Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
2.
J Virol ; 96(22): e0085522, 2022 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-36342297

RESUMEN

Human norovirus (HNoV) accounts for one-fifth of all acute viral gastroenteritis worldwide and an economic burden of ~$60 billion globally. The lack of treatment options against HNoV is in part due to the lack of cultivation systems. Recently, a model of infection in biopsy-derived human intestinal enteroids (HIE) has been described: 3D-HIE are first dispersed in 2D-monolayers and differentiated prior to infection, resulting in a labor-intensive, time-consuming procedure. Here, we present an alternative protocol for HNoV infection of 3D-HIE. We found that 3D-HIE differentiated as efficiently as 2D-monolayers. In addition, immunofluorescence-based quantification of UEA-1, a lectin that stains the villus brush border, revealed that ~80% of differentiated 3D-HIE spontaneously undergo polarity inversion, allowing for viral infection without the need for microinjection. Infection with HNoV GII.4-positive stool samples attained a fold-increase over inoculum of ~2 Log10 at 2 days postinfection or up to 3.5 Log10 when ruxolitinib, a JAK1/2-inhibitor, was added. Treatment of GII.4-infected 3D-HIE with the polymerase inhibitor 2'-C-Methylcytidine (2CMC) and other antivirals showed a reduction in viral infection, suggesting that 3D-HIE are an excellent platform to test anti-infectives. The transcriptional host response to HNoV was then investigated by RNA sequencing in infected versus uninfected 3D-HIE in the presence of ruxolitinib to focus on virus-associated signatures while limiting interferon-stimulated gene signatures. The analysis revealed upregulated hormone and neurotransmitter signal transduction pathways and downregulated glycolysis and hypoxia-response pathways upon HNoV infection. Overall, 3D-HIE have proven to be a highly robust model to study HNoV infection, screen antivirals, and to investigate the host response to HNoV infection. IMPORTANCE The human norovirus (HNoV) clinical and socio-economic impact calls for immediate action in the development of anti-infectives. Physiologically relevant in vitro models are hence needed to study HNoV biology, tropism, and mechanisms of viral-associated disease, and also as a platform to identify antiviral agents. Biopsy-derived human intestinal enteroids are a biomimetic of the intestinal epithelium and were recently described as a model that supports HNoV infection. However, the established protocol is time-consuming and labor-intensive. Therefore, we sought to develop a simplified and robust alternative model of infection in 3D enteroids that undergoes differentiation and spontaneous polarity inversion. Advantages of this model are the shorter experimental time, better infection yield, and spatial integrity of the intestinal epithelium. This model is potentially suitable for the study of other pathogens that infect intestinal cells from the apical surface but also for unraveling the interactions between intestinal epithelium and indigenous bacteria of the human microbiome.


Asunto(s)
Infecciones por Caliciviridae , Gastroenteritis , Norovirus , Humanos , Norovirus/fisiología , Pirazoles , Antivirales/farmacología
3.
JCI Insight ; 7(19)2022 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-36099053

RESUMEN

A high-fat diet (HFD) contributes to the increased incidence of colorectal cancer, but the mechanisms are unclear. We found that R-spondin 3 (Rspo3), a ligand for leucine-rich, repeat-containing GPCR 4 and 5 (LGR4 and LGR5), was the main subtype of R-spondins and was produced by myofibroblasts beneath the crypts in the intestine. HFD upregulated colonic Rspo3, LGR4, LGR5, and ß-catenin gene expression in specific pathogen-free rodents, but not in germ-free mice, and the upregulations were prevented by the bile acid (BA) binder cholestyramine or antibiotic treatment, indicating mediation by both BA and gut microbiota. Cholestyramine or antibiotic treatments prevented HFD-induced enrichment of members of the Lachnospiraceae and Rumincoccaceae, which can transform primary BA into secondary BA. Oral administration of deoxycholic acid (DCA), or inoculation of a combination of the BA deconjugator Lactobacillus plantarum and 7α-dehydroxylase-containing Clostridium scindens with an HFD to germ-free mice increased serum DCA and colonic Rspo3 mRNA levels, indicating that formation of secondary BA by gut microbiota is responsible for HFD-induced upregulation of Rspo3. In primary myofibroblasts, DCA increased Rspo3 mRNA via TGR5. Finally, we showed that cholestyramine or conditional deletion of Rspo3 prevented HFD- or DCA-induced intestinal proliferation. We conclude that secondary BA is responsible for HFD-induced upregulation of Rspo3, which, in turn, mediates HFD-induced intestinal epithelial proliferation.


Asunto(s)
Ácidos y Sales Biliares , Dieta Alta en Grasa , Animales , Antibacterianos , Proliferación Celular , Resina de Colestiramina , Ácido Desoxicólico , Dieta Alta en Grasa/efectos adversos , Intestinos , Leucina , Ligandos , Ratones , ARN Mensajero , Regulación hacia Arriba , beta Catenina/metabolismo
4.
Cell Rep ; 40(3): 111093, 2022 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-35858565

RESUMEN

Pathobionts employ unique metabolic adaptation mechanisms to maximize their growth in disease conditions. Adherent-invasive Escherichia coli (AIEC), a pathobiont enriched in the gut mucosa of patients with inflammatory bowel disease (IBD), utilizes diet-derived L-serine to adapt to the inflamed gut. Therefore, the restriction of dietary L-serine starves AIEC and limits its fitness advantage. Here, we find that AIEC can overcome this nutrient limitation by switching the nutrient source from the diet to the host cells in the presence of mucolytic bacteria. During diet-derived L-serine restriction, the mucolytic symbiont Akkermansia muciniphila promotes the encroachment of AIEC to the epithelial niche by degrading the mucus layer. In the epithelial niche, AIEC acquires L-serine from the colonic epithelium and thus proliferates. Our work suggests that the indirect metabolic network between pathobionts and commensal symbionts enables pathobionts to overcome nutritional restriction and thrive in the gut.


Asunto(s)
Infecciones por Escherichia coli , Adhesión Bacteriana , Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Expectorantes/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Nutrientes , Serina/metabolismo
5.
JCI Insight ; 6(23)2021 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-34710061

RESUMEN

Oral conditions are relatively common in patients with inflammatory bowel disease (IBD). However, the contribution of oral maladies to gut inflammation remains unexplored. Here, we investigated the effect of periodontitis on disease phenotypes of patients with IBD. In all, 60 patients with IBD (42 with ulcerative colitis [UC] and 18 with Crohn's disease [CD]) and 45 healthy controls (HCs) without IBD were recruited for this clinical investigation. The effects of incipient periodontitis on the oral and gut microbiome as well as IBD characteristics were examined. In addition, patients were prospectively monitored for up to 12 months after enrollment. We found that, in both patients with UC and those with CD, the gut microbiome was significantly more similar to the oral microbiome than in HCs, suggesting that ectopic gut colonization by oral bacteria is increased in patients with IBD. Incipient periodontitis did not further enhance gut colonization by oral bacteria. The presence of incipient periodontitis did not significantly affect the clinical outcomes of patients with UC and CD. However, the short CD activity index increased in patients with CD with incipient periodontitis but declined or was unchanged during the study period in patients without periodontitis. Thus, early periodontitis may associate with worse clinically symptoms in some patients with CD.


Asunto(s)
Enfermedad de Crohn/complicaciones , Periodontitis/etiología , Adulto , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Periodontitis/patología , Estudios Prospectivos , Factores de Riesgo
6.
JCI Insight ; 5(20)2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33055426

RESUMEN

High-fat feeding (HFF) leads to gut dysbiosis through unclear mechanisms. We hypothesize that bile acids secreted in response to high-fat diets (HFDs) may act on intestinal Paneth cells, leading to gut dysbiosis. We found that HFF resulted in widespread taxonomic shifts in the bacteria of the ileal mucosa, characterized by depletion of Lactobacillus and enrichment of Akkermansia muciniphila, Clostridium XIVa, Ruminococcaceae, and Lachnospiraceae, which were prevented by the bile acid binder cholestyramine. Immunohistochemistry and in situ hybridization studies showed that G protein-coupled bile acid receptor (TGR5) expressed in Paneth cells was upregulated in the rats fed HFD or normal chow supplemented with cholic acid. This was accompanied by decreased lysozyme+ Paneth cells and α-defensin 5 and 6 and increased expression of XBP-1. Pretreatment with ER stress inhibitor 4PBA or with cholestyramine prevented these changes. Ileal explants incubated with deoxycholic acid or cholic acid caused a decrease in α-defensin 5 and 6 and an increase in XBP-1, which was prevented by TGR5 antibody or 4PBA. In conclusion, this is the first demonstration to our knowledge that TGR5 is expressed in Paneth cells. HFF resulted in increased bile acid secretion and upregulation of TGR5 expression in Paneth cells. Bile acid toxicity in Paneth cells contributes to gut dysbiosis induced by HFF.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Disbiosis/genética , Microbioma Gastrointestinal/genética , Receptores Acoplados a Proteínas G/genética , Proteína 1 de Unión a la X-Box/genética , Akkermansia/genética , Akkermansia/patogenicidad , Animales , Ácidos y Sales Biliares/efectos adversos , Ácidos y Sales Biliares/biosíntesis , Clostridium/genética , Clostridium/patogenicidad , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Disbiosis/inducido químicamente , Disbiosis/metabolismo , Disbiosis/patología , Microbioma Gastrointestinal/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Lactobacillus/genética , Lactobacillus/metabolismo , Masculino , Células de Paneth/metabolismo , Células de Paneth/microbiología , Células de Paneth/patología , Ratas , alfa-Defensinas/genética
7.
Cell ; 182(2): 447-462.e14, 2020 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-32758418

RESUMEN

The precise mechanism by which oral infection contributes to the pathogenesis of extra-oral diseases remains unclear. Here, we report that periodontal inflammation exacerbates gut inflammation in vivo. Periodontitis leads to expansion of oral pathobionts, including Klebsiella and Enterobacter species, in the oral cavity. Amassed oral pathobionts are ingested and translocate to the gut, where they activate the inflammasome in colonic mononuclear phagocytes, triggering inflammation. In parallel, periodontitis results in generation of oral pathobiont-reactive Th17 cells in the oral cavity. Oral pathobiont-reactive Th17 cells are imprinted with gut tropism and migrate to the inflamed gut. When in the gut, Th17 cells of oral origin can be activated by translocated oral pathobionts and cause development of colitis, but they are not activated by gut-resident microbes. Thus, oral inflammation, such as periodontitis, exacerbates gut inflammation by supplying the gut with both colitogenic pathobionts and pathogenic T cells.


Asunto(s)
Colitis/patología , Enterobacter/fisiología , Microbioma Gastrointestinal , Klebsiella/fisiología , Boca/microbiología , Animales , Colitis/microbiología , Colon/microbiología , Colon/patología , Modelos Animales de Enfermedad , Enterobacter/aislamiento & purificación , Femenino , Inflamasomas/metabolismo , Interleucina-10/deficiencia , Interleucina-10/genética , Interleucina-1beta/metabolismo , Klebsiella/aislamiento & purificación , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Periodontitis/microbiología , Periodontitis/patología , Células Th17/citología , Células Th17/inmunología , Células Th17/metabolismo
8.
Nat Med ; 26(4): 608-617, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32066975

RESUMEN

The involvement of host immunity in the gut microbiota-mediated colonization resistance to Clostridioides difficile infection (CDI) is incompletely understood. Here, we show that interleukin (IL)-22, induced by colonization of the gut microbiota, is crucial for the prevention of CDI in human microbiota-associated (HMA) mice. IL-22 signaling in HMA mice regulated host glycosylation, which enabled the growth of succinate-consuming bacteria Phascolarctobacterium spp. within the gut microbiome. Phascolarctobacterium reduced the availability of luminal succinate, a crucial metabolite for the growth of C. difficile, and therefore prevented the growth of C. difficile. IL-22-mediated host N-glycosylation is likely impaired in patients with ulcerative colitis (UC) and renders UC-HMA mice more susceptible to CDI. Transplantation of healthy human-derived microbiota or Phascolarctobacterium reduced luminal succinate levels and restored colonization resistance in UC-HMA mice. IL-22-mediated host glycosylation thus fosters the growth of commensal bacteria that compete with C. difficile for the nutritional niche.


Asunto(s)
Bacterias/crecimiento & desarrollo , Bacterias/metabolismo , Clostridioides difficile/inmunología , Infecciones por Clostridium/prevención & control , Microbioma Gastrointestinal/fisiología , Interleucinas/fisiología , Animales , Bacterias/efectos de los fármacos , Clostridioides difficile/efectos de los fármacos , Infecciones por Clostridium/inmunología , Enterocolitis Seudomembranosa/inmunología , Enterocolitis Seudomembranosa/metabolismo , Enterocolitis Seudomembranosa/microbiología , Enterocolitis Seudomembranosa/prevención & control , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Glicosilación/efectos de los fármacos , Interacciones Microbiota-Huesped/efectos de los fármacos , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/inmunología , Humanos , Interleucinas/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Veillonellaceae/efectos de los fármacos , Veillonellaceae/crecimiento & desarrollo , Veillonellaceae/metabolismo , Interleucina-22
9.
Inflamm Bowel Dis ; 26(11): 1748-1758, 2020 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-31971239

RESUMEN

BACKGROUND: Ulcerative colitis (UC) carries an increased risk of primary and recurrent Clostridiodes difficile infection (rCDI), and CDI is associated with UC flares. We hypothesized that specific fecal microbial changes associate with UC flare and rCDI. METHODS: We conducted a prospective observational cohort study of 57 patients with UC and CDI, CDI only, and UC only. Stool samples were collected at baseline, at the end of antibiotic therapy, and after reconstitution for 16S rRNA sequencing. The primary outcomes were recurrent UC flare and rCDI. Logistic regression and Lasso models were constructed for analysis. RESULTS: There were 21 (45.7%) patients with rCDI, whereas 11 (34.4%) developed UC flare. Patients with rCDI demonstrated significant interindividual (P = 0.008) and intraindividual differences (P = 0.004) in community structure by Jensen-Shannon distance (JSD) compared with non-rCDI. Two cross-validated Lasso regression models predicted risk of rCDI: a baseline model with female gender, hospitalization for UC in the past year, increased Ruminococcaceae and Verrucomicrobia, and decreased Eubacteriaceae, Enterobacteriaceae, Lachnospiraceae, and Veillonellaceae (AuROC, 0.94); and a model 14 days after completion of antibiotics with female gender, increased Shannon diversity, Ruminococcaceae and Enterobacteriaceae, and decreased community richness and Faecalibacterium (AuROC, 0.9). Adding JSD between baseline and post-treatment samples to the latter model improved fit (AuROC, 0.94). A baseline model including UC hospitalization in the past year and increased Bacteroidetes was associated with increased risk for UC flare (AuROC, 0.88). CONCLUSION: Fecal microbial features at baseline and after therapy predict rCDI risk in patients with and without UC. These results may help risk stratify patients to guide management.


Asunto(s)
Antibacterianos/uso terapéutico , Clostridioides difficile , Infecciones por Clostridium/microbiología , Colitis Ulcerosa/microbiología , Microbioma Gastrointestinal/fisiología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Infecciones por Clostridium/tratamiento farmacológico , Heces/microbiología , Femenino , Humanos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , ARN Ribosómico 16S/análisis , Recurrencia , Brote de los Síntomas , Adulto Joven
10.
Nat Mater ; 19(1): 118-126, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31427744

RESUMEN

While conventional approaches for inflammatory bowel diseases mainly focus on suppressing hyperactive immune responses, it remains unclear how to address disrupted intestinal barriers, dysbiosis of the gut commensal microbiota and dysregulated mucosal immune responses in inflammatory bowel diseases. Moreover, immunosuppressive agents can cause off-target systemic side effects and complications. Here, we report the development of hyaluronic acid-bilirubin nanomedicine (HABN) that accumulates in inflamed colonic epithelium and restores the epithelium barriers in a murine model of acute colitis. Surprisingly, HABN also modulates the gut microbiota, increasing the overall richness and diversity and markedly augmenting the abundance of Akkermansia muciniphila and Clostridium XIVα, which are microorganisms with crucial roles in gut homeostasis. Importantly, HABN associated with pro-inflammatory macrophages, regulated innate immune responses and exerted potent therapeutic efficacy against colitis. Our work sheds light on the impact of nanotherapeutics on gut homeostasis, microbiome and innate immune responses for the treatment of inflammatory diseases.


Asunto(s)
Bilirrubina/farmacología , Colitis/inmunología , Colitis/terapia , Ácido Hialurónico/farmacología , Akkermansia , Animales , Disbiosis/inmunología , Femenino , Microbioma Gastrointestinal/inmunología , Células HT29 , Homeostasis , Humanos , Sistema Inmunológico , Inmunosupresores/uso terapéutico , Inflamación , Mucosa Intestinal/patología , Intestinos/patología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Microbiota , Nanomedicina , Nanopartículas/química , Permeabilidad , Especies Reactivas de Oxígeno/metabolismo , Verrucomicrobia
11.
J Clin Invest ; 128(1): 267-280, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29202473

RESUMEN

Foods high in fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAPs) exacerbate symptoms of irritable bowel syndrome (IBS); however, their mechanism of action is unknown. We hypothesized that a high-FODMAP (HFM) diet increases visceral nociception by inducing dysbiosis and that the FODMAP-altered gut microbial community leads to intestinal pathology. We fed rats an HFM and showed that HFM increases rat fecal Gram-negative bacteria, elevates lipopolysaccharides (LPS), and induces intestinal pathology, as indicated by inflammation, barrier dysfunction, and visceral hypersensitivity (VH). These manifestations were prevented by antibiotics and reversed by low-FODMAP (LFM) diet. Additionally, intracolonic administration of LPS or fecal supernatant (FS) from HFM-fed rats caused intestinal barrier dysfunction and VH, which were blocked by the LPS antagonist LPS-RS or by TLR4 knockdown. Fecal LPS was higher in IBS patients than in healthy subjects (HS), and IBS patients on a 4-week LFM diet had improved IBS symptoms and reduced fecal LPS levels. Intracolonic administration of FS from IBS patients, but not FS from HS or LFM-treated IBS patients, induced VH in rats, which was ameliorated by LPS-RS. Our findings indicate that HFM-associated gut dysbiosis and elevated fecal LPS levels induce intestinal pathology, thereby modulating visceral nociception and IBS symptomatology, and might provide an explanation for the success of LFM diet in IBS patients.


Asunto(s)
Carbohidratos de la Dieta/efectos adversos , Disbiosis , Microbioma Gastrointestinal , Intestinos/microbiología , Síndrome del Colon Irritable , Lipopolisacáridos/toxicidad , Nocicepción , Animales , Carbohidratos de la Dieta/farmacología , Disbiosis/inducido químicamente , Disbiosis/genética , Disbiosis/metabolismo , Disbiosis/microbiología , Técnicas de Silenciamiento del Gen , Síndrome del Colon Irritable/inducido químicamente , Síndrome del Colon Irritable/genética , Síndrome del Colon Irritable/metabolismo , Síndrome del Colon Irritable/microbiología , Masculino , Ratas , Ratas Wistar , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
12.
Science ; 356(6335): 315-319, 2017 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-28428425

RESUMEN

The high susceptibility of neonates to infections has been assumed to be due to immaturity of the immune system, but the mechanism remains unclear. By colonizing adult germ-free mice with the cecal contents of neonatal and adult mice, we show that the neonatal microbiota is unable to prevent colonization by two bacterial pathogens that cause mortality in neonates. The lack of colonization resistance occurred when Clostridiales were absent in the neonatal microbiota. Administration of Clostridiales, but not Bacteroidales, protected neonatal mice from pathogen infection and abrogated intestinal pathology upon pathogen challenge. Depletion of Clostridiales also abolished colonization resistance in adult mice. The neonatal bacteria enhanced the ability of protective Clostridiales to colonize the gut.


Asunto(s)
Clostridium/inmunología , Microbioma Gastrointestinal/inmunología , Intestinos/inmunología , Intestinos/microbiología , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Animales Recién Nacidos , Bacteroides/inmunología , Ciego/inmunología , Ciego/microbiología , Vida Libre de Gérmenes , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Factor 88 de Diferenciación Mieloide/genética , Proteínas Asociadas a Pancreatitis/metabolismo
13.
Am J Physiol Gastrointest Liver Physiol ; 312(1): G15-G23, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27810953

RESUMEN

The impact of omeprazole (OM), a widely used over-the-counter proton pump inhibitor, on weight gain has not been extensively explored. We examined what factors, e.g., diet composition, microbiota, genetic strain, and sex, might affect weight gain in mice fed a high caloric diet while on OM. Inbred C57BL/6J strain, a 50:50 hybrid (B6SJLF1/J) strain, and mice on a highly mixed genetic background were fed four diets: standard chow (STD, 6% fat), STD with 200 ppm OM (STD + O), a high-energy chow (HiE, 11% fat), and HiE chow with OM (HiE + O) for 17 wk. Metabolic analysis, body composition, and fecal microbiota composition were analyzed in C57BL/6J mice. Oral glucose tolerance tests were performed using mice on the mixed background. After 8 wk, female and male C57BL/6J mice on the HiE diets ate less, whereas males on the HiE diets compared with the STD diets gained weight. All diet treatments reduced energy expenditure in females but in males only those on the HiE + O diet. Gut microbiota composition differed in the C57BL/6J females but not the males. Hybrid B6SJLF1/J mice showed similar weight gain on all test diets. In contrast, mixed strain male mice fed a HiE + O diet gained ∼40% more weight than females on the same diet. In addition to increased weight gain, mixed genetic mice on the HiE + O diet cleared glucose normally but secreted more insulin. We concluded that sex and genetic background define weight gain and metabolic responses of mice on high caloric diets and OM.


Asunto(s)
Dieta Alta en Grasa , Omeprazol/farmacología , Inhibidores de la Bomba de Protones/farmacología , Aumento de Peso/efectos de los fármacos , Adiposidad/efectos de los fármacos , Adiposidad/genética , Animales , Ingestión de Energía/efectos de los fármacos , Ingestión de Energía/genética , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Conducta Alimentaria/efectos de los fármacos , Conducta Alimentaria/fisiología , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/genética , Antecedentes Genéticos , Masculino , Ratones , Ratones Endogámicos C57BL , Factores Sexuales , Aumento de Peso/genética
14.
Cell Mol Gastroenterol Hepatol ; 2(4): 468-481, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27795980

RESUMEN

BACKGROUND & AIMS: Gut dysbiosis is closely involved in the pathogenesis of inflammatory bowel disease (IBD). However, it remains unclear whether IBD-associated gut dysbiosis contributes to disease pathogenesis or is merely secondary to intestinal inflammation. We established a humanized gnotobiotic (hGB) mouse system to assess the functional role of gut dysbiosis associated with 2 types of IBD: Crohn's disease (CD) and ulcerative colitis (UC). METHODS: Germ-free mice were colonized by the gut microbiota isolated from patients with CD and UC, and healthy controls. Microbiome analysis, bacterial functional gene analysis, luminal metabolome analysis, and host gene expression analysis were performed in hGB mice. Moreover, the colitogenic capacity of IBD-associated microbiota was evaluated by colonizing germ-free colitis-prone interleukin 10-deficient mice with dysbiotic patients' microbiota. RESULTS: Although the microbial composition seen in donor patients' microbiota was not completely reproduced in hGB mice, some dysbiotic features of the CD and UC microbiota (eg, decreased diversity, alteration of bacterial metabolic functions) were recapitulated in hGB mice, suggesting that microbial community alterations, characteristic for IBD, can be reproduced in hGB mice. In addition, colonization by the IBD-associated microbiota induced a proinflammatory gene expression profile in the gut that resembles the immunologic signatures found in CD patients. Furthermore, CD microbiota triggered more severe colitis than healthy control microbiota when colonized in germ-free interleukin 10-deficient mice. CONCLUSIONS: Dysbiosis potentially contributes to the pathogenesis of IBD by augmenting host proinflammatory immune responses. Transcript profiling: GSE73882.

15.
Gut Microbes ; 5(4): 571-5, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25244596

RESUMEN

Rifaximin is a non-systemic, broad-spectrum antibiotic that acts against gram-positive, gram-negative, and anaerobic bacteria. Clinical studies indicate that rifaximin is beneficial in treating irritable bowel syndrome (IBS). The mechanism responsible for the beneficial effects of rifaximin is not clear. In a recent study, we reported that rifaximin alters the bacterial population in the ileum of rats, leading to a relative abundance of Lactobacillus species. These changes prevent gut inflammation and visceral hyperalgesia caused by chronic stress. To more closely mirror human clinical studies in which rifaximin is used to treat IBS symptoms, we performed additional studies and showed that rifaximin reversed mucosal inflammation and barrier dysfunction evoked by chronic stress. These beneficial effects were accompanied by a striking increase in the abundance of Lactobacillaceae and a marked reduction in the number of segmented filamentous bacteria after rifaximin treatment. These microbial changes may contribute to the antiinflammatory effects of rifaximin on the intestinal mucosa.


Asunto(s)
Antibacterianos/administración & dosificación , Antiinflamatorios/administración & dosificación , Bacterias/efectos de los fármacos , Biota/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Rifamicinas/administración & dosificación , Animales , Bacterias/inmunología , Modelos Animales de Enfermedad , Factores Inmunológicos/administración & dosificación , Inflamación/patología , Mucosa Intestinal/patología , Ratas Wistar , Rifaximina , Resultado del Tratamiento
16.
Gastroenterology ; 146(2): 484-96.e4, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24161699

RESUMEN

BACKGROUND & AIMS: Rifaximin is used to treat patients with functional gastrointestinal disorders, but little is known about its therapeutic mechanism. We propose that rifaximin modulates the ileal bacterial community, reduces subclinical inflammation of the intestinal mucosa, and improves gut barrier function to reduce visceral hypersensitivity. METHODS: We induced visceral hyperalgesia in rats, via chronic water avoidance or repeat restraint stressors, and investigated whether rifaximin altered the gut microbiota, prevented intestinal inflammation, and improved gut barrier function. Quantitative polymerase chain reaction (PCR) and 454 pyrosequencing were used to analyze bacterial 16S ribosomal RNA in ileal contents from the rats. Reverse transcription, immunoblot, and histologic analyses were used to evaluate levels of cytokines, the tight junction protein occludin, and mucosal inflammation, respectively. Intestinal permeability and rectal sensitivity were measured. RESULTS: Water avoidance and repeat restraint stress each led to visceral hyperalgesia, accompanied by mucosal inflammation and impaired mucosal barrier function. Oral rifaximin altered the composition of bacterial communities in the ileum (Lactobacillus species became the most abundant) and prevented mucosal inflammation, impairment to intestinal barrier function, and visceral hyperalgesia in response to chronic stress. Neomycin also changed the composition of the ileal bacterial community (Proteobacteria became the most abundant species). Neomycin did not prevent intestinal inflammation or induction of visceral hyperalgesia induced by water avoidance stress. CONCLUSIONS: Rifaximin alters the bacterial population in the ileum of rats, leading to a relative abundance of Lactobacillus. These changes prevent intestinal abnormalities and visceral hyperalgesia in response to chronic psychological stress.


Asunto(s)
Fármacos Gastrointestinales/farmacología , Hiperalgesia/prevención & control , Ileítis/prevención & control , Íleon/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Microbiota/efectos de los fármacos , Rifamicinas/farmacología , Administración Oral , Animales , Biomarcadores/metabolismo , Western Blotting , Citocinas/metabolismo , ADN Bacteriano/análisis , Esquema de Medicación , Fármacos Gastrointestinales/uso terapéutico , Hiperalgesia/etiología , Hiperalgesia/metabolismo , Hiperalgesia/microbiología , Ileítis/etiología , Ileítis/metabolismo , Ileítis/microbiología , Íleon/metabolismo , Íleon/microbiología , Inmunohistoquímica , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Masculino , Microbiota/genética , Neomicina/farmacología , Neomicina/uso terapéutico , Ocludina/metabolismo , Ratas , Ratas Wistar , Restricción Física , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rifamicinas/uso terapéutico , Rifaximina , Análisis de Secuencia de ADN , Estrés Psicológico
17.
J Immunol ; 190(12): 6607-15, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23667106

RESUMEN

Enteral nutrient deprivation via total parenteral nutrition (TPN) administration leads to local mucosal inflammatory responses, but the underlying mechanisms are unknown. Wild-type (WT) and MyD88(-/-) mice underwent jugular vein cannulation. One group received TPN without chow, and controls received standard chow. After 7 d, we harvested intestinal mucosally associated bacteria and isolated small-bowel lamina propria (LP) cells. Bacterial populations were analyzed using 454 pyrosequencing. LP cells were analyzed using quantitative PCR and multicolor flow cytometry. WT, control mucosally associated microbiota were Firmicutes-dominant, whereas WT TPN mice were Proteobacteria-domiant. Similar changes were observed in MyD88(-/-) mice with TPN administration. UniFrac analysis showed divergent small bowel and colonic bacterial communities in controls, merging toward similar microbiota (but distinct from controls) with TPN. The percentage of LP T regulatory cells significantly decreased with TPN in WT mice. F4/80(+)CD11b(+)CD11c(dull/-) macrophage-derived proinflammatory cytokines significantly increased with TPN. These proinflammatory immunologic changes were significantly abrogated in MyD88(-/-) TPN mice. Thus, TPN administration is associated with significant expansion of Proteobacteria within the intestinal microbiota and increased proinflammatory LP cytokines. Additionally, MyD88 signaling blockade abrogated decline in epithelial cell proliferation and epithelial barrier function loss.


Asunto(s)
Inflamación/patología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Factor 88 de Diferenciación Mieloide/inmunología , Nutrición Parenteral Total/efectos adversos , Animales , Citometría de Flujo , Inflamación/etiología , Inflamación/microbiología , Mucosa Intestinal/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Membrana Mucosa/microbiología , Membrana Mucosa/patología , Polimorfismo de Longitud del Fragmento de Restricción , Reacción en Cadena en Tiempo Real de la Polimerasa
18.
Gastroenterology ; 144(7): 1478-87, 1487.e1-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23470617

RESUMEN

BACKGROUND & AIMS: Stress alters brain-gut interactions and could exacerbate intestinal disorders, including irritable bowel syndrome. Alterations in the intestinal microbiota have been associated with irritable bowel syndrome. Maintenance of healthy microbiota requires nucleotide-binding oligomerization domain protein-like receptors, pyrin-domain containing (NLRP)-6 inflammasomes. We investigated the involvement of NLRP6 in water-avoidance stress (WAS)-induced intestinal disorders in mice. METHODS: B57BL6 mice were subjected to WAS for 1 hour each day for 10 days; body weights and intestinal inflammation and permeability were analyzed. We investigated signaling via the NLRP3 and NLRP6 inflammasomes, and the role of corticotropin-releasing hormone (CRH) in WAS-associated inflammation and NLRP6 inhibition. Mice that were not exposed to stress were co-housed with mice subjected to WAS to determine the effects of WAS-induced dysbiosis, measured by sequencing bacterial 16S ribosomal RNA. We also assessed the effects of a peroxisome proliferator-activated receptor-γ agonist and probiotics. RESULTS: WAS-induced small-bowel inflammation (enteritis) was associated with inhibition of NLRP6, but not NLRP3, and was prevented by a peroxisome proliferator-activated receptor-γ agonist, which induced epithelial expression of NLRP6. CRH was released during WAS and inhibited NLRP6 expression. WAS induced alterations in the gut microbiota of mice; co-housed nonstressed mice developed enteritis associated with increased CRH and decreased levels of NLRP6. Probiotic therapy reduced intestinal inflammation in mice with WAS-induced enteritis. CONCLUSIONS: Exposure of mice to stress inhibits NLRP6 and alters the composition of the gut microbiota, leading to intestinal inflammation. These findings might explain the benefits of probiotics for patients with stress-associated gastrointestinal disorders.


Asunto(s)
Hormona Liberadora de Corticotropina/fisiología , Enteritis/etiología , Receptores de Superficie Celular/fisiología , Estrés Psicológico/fisiopatología , Animales , Modelos Animales de Enfermedad , Enteritis/terapia , Femenino , Inflamasomas/metabolismo , Síndrome del Colon Irritable/fisiopatología , Síndrome del Colon Irritable/psicología , Metagenoma/fisiología , Ratones , Ratones Endogámicos C57BL , PPAR gamma/agonistas , Probióticos/uso terapéutico , Estrés Psicológico/complicaciones
19.
Appl Environ Microbiol ; 78(7): 2359-66, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22286988

RESUMEN

Little is known about the dynamics of early ecological succession during experimental conventionalization of the gastrointestinal (GI) tract; thus, we measured changes in bacterial communities over time, at two different mucosal sites (cecum and jejunum), with germfree C57BL/6 mice as the recipients of cecal contents (input community) from a C57BL/6 donor mouse. Bacterial communities were monitored using pyrosequencing of 16S rRNA gene amplicon libraries from the cecum and jejunum and analyzed by a variety of ecological metrics. Bacterial communities, at day 1 postconventionalization, in the cecum and jejunum had lower diversity and were distinct from the input community (dominated by either Escherichia or Bacteroides). However, by days 7 and 21, the recipient communities had become significantly diverse and the cecal communities resembled those of the donor and donor littermates, confirming that transfer of cecal contents results in reassembly of the community in the cecum 7 to 21 days later. However, bacterial communities in the recipient jejunum displayed significant structural heterogeneity compared to each other or the donor inoculum or the donor littermates, suggesting that the bacterial community of the jejunum is more dynamic during the first 21 days of conventionalization. This report demonstrates that (i) mature input communities do not simply reassemble at mucosal sites during conventionalization (they first transform into a "pioneering" community and over time take on the appearance, in membership and structure, of the original input community) and (ii) the specific mucosal environment plays a role in shaping the community.


Asunto(s)
Bacterias/clasificación , Bacterias/crecimiento & desarrollo , Ciego/microbiología , Ecosistema , Vida Libre de Gérmenes , Yeyuno/microbiología , Animales , Bacterias/genética , Bacterias/aislamiento & purificación , Bacteroides/genética , Bacteroides/crecimiento & desarrollo , Bacteroides/aislamiento & purificación , ADN Bacteriano/análisis , ADN Bacteriano/genética , ADN Bacteriano/aislamiento & purificación , ADN Ribosómico/genética , Heces/microbiología , Genes de ARNr , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa , Proteobacteria/genética , Proteobacteria/crecimiento & desarrollo , Proteobacteria/aislamiento & purificación , ARN Ribosómico 16S/genética , Análisis de Secuencia de ADN/métodos
20.
J Parasitol ; 90(3): 663-4, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15270122

RESUMEN

A total of 65 largemouth bass, Micropterus salmoides, and 27 smallmouth bass, M. dolomieu, collected in April-September 2000 and April-July 2001 from Gull Lake, Michigan, were examined for acanthocephalans. Leptorhynchoides thecatus and Neoechinorhynchus cylindratus infected all the bass examined. Leptorhynchoides thecatus had the highest mean intensity (258.2 +/- 185.4 in 2000 and 145.0 +/- 61.0 in 2001) of the species infecting smallmouth bass. Although N. cylindratus had higher mean intensities (42.1 +/- 37.9 in 2000 and 68.9 +/- 70.5 in 2001) than did L. thecatus in largemouth bass, the values were not significantly different between bass species. The prevalence, mean intensity, and mean abundance of Pomphorhynchus bulbocolli in the bass species were below the values for the other acanthocephalan species. Leptorhynchoides thecatus and N. cylindratus are the most abundant intestinal helminths in bass from Gull Lake.


Asunto(s)
Acantocéfalos/aislamiento & purificación , Lubina/parasitología , Enfermedades de los Peces/epidemiología , Helmintiasis Animal/epidemiología , Acantocéfalos/crecimiento & desarrollo , Animales , Femenino , Enfermedades de los Peces/parasitología , Agua Dulce , Helmintiasis Animal/parasitología , Masculino , Michigan/epidemiología , Prevalencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA