundance among patients with prior surgery in both cohorts. CONCLUSIONS: In 2 different populations, prior surgery in Crohn's disease is associated with altered fecal microbiome. Patients who had undergone ileal resection had reduction in the potentially beneficial bacteria E lenta and related actinobacteria and secondary bile acids, including isoLCA, suggesting that these could be biomarkers of patients at higher risk for disease progression.

2.
J Clin Invest ; 134(9)2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38512401

ABSTRACT

Bacterial translocation from the gut microbiota is a source of sepsis in susceptible patients. Previous work suggests that overgrowth of gut pathobionts, including Klebsiella pneumoniae, increases the risk of disseminated infection. Our data from a human dietary intervention study found that, in the absence of fiber, K. pneumoniae bloomed during microbiota recovery from antibiotic treatment. We thus hypothesized that dietary nutrients directly support or suppress colonization of this gut pathobiont in the microbiota. Consistent with our study in humans, complex carbohydrates in dietary fiber suppressed the colonization of K. pneumoniae and allowed for recovery of competing commensals in mouse models. In contrast, through ex vivo and in vivo modeling, we identified simple carbohydrates as a limiting resource for K. pneumoniae in the gut. As proof of principle, supplementation with lactulose, a nonabsorbed simple carbohydrate and an FDA-approved therapy, increased colonization of K. pneumoniae. Disruption of the intestinal epithelium led to dissemination of K. pneumoniae into the bloodstream and liver, which was prevented by dietary fiber. Our results show that dietary simple and complex carbohydrates were critical not only in the regulation of pathobiont colonization but also disseminated infection, suggesting that targeted dietary interventions may offer a preventative strategy in high-risk patients.


Subject(s)
Dietary Carbohydrates , Gastrointestinal Microbiome , Klebsiella Infections , Klebsiella pneumoniae , Klebsiella pneumoniae/metabolism , Humans , Mice , Animals , Klebsiella Infections/microbiology , Klebsiella Infections/prevention & control , Dietary Carbohydrates/metabolism , Female , Male , Dietary Fiber/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism , Intestines/microbiology
3.
Nat Microbiol ; 9(1): 85-94, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38168616

ABSTRACT

Bacterial toxins are well-studied virulence factors; however, recent studies have revealed their importance in bacterial niche adaptation. Enterotoxigenic Bacteroides fragilis (ETBF) expresses B. fragilis toxin (BFT) that we hypothesized may contribute to both colonic epithelial injury and niche acquisition. We developed a vertical transmission model for ETBF in mice that showed that BFT enabled ETBF to access a lamina propria (LP) niche during colonic microbiome development that was inaccessible to non-toxigenic B. fragilis. LP entry by ETBF required BFT metalloprotease activity, and showed temporal restriction to the pre-weaning period, dependent on goblet-cell-associated passages. In situ single-cell analysis showed bft expression at the apical epithelial surface and within the LP. BFT expression increased goblet cell number and goblet-cell-associated passage formation. These findings define a paradigm by which bacterial toxin expression specifies developmental niche acquisition, suggesting that a selective advantage conferred by a toxin may impact long-term host health.


Subject(s)
Bacterial Toxins , Animals , Mice , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Bacteria/metabolism , Colon/metabolism , Bacteroides fragilis/genetics
4.
Cell Mol Gastroenterol Hepatol ; 17(1): 131-148, 2024.
Article in English | MEDLINE | ID: mdl-37739064

ABSTRACT

BACKGROUND & AIMS: Altered plasma acylcarnitine levels are well-known biomarkers for a variety of mitochondrial fatty acid oxidation disorders and can be used as an alternative energy source for the intestinal epithelium when short-chain fatty acids are low. These membrane-permeable fatty acid intermediates are excreted into the gut lumen via bile and are increased in the feces of patients with inflammatory bowel disease (IBD). METHODS: Herein, based on studies in human subjects, animal models, and bacterial cultures, we show a strong positive correlation between fecal carnitine and acylcarnitines and the abundance of Enterobacteriaceae in IBD where they can be consumed by bacteria both in vitro and in vivo. RESULTS: Carnitine metabolism promotes the growth of Escherichia coli via anaerobic respiration dependent on the cai operon, and acetylcarnitine dietary supplementation increases fecal carnitine levels with enhanced intestinal colonization of the enteric pathogen Citrobacter rodentium. CONCLUSIONS: In total, these results indicate that the increased luminal concentrations of carnitine and acylcarnitines in patients with IBD may promote the expansion of pathobionts belonging to the Enterobacteriaceae family, thereby contributing to disease pathogenesis.


Subject(s)
Enterobacteriaceae , Inflammatory Bowel Diseases , Animals , Humans , Enterobacteriaceae/metabolism , Dysbiosis , Inflammatory Bowel Diseases/microbiology , Carnitine/metabolism , Fatty Acids/metabolism , Escherichia coli , Biomarkers
5.
bioRxiv ; 2023 Jun 11.
Article in English | MEDLINE | ID: mdl-37292978

ABSTRACT

Dysbiosis of the gut microbiota is increasingly appreciated as both a consequence and precipitant of human disease. The outgrowth of the bacterial family Enterobacteriaceae is a common feature of dysbiosis, including the human pathogen Klebsiella pneumoniae . Dietary interventions have proven efficacious in the resolution of dysbiosis, though the specific dietary components involved remain poorly defined. Based on a previous human diet study, we hypothesized that dietary nutrients serve as a key resource for the growth of bacteria found in dysbiosis. Through human sample testing, and ex-vivo , and in vivo modeling, we find that nitrogen is not a limiting resource for the growth of Enterobacteriaceae in the gut, contrary to previous studies. Instead, we identify dietary simple carbohydrates as critical in colonization of K. pneumoniae . We additionally find that dietary fiber is necessary for colonization resistance against K. pneumoniae , mediated by recovery of the commensal microbiota, and protecting the host against dissemination from the gut microbiota during colitis. Targeted dietary therapies based on these findings may offer a therapeutic strategy in susceptible patients with dysbiosis.

6.
Cell Host Microbe ; 22(4): 443-448.e5, 2017 Oct 11.
Article in English | MEDLINE | ID: mdl-28943327

ABSTRACT

Intestinal microbes are recognized for their role in human disease. Enterotoxigenic Bacteroides fragilis (ETBF) has been implicated in inflammatory bowel disease and colorectal cancer; however, colonization alone is insufficient to cause these illnesses. We hypothesized that homeostasis in healthy carriers is maintained by colonic mucus, the major constituent of which is the glycoprotein Muc2. We found that Muc2-deficient mice succumb to lethal disease from ETBF colonization in a B. fragilis toxin (BFT)-dependent manner. We identify a toxin regulator, the two-component system RprXY, which suppresses BFT expression in vitro and in vivo. Overexpression of either component was sufficient to prevent lethal disease in Muc2-deficient mice. Our studies demonstrate that homeostasis in the context of ETBF colonization is dependent on a dynamic interaction between intestinal mucus, a bacterial toxin, and a toxin regulatory system. Regulation of virulence may offer a therapeutic target to maintain intestinal homeostasis in susceptible patients.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Bacteroides fragilis/pathogenicity , Metalloendopeptidases/metabolism , Mucin-2/metabolism , Transcription Factors/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Toxins/genetics , Bacteroides fragilis/genetics , Colorectal Neoplasms/prevention & control , Humans , Inflammatory Bowel Diseases/prevention & control , Intestines/microbiology , Metalloendopeptidases/genetics , Mice , Mice, Inbred C57BL , Mucin-2/genetics , Mucus/metabolism , Transcription Factors/genetics , Virulence
7.
Gut Microbes ; 8(4): 374-383, 2017 07 04.
Article in English | MEDLINE | ID: mdl-28632016

ABSTRACT

The mature microbiome is a stable ecosystem that resists perturbation despite constant host exposure to exogenous microbes. However, the microbial mechanisms determining microbiome development and composition are poorly understood. We recently demonstrated that a non-toxigenic B. fragilis (NTBF) strain restricts enteric colonization by an enterotoxigenic (ETBF) strain dependent on a type VI secretion system (T6SS). We show here that a second enterotoxigenic strain is competent to colonize, dependent on the Bacteroides fragilis pathogenicity island (BFPAI). Additional data showing complex environmental regulation of the Bacteroides fragilis toxin (BFT) suggest that virulence factors may be adapted to modify the colonic niche to provide a strain-specific colonization advantage. We conclude that more complex models of host-microbe-microbiome interactions are needed to investigate this hypothesis.


Subject(s)
Bacteroides Infections/microbiology , Bacteroides fragilis/physiology , Bacteroides fragilis/pathogenicity , Genomic Islands , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacteroides fragilis/genetics , Humans , Mice , Mice, Inbred C57BL , Microbial Interactions , Virulence
8.
EMBO Rep ; 17(9): 1281-91, 2016 09.
Article in English | MEDLINE | ID: mdl-27432285

ABSTRACT

The microbiota is a major source of protection against intestinal pathogens; however, the specific bacteria and underlying mechanisms involved are not well understood. As a model of this interaction, we sought to determine whether colonization of the murine host with symbiotic non-toxigenic Bacteroides fragilis could limit acquisition of pathogenic enterotoxigenic B. fragilis We observed strain-specific competition with toxigenic B. fragilis, dependent upon type VI secretion, identifying an effector-immunity pair that confers pathogen exclusion. Resistance against host acquisition of a second non-toxigenic strain was also uncovered, revealing a broader function of type VI secretion systems in determining microbiota composition. The competitive exclusion of enterotoxigenic B. fragilis by a non-toxigenic strain limited toxin exposure and protected the host against intestinal inflammatory disease. Our studies demonstrate a novel role of type VI secretion systems in colonization resistance against a pathogen. This understanding of bacterial competition may be utilized to define a molecularly targeted probiotic strategy.


Subject(s)
Colitis/microbiology , Host-Pathogen Interactions , Intestinal Mucosa/microbiology , Microbial Interactions , Animals , Antibiosis , Bacteroides fragilis/classification , Bacteroides fragilis/genetics , Colitis/chemically induced , Colitis/pathology , Colitis/prevention & control , Disease Models, Animal , Immunity , Intestinal Mucosa/pathology , Mice
9.
Nat Med ; 22(5): 563-7, 2016 05.
Article in English | MEDLINE | ID: mdl-27089515

ABSTRACT

Bacteroides fragilis is the leading cause of anaerobic bacteremia and sepsis. Enterotoxigenic strains that produce B. fragilis toxin (BFT, fragilysin) contribute to colitis and intestinal malignancy, yet are also isolated in bloodstream infection. It is not known whether these strains harbor unique genetic determinants that confer virulence in extra-intestinal disease. We demonstrate that BFT contributes to sepsis in mice, and we identify a B. fragilis protease called fragipain (Fpn) that is required for the endogenous activation of BFT through the removal of its auto-inhibitory prodomain. Structural analysis of Fpn reveals a His-Cys catalytic dyad that is characteristic of C11-family cysteine proteases that are conserved in multiple pathogenic Bacteroides spp. and Clostridium spp. Fpn-deficient, enterotoxigenic B. fragilis has an attenuated ability to induce sepsis in mice; however, Fpn is dispensable in B. fragilis colitis, wherein host proteases mediate BFT activation. Our findings define a role for B. fragilis enterotoxin and its activating protease in the pathogenesis of bloodstream infection, which indicates a greater complexity of cellular targeting and activity of BFT than previously recognized. The expression of fpn by both toxigenic and nontoxigenic strains suggests that this protease may contribute to anaerobic sepsis in ways that extend beyond its role in toxin activation. It could thus potentially serve as a target for disease modification.


Subject(s)
Bacterial Proteins/metabolism , Bacteroides Infections/metabolism , Colitis/metabolism , Cysteine Proteases/metabolism , Metalloendopeptidases/metabolism , Sepsis/metabolism , Animals , Bacteremia/metabolism , Bacteremia/microbiology , Bacteria, Anaerobic , Bacterial Toxins/metabolism , Bacteroides Infections/microbiology , Bacteroides fragilis , Blotting, Western , Colitis/microbiology , Crystallography, X-Ray , Fluorescent Antibody Technique , Mice , Sepsis/microbiology , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...