Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.133
Filtrar
1.
Gut Microbes ; 16(1): 2394249, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39224018

RESUMEN

Inflammatory bowel diseases (IBD) etiology is multifactorial. Luminal microRNAs (miRNAs) have been suspected to play a role in the promotion of chronic inflammation, but the extent to which fecal miRNAs are interacting with the intestinal ecosystem in a way that contribute to diseases, including IBD, remains unknown. Here, fecal let-7b and miR-21 were found elevated, associated with inflammation, and correlating with multiple bacteria in IBD patients and IL-10-/- mice, model of spontaneous colitis. Using an in vitro microbiota modeling system, we revealed that these two miRNAs can directly modify the composition and function of complex human microbiota, increasing their proinflammatory potential. In vivo investigations revealed that luminal increase of let-7b drastically alters the intestinal microbiota and enhances macrophages' associated proinflammatory cytokines (TNF, IL-6, and IL-1ß). Such proinflammatory effects are resilient and dependent on the bacterial presence. Moreover, we identified that besides impairing the intestinal barrier function, miR-21 increases myeloperoxidase and antimicrobial peptides secretion, causing intestinal dysbiosis. More importantly, in vivo inhibition of let-7b and miR-21 with anti-miRNAs significantly improved the intestinal mucosal barrier function and promoted a healthier host-microbiota interaction in the intestinal lining, which altogether conferred protection against colitis. In summary, we provide evidence of the functional significance of fecal miRNAs in host-microbiota communication, highlighting their therapeutic potential in intestinal inflammation and dysbiosis-related conditions, such as IBD.


Asunto(s)
Colitis , Heces , Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , MicroARNs , MicroARNs/genética , MicroARNs/metabolismo , Animales , Humanos , Heces/microbiología , Ratones , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/metabolismo , Colitis/microbiología , Colitis/inducido químicamente , Colitis/genética , Inflamación/microbiología , Inflamación/metabolismo , Disbiosis/microbiología , Ratones Endogámicos C57BL , Femenino , Ratones Noqueados , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Masculino , Mucosa Intestinal/microbiología , Mucosa Intestinal/metabolismo , Citocinas/metabolismo , Macrófagos/inmunología , Macrófagos/microbiología , Macrófagos/metabolismo , Modelos Animales de Enfermedad , Interleucina-10/genética , Interleucina-10/metabolismo
2.
Microbiome ; 12(1): 177, 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-39300532

RESUMEN

BACKGROUND: Intake of dietary fiber is associated with a reduced risk of inflammatory bowel disease. ß-Glucan (BG), a bioactive dietary fiber, has potential health-promoting effects on intestinal functions; however, the underlying mechanism remains unclear. Here, we explore the role of BG in ameliorating colitis by modulating key bacteria and metabolites, confirmed by multiple validation experiments and loss-of-function studies, and reveal a novel bacterial cross-feeding interaction. RESULTS: BG intervention ameliorates colitis and reverses Lactobacillus reduction in colitic mice, and Lactobacillus abundance was significantly negatively correlated with the severity of colitis. It was confirmed by further studies that Lactobacillus johnsonii was the most significantly enriched Lactobacillus spp. Multi-omics analysis revealed that L. johnsonii produced abundant indole-3-lactic acid (ILA) leading to the activation of aryl hydrocarbon receptor (AhR) responsible for the mitigation of colitis. Interestingly, L. johnsonii cannot utilize BG but requires a cross-feeding with Bacteroides uniformis, which degrades BG and produces nicotinamide (NAM) to promote the growth of L. johnsonii. A proof-of-concept study confirmed that BG increases L. johnsonii and B. uniformis abundance and ILA levels in healthy individuals. CONCLUSIONS: These findings demonstrate the mechanism by which BG ameliorates colitis via L. johnsonii-ILA-AhR axis and reveal the important cross-feeding interaction between L. johnsonii and B. uniformis. Video Abstract.


Asunto(s)
Bacteroides , Colitis , Indoles , Lactobacillus johnsonii , beta-Glucanos , Animales , Indoles/metabolismo , Ratones , Colitis/microbiología , Colitis/terapia , beta-Glucanos/metabolismo , Bacteroides/metabolismo , Humanos , Lactobacillus johnsonii/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Ratones Endogámicos C57BL , Masculino , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Femenino , Lactobacillus/metabolismo
3.
Sci Adv ; 10(38): eado0016, 2024 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-39303027

RESUMEN

Fusobacterium nucleatum in colorectal cancer (CRC) tissue is implicated at multiple stages of the disease, while the mechanisms underlying bacterial translocation and colonization remain incompletely understood. Herein, we investigated whether extracellular vesicles derived from F. nucleatum (FnEVs) have impacts on bacterial colonization. In mice with colitis-related CRC, a notable enrichment of FnEVs was observed, leading to a significant increase in intratumor colonization by F. nucleatum and accelerated progression of CRC. The enrichment of FnEVs in clinical CRC tissues was demonstrated. Subsequently, we revealed that FnEVs undergo membrane fusion with CRC cells, leading to the transfer and retention of FomA on recipient cell surfaces. Given its ability to facilitate F. nucleatum autoaggregation through interaction with FN1441, the presence of FomA on CRC cell surfaces presents a target for bacterial adhesion. Collectively, the findings unveil a mechanism used by EVs to prepare a niche conducive for bacterial colonization in distal organs.


Asunto(s)
Adhesión Bacteriana , Neoplasias Colorrectales , Vesículas Extracelulares , Fusobacterium nucleatum , Fusobacterium nucleatum/fisiología , Vesículas Extracelulares/metabolismo , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/patología , Animales , Humanos , Ratones , Infecciones por Fusobacterium/microbiología , Línea Celular Tumoral , Proteínas Bacterianas/metabolismo , Colitis/microbiología , Colitis/patología , Modelos Animales de Enfermedad , Proteínas de la Membrana Bacteriana Externa
4.
PLoS Pathog ; 20(9): e1012541, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39250508

RESUMEN

Inflammatory bowel disease (IBD) is an immune system disorder primarily characterized by colitis, the exact etiology of which remains unclear. Traditional treatment approaches currently yield limited efficacy and are associated with significant side effects. Extensive research has indicated the potent therapeutic effects of probiotics, particularly Lactobacillus strains, in managing colitis. However, the mechanisms through which Lactobacillus strains ameliorate colitis require further exploration. In our study, we selected Lactobacillus gasseri ATCC33323 from the intestinal microbiota to elucidate the specific mechanisms involved in modulation of colitis. Experimental findings in a DSS-induced colitis mouse model revealed that L. gasseri ATCC33323 significantly improved physiological damage in colitic mice, reduced the severity of colonic inflammation, decreased the production of inflammatory factors, and preserved the integrity of the intestinal epithelial structure and function. It also maintained the expression and localization of adhesive proteins while improving intestinal barrier permeability and restoring dysbiosis in the gut microbiota. E-cadherin, a critical adhesive protein, plays a pivotal role in this protective mechanism. Knocking down E-cadherin expression within the mouse intestinal tract significantly attenuated the ability of L. gasseri ATCC33323 to regulate colitis, thus confirming its protective role through E-cadherin. Finally, transcriptional analysis and in vitro experiments revealed that L. gasseri ATCC33323 regulates CDH1 transcription by affecting NR1I3, thereby promoting E-cadherin expression. These findings contribute to a better understanding of the specific mechanisms by which Lactobacillus strains alleviate colitis, offering new insights for the potential use of L. gasseri as an alternative therapy for IBD, particularly in dietary supplementation.


Asunto(s)
Cadherinas , Colitis , Sulfato de Dextran , Mucosa Intestinal , Lactobacillus gasseri , Probióticos , Animales , Colitis/inducido químicamente , Colitis/microbiología , Colitis/metabolismo , Colitis/terapia , Cadherinas/metabolismo , Ratones , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Sulfato de Dextran/toxicidad , Probióticos/farmacología , Lactobacillus gasseri/metabolismo , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Humanos
5.
Food Res Int ; 195: 114950, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39277228

RESUMEN

Rosa roxburghii Tratt (RRT), a traditional Chinese plant known as the 'King of Vitamin C (VitC; ascorbic acid, AsA)', contains a wealth of nutrients and functional components, including polysaccharides, organic acids, flavonoids, triterpenes, and high superoxide dismutase (SOD) activity. The various functional components of RRT suggest that it may theoretically have a stronger potential for alleviating colitis compared to VitC. This study aims to verify whether RRT has a stronger ability to alleviate colitis than equimolar doses of VitC and to explore the mechanisms underlying this improvement. Results showed that RRT significantly mitigated body weight loss, intestinal damage, elevated inflammation levels, and compromised barriers in mice induced by Dextran sulfate sodium (DSS). Additionally, RRT enhanced the diversity and composition of intestinal microbiota in these DSS-induced mice. Colon RNA sequencing analysis revealed that compared to VitC, RRT further downregulated multiple immune-related signaling pathways, particularly the B cell receptor (BCR) pathway, which is centered around genes like Btk and its downstream PI3K-AKT, NF-κB, and MAPK signaling pathways. Correlation analysis between microbiota and genes demonstrated a significant relationship between the taxa improved by RRT and the key genes in the BCR and its downstream signaling pathways. Overall, RRT exhibited superior capabilities in alleviating DSS-induced colitis compared to VitC by decreasing intestinal inflammation and modulating BCR and its downstream signaling pathways, potentially regulated by the improved intestinal microbiota.


Asunto(s)
Ácido Ascórbico , Colitis , Sulfato de Dextran , Microbioma Gastrointestinal , Rosa , Transducción de Señal , Animales , Colitis/inducido químicamente , Colitis/metabolismo , Colitis/microbiología , Rosa/química , Ratones , Microbioma Gastrointestinal/efectos de los fármacos , Ratones Endogámicos C57BL , Extractos Vegetales/farmacología , Masculino , Colon/metabolismo , Colon/microbiología , Colon/efectos de los fármacos , Modelos Animales de Enfermedad
6.
NPJ Biofilms Microbiomes ; 10(1): 84, 2024 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-39266529

RESUMEN

The role of mast cells (MCs) in ulcerative colitis (UC) development is controversial. FcεRI, the IgE high-affinity receptor, is known to activate MCs. However, its role in UC remains unclear. In our study, Anti-FcεRI showed highly diagnostic value for UC. FcεRIα knockout in mice ameliorated DSS-induced colitis in a gut microbiota-dependent manner. Increased Lactobacillus abundance in FcεRIα deficient mice showed strongly correlation with the remission of colitis. RNA sequencing indicated activation of the NLRP6 inflammasome pathway in FcεRIα knockout mice. Additionally, Lactobacillus plantarum supplementation protected against inflammatory injury and goblet cell loss, with activation of the NLRP6 inflammasome during colitis. Notably, this effect was absent when the strain is unable to produce lactic acid. In summary, colitis was mitigated in FcεRIα deficient mice, which may be attributed to the increased abundance of Lactobacillus. These findings contribute to a better understanding of the relationship between allergic reactions, microbiota, and colitis.


Asunto(s)
Sulfato de Dextran , Microbioma Gastrointestinal , Receptores de IgE , Animales , Ratones , Colitis/prevención & control , Colitis/microbiología , Colitis/inducido químicamente , Colitis Ulcerosa/microbiología , Modelos Animales de Enfermedad , Inflamasomas/metabolismo , Lactobacillus , Lactobacillus plantarum/genética , Lactobacillus plantarum/fisiología , Mastocitos/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Probióticos , Receptores de IgE/genética
7.
Front Immunol ; 15: 1444045, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39229279

RESUMEN

Introduction: Colitis is an inflammatory bowel disease (IBD) characterized by immune cell dysregulation and alterations in the gut microbiome. In our previous report, we showed a natural product in cruciferous vegetables and ligand of the aryl hydrocarbon receptor (AhR), indole-3-carbinol (I3C), was able to reduce colitis-induced disease severity and microbial dysbiosis in an interleukin-22 (IL-22) dependent manner. Methods: In the current study, we performed single-cell RNA sequencing (scRNAseq) from colonocytes during colitis induction and supplementation with I3C and show how this treatment alters expression of genes involved in IL-22 signaling. To further define the role of IL-22 signaling in I3C-mediated protection during colitis and disease-associated microbial dysbiosis, we generated mice with AhR deficiency in RAR-related orphan receptor c (Rorc)-expressing cells (AhR ΔRorc ) which depletes this receptor in immune cells involved in production of IL-22. Colitis was induced in wildtype (WT), AhR ΔRorc , and littermate (LM) mice with or without I3C treatment. Results: Results showed AhR ΔRorc mice lost the efficacy effects of I3C treatment which correlated with a loss of ability to increase IL-22 by innate lymphoid type 3 (ILC3s), not T helper 22 (Th22) cells. 16S rRNA microbiome profiling studies showed AhR ΔRorc mice were unable to regulate disease-associated increases in Bacteroides, which differed between males and females. Lastly, inoculation with a specific disease-associated Bacteroides species, Bacteroides acidifaciens (B. acidifaciens), was shown to exacerbate colitis in females, but not males. Discussion: Collectively, this report highlights the cell and sex-specific role of AhR in regulating microbes that can impact colitis disease.


Asunto(s)
Bacteroides , Colitis , Interleucina-22 , Interleucinas , Receptores de Hidrocarburo de Aril , Receptores de Hidrocarburo de Aril/metabolismo , Receptores de Hidrocarburo de Aril/genética , Animales , Interleucinas/metabolismo , Colitis/inmunología , Colitis/microbiología , Femenino , Ratones , Masculino , Bacteroides/inmunología , Microbioma Gastrointestinal/inmunología , Disbiosis/inmunología , Ratones Endogámicos C57BL , Indoles/farmacología , Modelos Animales de Enfermedad , Factores Sexuales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Ratones Noqueados
8.
Cell Mol Life Sci ; 81(1): 397, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39261351

RESUMEN

Inflammatory bowel diseases (IBDs) are immune chronic diseases characterized by recurrent episodes, resulting in continuous intestinal barrier damage and intestinal microbiota dysbiosis. Safe strategies aimed at stabilizing and reducing IBDs recurrence have been vigorously pursued. Here, we constructed a recurrent intestinal injury Drosophila model and found that vitamin B12 (VB12), an essential co-factor for organism physiological functions, could effectively protect the intestine and reduce dextran sulfate sodium-induced intestinal barrier disruption. VB12 also alleviated microbial dysbiosis in the Drosophila model and inhibited the growth of gram-negative bacteria. We demonstrated that VB12 could mitigate intestinal damage by activating the hypoxia-inducible factor-1 signaling pathway in injured conditions, which was achieved by regulating the intestinal oxidation. In addition, we also validated the protective effect of VB12 in a murine acute colitis model. In summary, we offer new insights and implications for the potential supportive role of VB12 in the management of recurrent IBDs flare-ups.


Asunto(s)
Sulfato de Dextran , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Factor 1 Inducible por Hipoxia , Mucosa Intestinal , Transducción de Señal , Vitamina B 12 , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Vitamina B 12/farmacología , Vitamina B 12/metabolismo , Ratones , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Transducción de Señal/efectos de los fármacos , Sulfato de Dextran/toxicidad , Factor 1 Inducible por Hipoxia/metabolismo , Colitis/metabolismo , Colitis/inducido químicamente , Colitis/microbiología , Colitis/patología , Colitis/tratamiento farmacológico , Disbiosis/microbiología , Disbiosis/metabolismo , Ratones Endogámicos C57BL , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/patología , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Drosophila/metabolismo
9.
Molecules ; 29(17)2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39274922

RESUMEN

Previous studies have shown that Acanthopanax senticosus (AS) has a beneficial preventive and therapeutic effect on colitis. The fermentation of lactic acid bacteria (LAB) can alter the efficacy of AS by modifying or producing new compounds with potential bioactive properties. However, the specific components and mechanisms that enhance the efficacy are still unclear. In the present experiment, untargeted metabolomics was used to analyze the changes in active components before and after LAB fermentation of AS. The aim was to explain the mechanism of AS fermentation in treating colitis using a colitis model in mice. The results indicated that the fermentation of LAB could enhance the levels of total flavonoids and total polyphenols in FAS. Additionally, the beneficial components such as Delphinidin chloride, Diosmetin, Psoralidin, and Catechol significantly increased (p < 0.05). The colitis treatment experiment demonstrated that fermented AS could alleviate symptoms and improve the morphology of colitis in mice by enhancing antioxidant enzymes like CAT, T-SOD, and T-AOC. It also regulated the composition and abundance of intestinal flora species, such as Lactobacillus and Pseudogracilibacillus. The effectiveness of fermented AS was significantly superior to that of unfermented AS (p < 0.05). In conclusion, this study contributes to the application of lactic acid bacteria in AS fermentation and reveals the mechanism of fermentation AS for colitis.


Asunto(s)
Eleutherococcus , Fermentación , Microbioma Gastrointestinal , Lactobacillales , Metabolómica , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Eleutherococcus/química , Eleutherococcus/metabolismo , Ratones , Metabolómica/métodos , Lactobacillales/metabolismo , Colitis/microbiología , Colitis/metabolismo , Colitis/inducido químicamente , Masculino , Modelos Animales de Enfermedad , Metaboloma , Polifenoles/metabolismo , Polifenoles/farmacología , Flavonoides/metabolismo
10.
Nutrients ; 16(17)2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39275234

RESUMEN

Previous studies have indicated a critical role of intestinal bacteria in the pathogenesis of ulcerative colitis (UC). B. salyersiae is a commensal species from the human gut microbiota. However, what effect it has on UC development has not been investigated. In the present study, we explored this issue and demonstrated for the first time that oral administration of B. salyersiae CSP6, a bacterium previously isolated from the fecal sample of a healthy individual, protected against dextran sulfate sodium (DSS)-induced colitis in C57BL/6J mice. In particular, B. salyersiae CSP6 improved mucosal damage and attenuated gut dysbiosis in the colon of DSS-fed mice. Specifically, B. salyersiae CSP6 decreased the population of pathogenic Escherichia-Shigella spp. and increased the abundance of probiotic Dubosiella spp. and Bifidobacterium pseudolongum. Additionally, by reshaping the colonic microbiota, B. salyersiae CSP6 remarkably increased the fecal concentrations of equol, 8-deoxylactucin, and tiglic acid, three beneficial metabolites that have been well documented to exert strong anti-inflammatory effects. Altogether, our study provides novel evidence that B. salyersiae is a candidate probiotic species with potential anti-colitis properties in the human colon, which has applications for the development of next-generation probiotics.


Asunto(s)
Bacteroides , Colon , Sulfato de Dextran , Modelos Animales de Enfermedad , Heces , Microbioma Gastrointestinal , Ratones Endogámicos C57BL , Probióticos , Animales , Probióticos/farmacología , Humanos , Colon/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Ratones , Bacteroides/aislamiento & purificación , Heces/microbiología , Masculino , Colitis/microbiología , Colitis/inducido químicamente , Disbiosis/microbiología , Colitis Ulcerosa/microbiología
11.
Arch Microbiol ; 206(10): 398, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39254791

RESUMEN

Increasing awareness regarding health promotion and disease prevention has driven inclusion of fermented foods and beverages in the daily diet. These are the enormous sources of beneficial microbes, probiotics. This study aims to isolate yeast strains having probiotic potential and effectivity against colitis. Initially, ninety-two yeast strains were isolated from Haria, an ethnic fermented beverage of West Bengal, India. Primary screening was done by their acid (pH 4) and bile salt (0.3%) tolerance ability. Four potent isolates were selected and found effective against Entamoeba histolytica, as this human pathogen is responsible to cause colitis. They were identified as Saccharomyces cerevisiae. They showed luxurious growth even at 37 oC, tolerance up to 5% of NaCl, resistance to gastric juice and high bile salt (2.0%) and oro-gastrointestinal transit tolerance. They exhibited good auto-aggregation and co-aggregation ability and strong hydrophobicity. Finally, heat map and principal component analysis revealed that strain Y-89 was the best candidate. It was further characterised and found to have significant protective effects against DSS-induced colitis in experimental mice model. It includes improvement in colon length, body weight and organ indices; reduction in disease activity index; reduction in cholesterol, LDL, SGPT, SGOT, urea and creatinine levels; improvement in HDL, ALP, total protein and albumin levels; decrease in coliform count and restoration of tissue damage. This study demonstrates that the S. cerevisiae strain Y-89 possesses remarkable probiotic traits and can be used as a potential bio-therapeutic candidate for the prevention of colitis.


Asunto(s)
Colitis , Alimentos Fermentados , Probióticos , Saccharomyces cerevisiae , Probióticos/administración & dosificación , Probióticos/farmacología , Animales , Ratones , India , Colitis/microbiología , Colitis/inducido químicamente , Colitis/prevención & control , Alimentos Fermentados/microbiología , Modelos Animales de Enfermedad , Bebidas/microbiología , Masculino , Entamoeba histolytica , Humanos , Fermentación
12.
ISME J ; 18(1)2024 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-39180723

RESUMEN

Tryptophan (Trp) has been shown to regulate immune function by modulating gut serotonin (5-HT) metabolism and signaling. However, the mechanisms underlying the microbial modulation of gut 5-HT signaling in gut inflammation with gut microbiota dysbiosis require further investigation. Here, we investigated the effects of Trp supplementation on the composition and metabolism of the gut microbiome and 5-HT signaling-related gut immune function using a dextran sodium sulfate (DSS)-induced colitis mouse model coupled with antibiotic exposure. The results showed that antibiotic treatment before but not during DSS treatment decreased the immunoregulatory effects of Trp and aggravated gut inflammation and body weight loss in mice. Metagenomic analysis revealed that the fecal microbiota transplantation of Trp-enriched gut microbiota to recipient mice subject to antibiotic pre-exposure and DSS treatment alleviated inflammation by increasing the relative abundances of Lactobacillus and Parabacteroides and the microbial production of indole coupled with the activation of the 5-HT receptor 2B (HTR2B) in the colon. Transcriptomic analysis showed that HTR2B agonist administration strengthened the beneficial effects of Trp in DSS-induced colitis mice with antibiotic exposure by reducing gut lipopolysaccharide-binding protein (LBP) production, IκB-α/nuclear factor-κB signaling, and M1 macrophage polarization. Indole treatment reduced LBP production and M1 macrophage polarization both in mice with DSS-induced colitis and in lipopolysaccharide-treated mouse macrophages; however, the HTR2B antagonist reversed the effects of indole. Our findings provide the basis for developing new dietary and therapeutic interventions to improve gut microbiota dysbiosis-associated inflammatory gut disorders and diseases.


Asunto(s)
Proteínas Portadoras , Colitis , Colon , Sulfato de Dextran , Modelos Animales de Enfermedad , Disbiosis , Microbioma Gastrointestinal , Indoles , Macrófagos , Ratones Endogámicos C57BL , Triptófano , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Disbiosis/microbiología , Ratones , Colitis/inducido químicamente , Colitis/inmunología , Colitis/microbiología , Triptófano/metabolismo , Indoles/farmacología , Macrófagos/inmunología , Macrófagos/efectos de los fármacos , Colon/microbiología , Colon/inmunología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Fase Aguda/metabolismo , Masculino , Trasplante de Microbiota Fecal , Antibacterianos/farmacología , Transducción de Señal , Glicoproteínas de Membrana
13.
Biomed Pharmacother ; 179: 117292, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39151314

RESUMEN

A type of colorectal cancer (CRC),Colitis-associated colorectal cancer (CAC), is closely associated with chronic inflammation and gut microbiota dysbiosis. Berberine (BBR) has a long history in the treatment of intestinal diseases, which has been reported to inhibit colitis and CRC. However, the mechanism of its action is still unclear. Here, this study aimed to explore the potential protective effects of BBR on azoxymethane (AOM)/dextransulfate sodium (DSS)-induced colitis and tumor mice, and to elucidate its potential molecular mechanisms by microbiota, genes and metabolic alterations. The results showed that BBR inhibited the gut inflammation and improved the function of mucosal barrier to ameliorate AOM/DSS-induced colitis. And BBR treatment significantly reduced intestinal tumor development and ki-67 expression of intestinal tissue along with promoted apoptosis. Through microbiota analysis based on the 16 S rRNA gene, we found that BBR treatment improved intestinal microbiota imbalance in AOM/DSS-induced colitis and tumor mice, which were characterized by an increase of beneficial bacteria, for instance Akkermanisa, Lactobacillus, Bacteroides uniformis and Bacteroides acidifaciens. In addition, transcriptome analysis showed that BBR regulated colonic epithelial signaling pathway in CAC mice particularly by tryptophan metabolism and Wnt signaling pathway. Notably, BBR treatment resulted in the enrichment of amino acids metabolism and microbiota-derived SCFA metabolites. In summary, our research findings suggest that the gut microbiota-amino acid metabolism-Wnt signaling pathway axis plays critical role in maintaining intestinal homeostasis, which may provide new insights into the inhibitory effects of BBR on colitis and colon cancer.


Asunto(s)
Azoximetano , Berberina , Neoplasias Asociadas a Colitis , Colitis , Sulfato de Dextran , Microbioma Gastrointestinal , Metabolómica , Transcriptoma , Berberina/farmacología , Berberina/uso terapéutico , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Ratones , Azoximetano/toxicidad , Colitis/microbiología , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Neoplasias Asociadas a Colitis/microbiología , Neoplasias Asociadas a Colitis/tratamiento farmacológico , Neoplasias Asociadas a Colitis/patología , Masculino , Ratones Endogámicos C57BL , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/tratamiento farmacológico , Disbiosis , Modelos Animales de Enfermedad
14.
Sci Rep ; 14(1): 18618, 2024 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-39127850

RESUMEN

This study aimed to investigate whether class A1 scavenger receptor (SR-A1) regulated macrophage polarization and gut microbial alteration during intestinal inflammation of colitis. A murine colitis model was established by feeding with dextran sulfate sodium (DSS), and treatment groups were injected intravenously with SR-A1 antibody. Results showed a preventive effect on colitis symptoms and fewer inflammatory cell infiltrates in treatment groups. Down-regulation of inflammatory cytokines and up-regulation of anti-inflammatory cytokine related to macrophages were seen in murine PBMC and LPMC after injected with SR-A1 antibody. The percentage of M2 macrophages was also elevated in treatment groups. In addition, SR-A1 antibody treatment resulted in the decreased apoptosis and increased proliferation of colonic epithelial cells. Other findings indicated that SR-A1 antibody injection could mediate its anti-inflammatory effect via inhibiting TLR4-MyD88-NF-kB signaling pathway and alterating the gut microbiota composition. Our research identified SR-A1 as a potential therapeutic target in inflammatory bowel disease (IBD).


Asunto(s)
Colitis , Microbioma Gastrointestinal , Macrófagos , Receptores Depuradores de Clase A , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Colitis/inmunología , Colitis/inducido químicamente , Colitis/microbiología , Colitis/metabolismo , Ratones , Macrófagos/inmunología , Macrófagos/metabolismo , Receptores Depuradores de Clase A/metabolismo , Sulfato de Dextran/toxicidad , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 4/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Transducción de Señal , Modelos Animales de Enfermedad , Citocinas/metabolismo , Anticuerpos , FN-kappa B/metabolismo , Ratones Endogámicos C57BL , Masculino , Apoptosis/efectos de los fármacos
15.
J Extracell Vesicles ; 13(8): e12487, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39166405

RESUMEN

Inflammatory bowel disease (IBD) is a chronic disorder characterized by recurrent gastrointestinal inflammation, lacking a precise aetiology and definitive cure. The gut microbiome is vital in preventing and treating IBD due to its various physiological functions. In the interplay between the gut microbiome and human health, extracellular vesicles secreted by gut bacteria (BEVs) are key mediators. Herein, we explore the role of Roseburia intestinalis (R)-derived EVs (R-EVs) as potent anti-inflammatory mediators in treating dextran sulfate sodium-induced colitis. R was selected as an optimal BEV producer for IBD treatment through ANCOM analysis. R-EVs with a 76 nm diameter were isolated from R using a tangential flow filtration system. Orally administered R-EVs effectively accumulated in inflamed colonic tissues and increased the abundance of Bifidobacterium on microbial changes, inhibiting colonic inflammation and prompting intestinal recovery. Due to the presence of Ile-Pro-Ile in the vesicular structure, R-EVs reduced the DPP4 activity in inflamed colonic tissue and increased the active GLP-1, thereby downregulating the NFκB and STAT3 via the PI3K pathway. Our results shed light on the impact of BEVs on intestinal recovery and gut microbiome alteration in treating IBD.


Asunto(s)
Colitis , Vesículas Extracelulares , Microbioma Gastrointestinal , Vesículas Extracelulares/metabolismo , Animales , Colitis/metabolismo , Colitis/microbiología , Colitis/terapia , Ratones , Inflamación/metabolismo , Sulfato de Dextran , Humanos , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/terapia , Enfermedades Inflamatorias del Intestino/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Ratones Endogámicos C57BL , Masculino , Dipeptidil Peptidasa 4/metabolismo , FN-kappa B/metabolismo , Clostridiales/metabolismo
16.
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
17.
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
18.
Gut Microbes ; 16(1): 2387857, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39171684

RESUMEN

Imbalances in proteolytic activity have been linked to the development of inflammatory bowel diseases (IBD) and experimental colitis. Proteases in the intestine play important roles in maintaining homeostasis, but exposure of mucosal tissues to excess proteolytic activity can promote pathology through protease-activated receptors (PARs). Previous research implicates microbial proteases in IBD, but the underlying pathways and specific interactions between microbes and PARs remain unclear. In this study, we investigated the role of microbial proteolytic activation of the external domain of PAR2 in intestinal injury using mice expressing PAR2 with a mutated N-terminal external domain that is resistant to canonical activation by proteolytic cleavage. Our findings demonstrate the key role of proteolytic cleavage of the PAR2 external domain in promoting intestinal permeability and inflammation during colitis. In wild-type mice expressing protease-sensitive PAR2, excessive inflammation leads to the expansion of bacterial taxa that cleave the external domain of PAR2, exacerbating colitis severity. In contrast, mice expressing mutated protease-resistant PAR2 exhibit attenuated colitis severity and do not experience the same proteolytic bacterial expansion. Colonization of wild-type mice with proteolytic PAR2-activating Enterococcus and Staphylococcus worsens colitis severity. Our study identifies a previously unknown interaction between proteolytic bacterial communities, which are shaped by inflammation, and the external domain of PAR2 in colitis. The findings should encourage new therapeutic developments for IBD by targeting excessive PAR2 cleavage by bacterial proteases.


Asunto(s)
Colitis , Proteolisis , Receptor PAR-2 , Animales , Receptor PAR-2/metabolismo , Receptor PAR-2/genética , Colitis/microbiología , Colitis/patología , Colitis/metabolismo , Ratones , Microbioma Gastrointestinal , Ratones Endogámicos C57BL , Inflamación/metabolismo , Inflamación/microbiología , Enterococcus/genética , Enterococcus/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Bacterias/genética , Bacterias/metabolismo , Bacterias/clasificación , Bacterias/enzimología , Modelos Animales de Enfermedad , Humanos , Dominios Proteicos , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología
19.
mBio ; 15(9): e0204324, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39162488

RESUMEN

E3 ubiquitin ligase (E3) plays a vital role in regulating inflammatory responses by mediating ubiquitination. Previous studies have shown that ankyrin repeat and SOCS box-containing protein 3 (ASB3) is involved in immunomodulatory functions associated with cancer. However, the impact of ASB3 on the dynamic interplay of microbiota and inflammatory responses in inflammatory bowel disease (IBD) is unclear. Here, we systematically identify the E3 ligase ASB3 as a facilitative regulator in the development and progression of IBD. We observed that ASB3 exhibited significant upregulation in the lesions of patients with IBD. ASB3-/- mice are resistant to dextran sodium sulfate-induced colitis. IκBα phosphorylation levels and production of proinflammatory factors IL-1ß, IL-6, and TNF-α were reduced in the colonic tissues of ASB3-/- mice compared to WT mice. This colitis-resistant phenotype was suppressed after coprophagic microbial transfer and reversed after combined antibiotics removed the gut commensal microbiome. Mechanistically, ASB3 specifically catalyzes K48-linked polyubiquitination of TRAF6 in intestinal epithelial cells. In contrast, in ASB3-deficient organoids, the integrity of the TRAF6 protein is shielded, consequently decelerating the onset of intestinal inflammation. ASB3 is associated with dysregulation of the colitis microbiota and promotes proinflammatory factors' production by disrupting TRAF6 stability. Strategies to limit the protein level of ASB3 in intestinal epithelial cells may help in the treatment of colitis. IMPORTANCE: Ubiquitination is a key process that controls protein stability. We determined the ubiquitination of TRAF6 by ASB3 in intestinal epithelial cells during colonic inflammation. Inflammatory bowel disease patients exhibit upregulated ASB3 expression at focal sites, supporting the involvement of degradation of TRAF6, which promotes TLR-Myd88/TRIF-independent NF-κB aberrant activation and intestinal microbiota imbalance. Sustained inflammatory signaling in intestinal epithelial cells and dysregulated protective probiotic immune responses mediated by ASB3 collectively contribute to the exacerbation of inflammatory bowel disease. These findings provide insights into the pathogenesis of inflammatory bowel disease and suggest a novel mechanism by which ASB3 increases the risk of colitis. Our results suggest that future inhibition of ASB3 in intestinal epithelial cells may be a novel clinical strategy.


Asunto(s)
Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Ratones Noqueados , Factor 6 Asociado a Receptor de TNF , Animales , Humanos , Ratones , Colitis/microbiología , Colitis/inducido químicamente , Colitis/genética , Colitis/metabolismo , Modelos Animales de Enfermedad , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/inmunología , Ratones Endogámicos C57BL , Estabilidad Proteica , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Factor 6 Asociado a Receptor de TNF/genética , Ubiquitinación
20.
Int J Biol Macromol ; 278(Pt 3): 134860, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39163956

RESUMEN

Exploring nutritional therapies that manipulate tryptophan metabolism to activate AhR signaling represents a promising approach for mitigating chronic colitis. Arabinoxylan is a bioactive constituent abundant in wheat bran. Here, we comprehensively investigated anti-colitis potentials of wheat bran arabinoxylan (WBAX), its synbiotic and postbiotic derived from WBAX and Limosilactobacillus reuteri WX-94 (i.e., a probiotic strain exhibiting tryptophan metabolic activity). WBAX fueled L. reuteri and promoted microbial conversion of tryptophan to AhR ligands during in vitro fermentation in the culture medium and in the fecal microbiota from type 2 diabetes. The WBAX postbiotic outperformed WBAX and its synbiotic in augmenting efficacy of tryptophan in restoring DSS-disturbed serum immune markers, colonic tight junction proteins and gene profiles involved in amino acid metabolism and FoxO signaling. The WBAX postbiotic remodeled gut microbiota and superiorly enhanced AhR ligands (i.e., indole metabolites and bile acids), alongside with elevation in colonic AhR and IL-22. Associations between genera and metabolites modified by the postbiotic and colitis in human were verified and strong binding capacities between metabolites and colitis-related targets were demonstrated by molecular docking. Our study advances the novel perspective of WBAX in manipulating tryptophan metabolism and anti-colitis potentials of WBAX postbiotic via promoting gut microbiota-dependent AhR signaling.


Asunto(s)
Colitis , Fibras de la Dieta , Microbioma Gastrointestinal , Limosilactobacillus reuteri , Simbióticos , Xilanos , Xilanos/farmacología , Xilanos/química , Xilanos/metabolismo , Limosilactobacillus reuteri/metabolismo , Colitis/metabolismo , Colitis/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Fibras de la Dieta/metabolismo , Humanos , Animales , Ratones , Receptores de Hidrocarburo de Aril/metabolismo , Triptófano/metabolismo , Simulación del Acoplamiento Molecular , Fermentación , Masculino , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/microbiología , Multiómica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA