Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 168
Filtrar
1.
Food Res Int ; 192: 114801, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39147503

RESUMEN

This study aimed to evaluate the effects of fermented beetroot ketchup enriched with Lactobacillus johnsonii K4 and non-fermented beetroot ketchup on pooled fecal microbiota from healthy adults in in vitro colon model. The research focused on how these products influenced the composition and functionality of the gut microbiota, as well as metabolite production, using the validated dynamic in vitro colon model, TNO Intestinal Model (TIM-2). After an initial starvation phase, a single 60 g dose of predigested and freeze-dried ketchup was introduced into the model. The potential probiotic strain Lactobacillus johnsonii K4 was added over three days. A carbohydrate mixture of standard ileal effluent medium (SIEM) served as the control. Our analysis identified 21 bacterial taxa that were significantly modulated (q-value < 0.2) when comparing ketchup samples to control samples. Notably, the ketchup samples led to an increase in butyrate-producing taxa, including Faecalibacterium, Blautia, Ruminococcaceae, Ruminiclostridium 6, and Anaerostipes. Conversely, there was a reduction in potentially pathogenic genera Desulfovibrio and Escherichia-Shigella. Distinct profiles of short-chain fatty acids (SCFA) were observed among the fermented ketchup, non-fermented ketchup, and control samples. Non-fermented ketchup resulted in higher proportions of acetate, propionate, and butyrate compared to the other interventions. This may be related to the fermentation with lactic acid bacteria in fermented samples with lower levels of substrate for SCFAs production. However, fermented ketchup sample has higher relative abundance of beneficial bacteria like Lactobacillus, Weissella and Dorea in gut microbiota. These findings suggest that beetroot ketchup, can positively influence gut microbiota composition and function, highlighting its potential benefits for human health.


Asunto(s)
Colon , Ácidos Grasos Volátiles , Heces , Fermentación , Microbioma Gastrointestinal , Microbioma Gastrointestinal/fisiología , Colon/microbiología , Colon/metabolismo , Humanos , Heces/microbiología , Ácidos Grasos Volátiles/metabolismo , Probióticos , Beta vulgaris/microbiología , Beta vulgaris/química , Adulto , Lactobacillus/metabolismo , Alimentos Fermentados/microbiología , Bacterias/metabolismo , Bacterias/clasificación , Butiratos/metabolismo , Masculino
2.
Microbiome ; 12(1): 152, 2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-39152482

RESUMEN

BACKGROUND: H2S imbalances in the intestinal tract trigger Crohn's disease (CD), a chronic inflammatory gastrointestinal disorder characterized by microbiota dysbiosis and barrier dysfunction. However, a comprehensive understanding of H2S generation in the gut, and the contributions of both microbiota and host to systemic H2S levels in CD, remain to be elucidated. This investigation aimed to enhance comprehension regarding the sulfidogenic potential of both the human host and the gut microbiota. RESULTS: Our analysis of a treatment-naive CD cohorts' fecal metagenomic and biopsy metatranscriptomic data revealed reduced expression of host endogenous H2S generation genes alongside increased abundance of microbial exogenous H2S production genes in correlation with CD. While prior studies focused on microbial H2S production via dissimilatory sulfite reductases, our metagenomic analysis suggests the assimilatory sulfate reduction (ASR) pathway is a more significant contributor in the human gut, given its high prevalence and abundance. Subsequently, we validated our hypothesis experimentally by generating ASR-deficient E. coli mutants ∆cysJ and ∆cysM through the deletion of sulfite reductase and L-cysteine synthase genes. This alteration significantly affected bacterial sulfidogenic capacity, colon epithelial cell viability, and colonic mucin sulfation, ultimately leading to colitis in murine model. Further study revealed that gut microbiota degrade sulfopolysaccharides and assimilate sulfate to produce H2S via the ASR pathway, highlighting the role of sulfopolysaccharides in colitis and cautioning against their use as food additives. CONCLUSIONS: Our study significantly advances understanding of microbial sulfur metabolism in the human gut, elucidating the complex interplay between diet, gut microbiota, and host sulfur metabolism. We highlight the microbial ASR pathway as an overlooked endogenous H2S producer and a potential therapeutic target for managing CD. Video Abstract.


Asunto(s)
Enfermedad de Crohn , Microbioma Gastrointestinal , Sulfuro de Hidrógeno , Sulfatos , Enfermedad de Crohn/microbiología , Humanos , Sulfuro de Hidrógeno/metabolismo , Animales , Ratones , Sulfatos/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Heces/microbiología , Disbiosis/microbiología , Colon/microbiología , Metagenómica , Oxidación-Reducción , Modelos Animales de Enfermedad , Femenino
3.
Pharmazie ; 79(7): 151-158, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39152558

RESUMEN

Substances metabolised by the intestinal microbiota can be used as colon markers and are gaining importance. The flavonoid glycoside baicalin has been described in the literature to be metabolised by the intestinal microbiota. The aim of this work was to investigate how the biotransformation of baicalin to baicalein is related to the intestinal microbiota. For this purpose, stool samples from healthy volunteers with different dietary habits were used. From the pre-cultured stool samples, different standard microbiota were obtained which were used for the subsequent metabolism studies in the in vitro model MimiCol. MimiCol represents the ascending section of the colon, the colon ascendens, in terms of available volume, pH-value, redox potential and bacterial abundance. While during the experiments with added standard microbiota a metabolism of baicalin to baicalein could be detected, this was not the case in a series of experiments without added microbiota. This confirmed the hypothesis that the metabolism of baicalin relies on the bacterial species that are present in the colon. The data collected in the MimiCol therefore support the use of baicalin as a potential marker for the determination of the colon arrival. This can be explained by the fact that baicalin in its native form is poorly absorbed from the gastrointestinal tract. Enzymes of the colonic microbiota, namely ß-glucuronidases, hydrolyze baicalin to the aglycone baicalein. The resulting aglycone can be absorbed through the intestinal mucosa and detected in blood plasma. This potentially enables the use of baicalin as a marker to determine the time of arrival in the colon.


Asunto(s)
Colon , Heces , Flavanonas , Flavonoides , Microbioma Gastrointestinal , Flavonoides/metabolismo , Flavonoides/farmacocinética , Humanos , Microbioma Gastrointestinal/fisiología , Heces/microbiología , Flavanonas/metabolismo , Colon/metabolismo , Colon/microbiología , Biotransformación , Adulto , Bacterias/metabolismo , Masculino
4.
Gut Microbes ; 16(1): 2392874, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39163515

RESUMEN

Alterations in intestinal permeability and the gut microbiome caused by alcohol abuse are associated with alcoholic liver disease and with worsening of inflammatory bowel diseases (IBD) symptoms. To resolve the direct effects of chronic ethanol consumption on the colon and its microbiome in the absence of acute or chronic alcohol-induced liver disease, we developed a mouse model of chronic binge drinking that uncovers how alcohol may enhance susceptibility to colitis via the microbiota. Employing daily ethanol gavage, we recapitulate key features of binge ethanol consumption. We found that binge ethanol drinking worsens intestinal infection, colonic injury and inflammation, and this effect persists beyond the drinking period. Using gnotobiotics, we showed that alcohol-driven susceptibility to colitis is microbiota-dependent and transferable to ethanol-naïve mice by microbiome transplantation. Allobaculum spp. expanded in binge drinking mice, and administration of Allobaculum fili was sufficient to enhance colitis in non-drinking mice. Our study provides a model to study binge drinking-microbiota interactions and their effects on host disease and reinforces the pathogenic function of Allobaculum spp. as colitogenic bacteria. Our findings illustrate how chronic binge drinking-induced alterations of the microbiome may affect susceptibility to IBD onset or flares.


Asunto(s)
Consumo Excesivo de Bebidas Alcohólicas , Colitis , Colon , Microbioma Gastrointestinal , Ratones Endogámicos C57BL , Animales , Consumo Excesivo de Bebidas Alcohólicas/complicaciones , Microbioma Gastrointestinal/efectos de los fármacos , Ratones , Colitis/microbiología , Colitis/inducido químicamente , Colon/microbiología , Colon/patología , Modelos Animales de Enfermedad , Bacterias/clasificación , Bacterias/aislamiento & purificación , Bacterias/genética , Etanol/efectos adversos , Susceptibilidad a Enfermedades , Masculino , Vida Libre de Gérmenes , Inflamación/microbiología , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/patología
5.
Front Immunol ; 15: 1445838, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39165355

RESUMEN

Objective: Modified Gegen Qinlian Decoction (MGQD) has been shown to effectively relieve ulcerative colitis (UC) without a known pharmacological mechanism. In this study, the anti-colitis efficaciousness of MGQD and its underlying mechanisms in UC were evaluated. Methods: Mice with colitis were administered MGQD for 7 days. Following the evaluation of clinical symptoms, gut microbiota in the feces of UC mice was examined using 16S rRNA sequencing and bile acids (BAs) were examined using LC/MS. Gut microbiota consumption and fecal microbiota transplantation (FMT) were used to explore the involvement of gut microbiota in the anti-UC action of MGQD. Results: MGQD relieved colitis as shown by weight loss protection, a lower disease activity index (DAI), restoration of intestinal length reduction, and lower histopathologic scores. MGQD also restored crypt stem cell proliferation and function of colonic goblet cells, and promoted MUC2 protein secretion. Interestingly, investigations using gut bacterial depletion and FMT showed that MGQD attenuated colonic damage in a gut-dependent way. The modulation of the gut microbiota by MGQD might be attributed to a decrease in Odoribacter and an increase in norank_f_Muribaculaceae. In addition, MGQD modulated the metabolism of BAs while restoring the structure of the gut microbiota. Conclusion: MGQD significantly alleviated colitis in mice, which may be associated with the modulation of gut microbiota and BA metabolism and restoration of function of goblet cells. However, factors other than the gut microbiota may also be involved in the amelioration of UC by MGQD.


Asunto(s)
Ácidos y Sales Biliares , Colitis Ulcerosa , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos , Microbioma Gastrointestinal , Células Caliciformes , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Colitis Ulcerosa/microbiología , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/metabolismo , Colitis Ulcerosa/terapia , Ratones , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Células Caliciformes/efectos de los fármacos , Células Caliciformes/metabolismo , Ácidos y Sales Biliares/metabolismo , Masculino , Trasplante de Microbiota Fecal , Ratones Endogámicos C57BL , Colon/patología , Colon/metabolismo , Colon/efectos de los fármacos , Colon/microbiología
6.
Sci Rep ; 14(1): 18866, 2024 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-39143192

RESUMEN

Isomalto/malto-polysaccharides (IMMPs) are a novel type of soluble dietary fibres with a prebiotic potential promoting growth of beneficial microbes in the gut. However, the mode of action of IMMPs remains unknown. Previous studies on IMMPs showed an increase in total bacteria, especially lactobacilli, and higher production of short chain fatty acids (SCFA) when IMMPs were fed to rats or used during in vitro fermentation. Here we used metatranscriptomics to investigate how IMMPs with different amounts of α - (1 → 6) glycosidic linkages affected microbial function during incubation with human fecal inoculum. We showed that active microbial community dynamics during fermentation varied depending on the type of IMMP used and that the observed changes were reflected in the community gene expression profiles. Based on metatranscriptome analysis, members of Bacteroides, Lactobacillus and Bifidobacterium were the predominant degraders of IMMPs, and the increased gene expression in these bacteria correlated with high amounts of α - (1 → 6) glycosidic linkages. We also noted an increase in relative abundance of these bacteria and an activation of pathways involved in SCFA synthesis. Our findings could provide a baseline for more targeted approaches in designing prebiotics for specific bacteria and to achieve more controlled modulation of microbial activity towards desired health outcomes.


Asunto(s)
Heces , Microbioma Gastrointestinal , Prebióticos , Humanos , Microbioma Gastrointestinal/efectos de los fármacos , Heces/microbiología , Fermentación , Polisacáridos/metabolismo , Colon/microbiología , Colon/metabolismo , Ácidos Grasos Volátiles/metabolismo , Transcriptoma , Perfilación de la Expresión Génica , Fibras de la Dieta/metabolismo , Lactobacillus/metabolismo , Lactobacillus/genética , Bacterias/genética , Bacterias/metabolismo
7.
Microbiome ; 12(1): 147, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39113097

RESUMEN

BACKGROUND: Understanding the cause vs consequence relationship of gut inflammation and microbial dysbiosis in inflammatory bowel diseases (IBD) requires a reproducible mouse model of human-microbiota-driven experimental colitis. RESULTS: Our study demonstrated that human fecal microbiota transplant (FMT) transfer efficiency is an underappreciated source of experimental variability in human microbiota-associated (HMA) mice. Pooled human IBD patient fecal microbiota engrafted germ-free (GF) mice with low amplicon sequence variant (ASV)-level transfer efficiency, resulting in high recipient-to-recipient variation of microbiota composition and colitis severity in HMA Il-10-/- mice. In contrast, mouse-to-mouse transfer of mouse-adapted human IBD patient microbiota transferred with high efficiency and low compositional variability resulting in highly consistent and reproducible colitis phenotypes in recipient Il-10-/- mice. Engraftment of human-to-mouse FMT stochastically varied with individual transplantation events more than mouse-adapted FMT. Human-to-mouse FMT caused a population bottleneck with reassembly of microbiota composition that was host inflammatory environment specific. Mouse-adaptation in the inflamed Il-10-/- host reassembled a more aggressive microbiota that induced more severe colitis in serial transplant to Il-10-/- mice than the distinct microbiota reassembled in non-inflamed WT hosts. CONCLUSIONS: Our findings support a model of IBD pathogenesis in which host inflammation promotes aggressive resident bacteria, which further drives a feed-forward process of dysbiosis exacerbated by gut inflammation. This model implies that effective management of IBD requires treating both the dysregulated host immune response and aggressive inflammation-driven microbiota. We propose that our mouse-adapted human microbiota model is an optimized, reproducible, and rigorous system to study human microbiome-driven disease phenotypes, which may be generalized to mouse models of other human microbiota-modulated diseases, including metabolic syndrome/obesity, diabetes, autoimmune diseases, and cancer. Video Abstract.


Asunto(s)
Modelos Animales de Enfermedad , Disbiosis , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Interleucina-10 , Animales , Humanos , Ratones , Enfermedades Inflamatorias del Intestino/microbiología , Disbiosis/microbiología , Interleucina-10/genética , Colitis/microbiología , Heces/microbiología , Colon/microbiología , Ratones Noqueados , Ratones Endogámicos C57BL , Femenino , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Inflamación , Masculino
8.
Gut Microbes ; 16(1): 2387877, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39133871

RESUMEN

Colibactin is a recently characterized pro-carcinogenic genotoxin produced by pks+ Escherichia coli. We hypothesized that cystic fibrosis (CF)-associated dysfunctional mucus structure increases the vulnerability of host mucosa to colibactin-induced DNA damage. In this pilot study, we tested healthy-appearing mucosal biopsy samples obtained during screening and surveillance colonoscopies of adult CF and non-CF patients for the presence of pks+ E. coli, and we investigated the possibility of detecting a novel colibactin-specific DNA adduct that has not been yet been demonstrated in humans. While CF patients had a lower incidence of pks+ E. coli carriage (~8% vs 29%, p = 0.0015), colibactin-induced DNA adduct formation was detected, but only in CF patients and only in those who were not taking CFTR modulator medications. Moreover, the only patient found to have colon cancer during this study had CF, harbored pks+ E. coli, and had colibactin-induced DNA adducts in the mucosal samples. Larger studies with longitudinal follow-up should be done to extend these initial results and further support the development of colibactin-derived DNA adducts to stratify patients and their risk.


Asunto(s)
Colon , Fibrosis Quística , Aductos de ADN , Escherichia coli , Mucosa Intestinal , Moco , Péptidos , Policétidos , Fibrosis Quística/microbiología , Fibrosis Quística/metabolismo , Humanos , Policétidos/metabolismo , Aductos de ADN/metabolismo , Adulto , Escherichia coli/genética , Escherichia coli/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Péptidos/metabolismo , Masculino , Colon/microbiología , Colon/patología , Colon/metabolismo , Femenino , Proyectos Piloto , Moco/metabolismo , Moco/microbiología , Persona de Mediana Edad , Adulto Joven , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología
9.
Nutrients ; 16(15)2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39125398

RESUMEN

Persimmon fruit processing-derived waste and by-products, such as peels and pomace, are important sources of dietary fiber and phytochemicals. Revalorizing these by-products could help promote circular nutrition and agricultural sustainability while tackling dietary deficiencies and chronic diseases. In this study, fiber-rich fractions were prepared from the by-products of Sharoni and Brilliant Red persimmon varieties. These fractions were quantified for their phenolic composition and assessed for their ability to promote the growth of beneficial human colonic Firmicutes species and for their in vitro anti-inflammatory potential. Gallic and protocatechuic acids, delphinidin, and cyanidin were the main phenolics identified. Faecalibacterium prausnitzii strains showed significantly higher growth rates in the presence of the Brilliant Red fraction, generating more than double butyrate as a proportion of the total short-chain fatty acids (39.5% vs. 17.8%) when compared to glucose. The fiber-rich fractions significantly decreased the inflammatory effect of interleukin-1ß in Caco-2 cells, and the fermented fractions (both from Sharoni and Brilliant Red) significantly decreased the inflammatory effect of interleukin-6 and tumor necrosis factor-α in the RAW 264.7 cells. Therefore, fiber-rich fractions from persimmon by-products could be part of nutritional therapies as they reduce systemic inflammation, promote the growth of beneficial human gut bacteria, and increase the production of beneficial microbial metabolites such as butyrate.


Asunto(s)
Antiinflamatorios , Colon , Fibras de la Dieta , Diospyros , Humanos , Fibras de la Dieta/farmacología , Fibras de la Dieta/análisis , Diospyros/química , Ratones , Antiinflamatorios/farmacología , Colon/microbiología , Colon/efectos de los fármacos , Colon/metabolismo , Animales , Células RAW 264.7 , Células CACO-2 , Microbioma Gastrointestinal/efectos de los fármacos , Firmicutes , Faecalibacterium prausnitzii , Frutas/química , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Hidroxibenzoatos/farmacología , Hidroxibenzoatos/análisis , Fenoles/farmacología , Fenoles/análisis , Fermentación , Ácido Gálico/farmacología , Antocianinas/farmacología , Antocianinas/análisis
10.
J Appl Microbiol ; 135(7)2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38986506

RESUMEN

AIMS: This study aimed to compare the effects of linear and branched fructooligosaccharides (FOS) extracted from chicory and grass (Lolium perenne), respectively on human microbiota composition, diversity, and metabolism. METHODS AND RESULTS: To test the effects of linear and branched FOS on human microbiota we used the artificial in vitro human colon model (TIM-2). Microbiota composition and diversity were assessed by V3-V4 16S rRNA metagenomic sequencing, followed by differential taxa abundance and alpha/beta diversity analyses. SCFA/BCFA production was evaluated by gas chromatography-mass spectrometry. As a result, branched FOS had the most beneficial effects on microbial diversity and metabolite production. Also, branched FOS significantly increased the abundance of commensal bacteria associated with maintaining healthy gut functions and controlling inflammation, such as Butyricicoccus, Erysipelotrichaceae, Phascolarctobacterium, and Sutterella. Linear FOS also significantly increased the abundance of some other commensal gut bacteria (Anaerobutyricum, Lachnospiraceae, Faecalibacterium), but there were no differences in diversity metrics compared to the control. CONCLUSIONS: The study revealed that branched FOS had the most beneficial effects compared to the linear FOS in vitro, concerning microbiota modulation, and metabolite production, making this a good candidate for further studies in food biotechnology.


Asunto(s)
Bacterias , Colon , Microbioma Gastrointestinal , Oligosacáridos , ARN Ribosómico 16S , Microbioma Gastrointestinal/efectos de los fármacos , Oligosacáridos/farmacología , Oligosacáridos/metabolismo , Humanos , Bacterias/genética , Bacterias/clasificación , Bacterias/metabolismo , Bacterias/aislamiento & purificación , Bacterias/efectos de los fármacos , Colon/microbiología , Colon/metabolismo , ARN Ribosómico 16S/genética , Lolium/microbiología , Cichorium intybus , Heces/microbiología
11.
J Anim Sci ; 1022024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38995038

RESUMEN

Fermentation of dietary and endogenous protein in the hindgut is generally considered detrimental to the health of pigs. We investigated the in vitro fermentation potential of porcine endogenous protein in ileal digesta and colonic mucus, using a N-free buffer with an excess of fermentable carbohydrates. Urea, whey protein isolate (WPI, positive control), WPI hydrolysate (WPIH), and combinations of the latter two were used to validate the assay. A new biphasic model, including a linear end simulation, fitted to the gas production data over a 48-h period identified the time point when substrate fermentation ended. A higher degree of hydrolysis of WPI resulted in a higher maximum gas production rate (Rmax, P < 0.01). Differences in Rmax and the time required to reach Rmax were observed among ileal digesta samples, with Rmax increasing with the insoluble protein content, and the highest Rmax occurring with colonic mucus samples (P < 0.05). The endogenous proteins entering the large intestine of pigs can ferment more rapidly compared to highly soluble and digestible protein sources, with Rmax positively correlated with decreasing solubility of endogenous nitrogenous components.


Protein fermentation in the hindgut of pigs can impact their health, affecting factors like growth rates and feed efficiency. Besides dietary protein, up to 50% of the protein entering the large intestine of growing pigs may be of endogenous origin. Therefore, we explored the fermentation potential of endogenous proteins compared to a well-known protein source, whey protein isolate (WPI). In developing and validating an in vitro gas production technique, we employed urea, WPI, WPI hydrolysate, and various combinations as substrates. The study introduces a new biphasic model for in vitro gas production, offering a detailed analysis of the fermentation process over a 48-h period. Our results revealed that porcine endogenous proteins can undergo rapid fermentation because the maximum gas production rate was higher compared to WPI. This insight is crucial for understanding the dynamics of protein fermentation in pigs. Additionally, we explored the solubility and molecular size of proteins, providing a comprehensive understanding of their fermentation characteristics. We found that endogenous proteins were less soluble compared to WPI but contained more smaller peptides. Unraveling the complexities of protein fermentation in pigs contributes to improvement of feed formulation for optimal gut health.


Asunto(s)
Proteínas en la Dieta , Fermentación , Animales , Porcinos , Proteínas en la Dieta/metabolismo , Digestión/fisiología , Íleon/metabolismo , Colon/metabolismo , Colon/microbiología , Proteína de Suero de Leche/metabolismo , Contenido Digestivo/química
12.
Sci Rep ; 14(1): 15335, 2024 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-38961176

RESUMEN

Anastomotic leakage (AL) is a potentially life-threatening complication following colorectal cancer (CRC) resection. In this study, we aimed to unravel longitudinal changes in microbial structure before, during, and after surgery and to determine if microbial alterations may be predictive for risk assessment between sufficient anastomotic healing (AS) and AL prior surgery. We analysed the microbiota of 134 colon mucosal biopsies with 16S rRNA V1-V2 gene sequencing. Samples were collected from three location sites before, during, and after surgery, and patients received antibiotics after the initial collection and during surgery. The microbial structure showed dynamic surgery-related changes at different time points. Overall bacterial diversity and the abundance of some genera such as Faecalibacterium or Alistipes decreased over time, while the genera Enterococcus and Escherichia_Shigella increased. The distribution of taxa between AS and AL revealed significant differences in the abundance of genera such as Prevotella, Faecalibacterium and Phocaeicola. In addition to Phocaeicola, Ruminococcus2 and Blautia showed significant differences in abundance between preoperative sample types. ROC analysis of the predictive value of these genera for AL revealed an AUC of 0.802 (p = 0.0013). In summary, microbial composition was associated with postoperative outcomes, and the abundance of certain genera may be predictive of postoperative complications.


Asunto(s)
Fuga Anastomótica , Microbioma Gastrointestinal , Humanos , Masculino , Femenino , Anciano , Fuga Anastomótica/etiología , Fuga Anastomótica/microbiología , Persona de Mediana Edad , Microbioma Gastrointestinal/genética , Neoplasias Colorrectales/cirugía , Neoplasias Colorrectales/microbiología , ARN Ribosómico 16S/genética , Cirugía Colorrectal/efectos adversos , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Colon/microbiología , Colon/cirugía , Colon/patología , Prueba de Estudio Conceptual
13.
J Pharm Biomed Anal ; 249: 116351, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39018720

RESUMEN

Ulcerative colitis (UC) is a chronic inflammatory bowel disease (IBD) that primarily affects mucosa and submucosa of colon and rectum. Although the exact etiology of UC remains elusive, increasing evidence has demonstrated that the gut microbiome and its interaction with host metabolism plays an important role in UC development. The objective of this study was to investigate the therapeutic potential and mechanism of dimeric proanthocyanidins (PAC) enriched from ethyl acetate extract of Ephedra roots on UC from the perspective of gut microbiota and metabolic regulation. In this study, a bio-guided strategy integrating LC-MS analysis, DMAC assay, antioxidant screening, and antiinflammation activity screening was used to enrich dimeric PAC from Ephedra roots, then untargeted metabolomics combined with gut microbiota analysis was performed to investigate the therapeutic mechanism of PRE on UC. This is the first study that combines a bio-guided strategy to enrich dimeric PAC from Ephedra roots and a comprehensive analysis of their effects on gut microbiota and host metabolism. Oral administration of PRE was found to significantly relieve dextran sodium sulfate (DSS)-induced ulcerative colitis symptoms in mice, characterized by the reduced disease activity index (DAI), increased colon length and improved colon pathological damage, together with the down-regulation of colonic inflammatory and oxidative stress levels. In addition, 16 S rRNA sequencing combined with untargeted metabolomics was conducted to reveal the effects of PRE on gut microbiota composition and serum metabolites. PRE improved gut microbiota dysbiosis through increasing the relative abundance of beneficial bacteria Lachnospiraceae_NK4A136_group and decreasing the level of potentially pathogenic bacteria such as Escherichia-Shigella. Serum metabolomics showed that the disturbed tryptophan and glycerophospholipid metabolism in UC mice was restored after PRE treatment. Collectively, PRE was proved to be a promising anti-UC candidate, which deserves further investigation in future research.


Asunto(s)
Colitis Ulcerosa , Sulfato de Dextran , Ephedra sinica , Microbioma Gastrointestinal , Metabolómica , Raíces de Plantas , Proantocianidinas , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/fisiología , Animales , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/microbiología , Colitis Ulcerosa/metabolismo , Ratones , Metabolómica/métodos , Proantocianidinas/farmacología , Proantocianidinas/aislamiento & purificación , Ephedra sinica/química , Masculino , Extractos Vegetales/farmacología , Modelos Animales de Enfermedad , Colon/efectos de los fármacos , Colon/metabolismo , Colon/microbiología , Ratones Endogámicos C57BL
14.
Microbiol Res ; 286: 127812, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38954992

RESUMEN

Clostridioides difficile infection (CDI) poses a significant health threat due to high recurrence rates. Antimicrobial agents are commonly used to manage CDI-related diarrhoea; however, by aggravating intestinal dysbiosis, antibiotics enable C. difficile spores germination and production of toxins, the main virulence factors. Therefore, the binding of exotoxins using adsorbents represents an attractive alternative medication for the prevention and treatment of relapses. In this study, we provided evidence that the natural insoluble polysaccharides, named ABR119, extracted by plant cell cultures, effectively trap C. difficile toxins. In our experiments, ABR119 exhibited no cytotoxicity in vitro and was safely administered in vivo. In the animal model of C. difficile-associated colitis, ABR119 (50 mg/kg body weight) significantly reduced the colonic myeloperoxidase activity and severity of inflammation, preventing body weight loss. These effects were not evident when we treated animals with wheat bran polysaccharides. We did not detect bacterial killing effects of ABR119 against C. difficile nor against bacterial species of the normal gut microbiota. Moreover, ABR119 did not interfere in vitro with the antimicrobial activities of most clinically used antibiotics. In summary, ABR119 holds promise for treating and preventing C. difficile colitis by trapping the bacterial toxins, warranting further studies to assess the ABR119 potential in human infections caused by C. difficile.


Asunto(s)
Antibacterianos , Toxinas Bacterianas , Clostridioides difficile , Infecciones por Clostridium , Colitis , Modelos Animales de Enfermedad , Polisacáridos , Polisacáridos/farmacología , Polisacáridos/metabolismo , Clostridioides difficile/efectos de los fármacos , Clostridioides difficile/patogenicidad , Animales , Colitis/microbiología , Colitis/tratamiento farmacológico , Colitis/prevención & control , Colitis/inducido químicamente , Infecciones por Clostridium/prevención & control , Infecciones por Clostridium/microbiología , Infecciones por Clostridium/tratamiento farmacológico , Toxinas Bacterianas/metabolismo , Antibacterianos/farmacología , Humanos , Células Vegetales , Ratones , Colon/microbiología , Colon/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos
15.
Lab Chip ; 24(15): 3690-3703, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38973701

RESUMEN

Changes in the abundance of certain bacterial species within the colorectal microbiota correlate with colorectal cancer (CRC) development. While carcinogenic mechanisms of single pathogenic bacteria have been characterized in vitro, limited tools are available to investigate interactions between pathogenic bacteria and both commensal microbiota and colonocytes in a physiologically relevant tumor microenvironment. To address this, we developed a microfluidic device that can be used to co-culture colonocyte spheroids and colorectal microbiota. The device was used to explore the effect of Fusobacterium nucleatum, an opportunistic pathogen associated with colorectal cancer development in humans, on colonocyte gene expression and microbiota composition. F. nucleatum altered the transcription of genes involved in cytokine production, epithelial-to-mesenchymal transition, and proliferation in colonocytes in a contact-independent manner; however, most of these effects were significantly diminished by the presence of commensal microbiota. Interestingly, F. nucleatum significantly altered the abundance of multiple bacterial clades associated with mucosal immune responses and cancer development in the colon. Our results highlight the importance of evaluating the potential carcinogenic activity of pathogens in the context of a commensal microbiota, and the potential to discover novel inter-species microbial interactions in the CRC microenvironment.


Asunto(s)
Técnicas de Cocultivo , Colon , Neoplasias Colorrectales , Fusobacterium nucleatum , Humanos , Técnicas de Cocultivo/instrumentación , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/metabolismo , Colon/microbiología , Colon/patología , Dispositivos Laboratorio en un Chip , Técnicas Analíticas Microfluídicas/instrumentación , Transición Epitelial-Mesenquimal , Microbiota , Proliferación Celular
16.
J Agric Food Chem ; 72(29): 16221-16236, 2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-38996349

RESUMEN

A hundred million tons of young apples are thinned and discarded in the orchard per year, aiming to increase the yield and quality of apples. We fermented thinned young apples using a potential probiotic fungus, Eurotium cristatum, which notably disrupted the microstructure of raw samples, as characterized by the scanning electron microscope. Fermentation substantially altered the metabolite profiles of samples, which are predicted to alleviate colitis via regulating inflammatory response and response to lipopolysaccharide by using network pharmacology analysis. In vivo, oral gavage of water extracts of E. cristatum fermented young apples (E.YAP) effectively alleviated DSS-induced colitis, restored the histopathology damage, reduced the levels of inflammatory cytokines, and promoted colonic expressions of tight junction proteins. Moreover, E.YAP ameliorated gut dysbacteriosis by increasing abundances of Lactobacillus,Blautia, Muribaculaceae, and Prevotellaceae_UCG-001 while inhibiting Turicibacter, Alistipes, and Desulfovibrio. Importantly, E.YAP increased colonic bile acids, such as CA, TCA, DCA, TUDCA, and LCA, thereby alleviating colitis via PXR/NF-κB signaling. Furthermore, a synbiotic combination with Limosilactobacillus reuteri WX-94, a probiotic strain isolated from feces of healthy individuals with anti-inflammatory properties, augmented anticolitis capacities of E.YAP. Our findings demonstrate that E.YAP could be a novel, potent, food-based anti-inflammatory prebiotic for relieving inflammatory injuries.


Asunto(s)
Bacterias , Colitis , Eurotium , Fermentación , Malus , Ratones Endogámicos C57BL , Animales , Malus/química , Ratones , Colitis/microbiología , Colitis/metabolismo , Colitis/inducido químicamente , Humanos , Masculino , Eurotium/metabolismo , Eurotium/química , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Bacterias/metabolismo , Microbioma Gastrointestinal/efectos de los fármacos , Probióticos/administración & dosificación , Probióticos/farmacología , Frutas/química , Frutas/microbiología , Colon/microbiología , Colon/metabolismo , Colon/inmunología
17.
Food Funct ; 15(16): 8370-8385, 2024 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-39023128

RESUMEN

Miao sour soup (MSS), a daily fermented food in Guizhou, China, is rich in microorganisms with various beneficial activities, including anti-inflammatory and antioxidant activities. However, the therapeutic effects of MSS on IBD remain unexplored. This study aimed to investigate the protective effect of MSS against colitis in mice. In this study, we examined the microbial community structure of MSS by metagenomic sequencing and also explored the protective effect of MSS on DSS-induced colitis in mice. We investigated the effects of MSS on intestinal inflammatory response and intestinal barrier function in mice. Finally, the changes in intestinal flora were analyzed based on the 16S rRNA gene sequencing results. Significantly, the experiment result shows that MSS ameliorated the severity of DSS-induced disease in mice by mitigating colitis-associated weight loss, reducing the disease activity index of IBD, alleviating colonic hemorrhagic lesions, increasing colon length, and improving colonic tissue damage. Moreover, MSS preserved intestinal barrier integrity and restored intestinal epithelial function in mice. Additionally, MSS modulated the structure and composition of the intestinal flora. Furthermore, MSS downregulated pro-inflammatory factors and attenuated the NF-κB p65 expression, thereby mitigating the inflammatory response. These findings highlight the protective effect of MSS against DSS-induced colitis, providing substantial scientific support for potential applications of MSS as a functional food.


Asunto(s)
Colitis , Sulfato de Dextran , Microbioma Gastrointestinal , Mucosa Intestinal , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Ratones , Colitis/inducido químicamente , Sulfato de Dextran/efectos adversos , Masculino , Mucosa Intestinal/microbiología , Ratones Endogámicos C57BL , Alimentos Fermentados , Modelos Animales de Enfermedad , Colon/microbiología , Colon/metabolismo , ARN Ribosómico 16S/genética , Bacterias/clasificación , Bacterias/aislamiento & purificación , Bacterias/genética , Funcion de la Barrera Intestinal
18.
Food Funct ; 15(16): 8496-8509, 2024 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-39056151

RESUMEN

Egg white protein ovomucin and its hydrolysates were previously reported to exhibit anti-inflammatory and anti-adhesive activities. However, their potential to regulate pathogen colonization and disease severity has not been fully characterized. To investigate the effects of ovomucin (OVM) and its hydrolysates including ovomucin-Protex 26L (OP) and -pepsin/pancreatin (OPP) on host resistance to pathogen infection, a well-documented colitis model in mice for attaching and effacing E. coli pathogens, Citrobacter rodentium, was used in the current study. C57Bl/6J female mice were fed on a basal diet supplemented with OVM or its hydrolysates for 3 weeks prior to the C. rodentium challenge, with the dietary treatments continued for seven days. Body weight was not affected throughout the experimental period. OP supplementation resulted in lower (P < 0.05) pathogen loads at 7 dpi. Attenuated colitis severity was observed in mice that received OVM and OP, as indicated by reduced colonic pathological scores and pro-inflammatory responses compared with the infected control group. In contrast, OPP consumption resulted in enhanced C. rodentium colonization and disease severity. Notably, reduced microbial diversity indices of the gut microbiota were observed in the OPP-supplemented mice compared with the OVM- and OP-supplemented groups. This study showed the potential of OVM and OP to alleviate the severity of colitis induced by infection while also suggesting the opposite outcome of OPP in mitigating enteric infection.


Asunto(s)
Citrobacter rodentium , Colitis , Infecciones por Enterobacteriaceae , Ratones Endogámicos C57BL , Ovomucina , Animales , Ratones , Femenino , Colitis/inducido químicamente , Colitis/microbiología , Infecciones por Enterobacteriaceae/microbiología , Microbioma Gastrointestinal , Modelos Animales de Enfermedad , Colon/microbiología , Colon/patología , Colon/metabolismo , Hidrolisados de Proteína/farmacología
19.
J Pharm Biomed Anal ; 249: 116367, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39029356

RESUMEN

Angelica sinensis (Oliv.) Diels (AS) is a commonly used herbal medicine and culinary spice known for its gastrointestinal protective properties. Angelica sinensis oil (AO) is the main bioactive component of AS. However, the therapeutic effects and mechanisms of AO on the gastrointestinal tract remain unclear. In this study, we aim to investigated the potential of AO in restoring gut microbiota disorder and metabolic disruptions associated with ulcerative colitis (UC). A systematic chemical characterization of AO was conducted using GC×GC-Q TOF-MS. A UC mouse model was established by freely drinking DSS to assess the efficacy of AO. Utilizing 16 S rRNA sequencing in combination with untargeted metabolomics analysis of serum, we identified alterations in gut microbiota, differential metabolites, and pathways influenced by AO in UC treatment, thereby elucidating the therapeutic mechanism of AO in UC management. Pharmacodynamic results indicated that AO effectively inhibited the content of inflammation mediators, such as Interleukin-1ß, Interleukin-6 and tumor necrosis factor-α, and proserved colon tissue integrity in UC mice. Furthermore, AO significantly downregulated the abundance of pathogenic bacteria (Bacteroidetes, Proteobacteria, and Desulfobacteriaceae) while increasing the abundance of beneficial bacteria (Firmicutes, Blautia, Akkermansia, and Lachnospiraceae). Metabolomics analysis highlighted significant disruptions in endogenous metabolism in UC mice, with a notable restoration of SphK1 and S1P levels following AO administration. Besides, we discovered that AO regulated the balance of sphingolipid metabolism and protected the intestinal barrier, potentially through the SphK1/MAPK signaling pathway. Overall, this study indicated that AO effectively ameliorates the clinical manifestations of UC by synergistically regulating gut microbe and metabolite homeostasis. AO emerges as a potential functional and therapeutic ingredient for UC treatment.


Asunto(s)
Angelica sinensis , Colitis Ulcerosa , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Metabolómica , ARN Ribosómico 16S , Animales , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/microbiología , Colitis Ulcerosa/metabolismo , Ratones , Metabolómica/métodos , Microbioma Gastrointestinal/efectos de los fármacos , Angelica sinensis/química , ARN Ribosómico 16S/genética , Masculino , Aceites de Plantas/farmacología , Ratones Endogámicos C57BL , Sulfato de Dextran , Colon/efectos de los fármacos , Colon/metabolismo , Colon/microbiología
20.
Phytomedicine ; 132: 155897, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39032279

RESUMEN

BACKGROUND: Intestinal health is affected by heredity, lifestyle, and structure of gut microbiota. The imbalance of symbiotic and harmful bacteria in gut microbiota may increase the occurrence of colonic inflammation. Supplementary A. muciniphila can improve the survival rate of colitis mice, reduce colon tissue injury, and the expression of anti-inflammatory factors was upregulated. Artemisia argyi has been reported to have anti-inflammatory, antioxidant, bactericidal, and immunomodulatory effects. However, its anti-inflammatory effect and mechanism, and its influence on gut microbiota and metabolites are still unclear yet. PURPOSE: To explore whether Artemisia argyi Polyphenols(AAPs) can alleviate ulcerative colitis (UC) by changing gut microbiota. METHODS: The therapeutic effect of AAPs on colitis was investigated by inducing ulcerative colitis in mice using dextran sodium sulfate (DSS) and administering different doses of AAPs orally to mice. Exploring the levels of inflammatory proteins, oxidative stress proteins, and barrier proteins using western blotting and immunofluorescence, and explored the structural changes of gut microbiota and its metabolites. Meanwhile, in order to explore whether the role of AAPs in alleviating colitis is based on the regulation of gut microbiota structure, we conducted fecal microbiota transplantation (FMT). RESULTS: It showed that AAPs and FMT trial alleviated DSS-induced colonic injury, including clinical parameters and pathological injury of colon tissue, reduction in the expression of inflammatory proteins: IL-6, TNF-α, p-p65, p-IκBα, and increase in the expression of antioxidant proteins: Nrf2, NQO-1 and HO-1 and barrier proteins: Claudin-1, Occludin, ZO-1 and MUC2. AAPs and FMT promoted the content of beneficial bacteria, such as Butyricimonas and Lactobacillus, and the content of beneficial metabolites for instance acetic acid, butyric acid, and valeric acid has also increased. CONCLUSION: These results suggested that AAPs might improve DSS-induced colonic injury by changing the structural of gut microbiota while promoting the synthesis of fatty acids in the intestine, thereby providing a theoretical basis for using AAPs to treat ulcerative colitis.


Asunto(s)
Artemisia , Colitis Ulcerosa , Sulfato de Dextran , Microbioma Gastrointestinal , Polifenoles , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Polifenoles/farmacología , Artemisia/química , Ratones , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/tratamiento farmacológico , Masculino , Ratones Endogámicos C57BL , Colon/efectos de los fármacos , Colon/microbiología , Antiinflamatorios/farmacología , Modelos Animales de Enfermedad , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/farmacología , Extractos Vegetales/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA