Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Clin Invest ; 134(9)2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38512401

RESUMEN

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.


Asunto(s)
Carbohidratos de la Dieta , Microbioma Gastrointestinal , Infecciones por Klebsiella , Klebsiella pneumoniae , Klebsiella pneumoniae/metabolismo , Humanos , Ratones , Animales , Infecciones por Klebsiella/microbiología , Infecciones por Klebsiella/prevención & control , Carbohidratos de la Dieta/metabolismo , Femenino , Masculino , Fibras de la Dieta/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/metabolismo , Intestinos/microbiología
2.
Nat Microbiol ; 9(1): 85-94, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38168616

RESUMEN

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.


Asunto(s)
Toxinas Bacterianas , Animales , Ratones , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Bacterias/metabolismo , Colon/metabolismo , Bacteroides fragilis/genética
3.
Cell Mol Gastroenterol Hepatol ; 17(1): 131-148, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37739064

RESUMEN

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.


Asunto(s)
Enterobacteriaceae , Enfermedades Inflamatorias del Intestino , Animales , Humanos , Enterobacteriaceae/metabolismo , Disbiosis , Enfermedades Inflamatorias del Intestino/microbiología , Carnitina/metabolismo , Ácidos Grasos/metabolismo , Escherichia coli , Biomarcadores
4.
bioRxiv ; 2023 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-37292978

RESUMEN

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.

5.
Cell Host Microbe ; 22(4): 443-448.e5, 2017 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-28943327

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Bacteroides fragilis/patogenicidad , Metaloendopeptidasas/metabolismo , Mucina 2/metabolismo , Factores de Transcripción/metabolismo , Animales , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Toxinas Bacterianas/genética , Bacteroides fragilis/genética , Neoplasias Colorrectales/prevención & control , Humanos , Enfermedades Inflamatorias del Intestino/prevención & control , Intestinos/microbiología , Metaloendopeptidasas/genética , Ratones , Ratones Endogámicos C57BL , Mucina 2/genética , Moco/metabolismo , Factores de Transcripción/genética , Virulencia
6.
Gut Microbes ; 8(4): 374-383, 2017 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-28632016

RESUMEN

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.


Asunto(s)
Infecciones por Bacteroides/microbiología , Bacteroides fragilis/fisiología , Bacteroides fragilis/patogenicidad , Islas Genómicas , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Bacteroides fragilis/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Interacciones Microbianas , Virulencia
7.
EMBO Rep ; 17(9): 1281-91, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27432285

RESUMEN

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.


Asunto(s)
Colitis/microbiología , Interacciones Huésped-Patógeno , Mucosa Intestinal/microbiología , Interacciones Microbianas , Animales , Antibiosis , Bacteroides fragilis/clasificación , Bacteroides fragilis/genética , Colitis/inducido químicamente , Colitis/patología , Colitis/prevención & control , Modelos Animales de Enfermedad , Inmunidad , Mucosa Intestinal/patología , Ratones
8.
Nat Med ; 22(5): 563-7, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27089515

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Infecciones por Bacteroides/metabolismo , Colitis/metabolismo , Proteasas de Cisteína/metabolismo , Metaloendopeptidasas/metabolismo , Sepsis/metabolismo , Animales , Bacteriemia/metabolismo , Bacteriemia/microbiología , Bacterias Anaerobias , Toxinas Bacterianas/metabolismo , Infecciones por Bacteroides/microbiología , Bacteroides fragilis , Western Blotting , Colitis/microbiología , Cristalografía por Rayos X , Técnica del Anticuerpo Fluorescente , Ratones , Sepsis/microbiología , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA