Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.311
Filter
1.
Sci Rep ; 14(1): 23701, 2024 10 10.
Article in English | MEDLINE | ID: mdl-39390011

ABSTRACT

Fecal samples were collected from 640 individuals in Korea, including 523 patients with IBD (223 with Crohn's disease [CD] and 300 with ulcerative colitis [UC]) and 117 healthy controls. The samples were subjected to cross-sectional gut metagenomic analysis using 16 S rRNA sequencing and bioinformatics analysis. Patients with IBD, particularly those with CD, exhibited significantly lower alpha diversities than the healthy subjects. Differential abundance analysis revealed dysbiotic signatures, characterized by an expansion of the genus Escherichia-Shigella in patients with CD. Functional annotations showed that functional pathways related to bacterial pathogenesis and production of hydrogen sulfide (H2S) were strongly upregulated in patients with CD. A dysbiosis score, calculated based on functional characteristics, highly correlated with disease severity. Markers distinguishing between healthy subjects and patients with IBD showed accurate classification based on a small number of microbial taxa, which may be used to diagnose ambiguous cases. These findings confirm the taxonomic and functional dysbiosis of the gut microbiota in patients with IBD, especially those with CD. Taxa indicative of dysbiosis may have significant implications for future clinical research on the management and diagnosis of IBD.


Subject(s)
Biomarkers , Dysbiosis , Feces , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , RNA, Ribosomal, 16S , Humans , Gastrointestinal Microbiome/genetics , Dysbiosis/diagnosis , Dysbiosis/microbiology , Female , Male , Republic of Korea/epidemiology , Adult , Middle Aged , RNA, Ribosomal, 16S/genetics , Feces/microbiology , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/diagnosis , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/diagnosis , Metagenomics/methods , Crohn Disease/microbiology , Crohn Disease/diagnosis , Case-Control Studies , Cross-Sectional Studies , Young Adult , Aged
2.
Front Biosci (Landmark Ed) ; 29(9): 338, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39344335

ABSTRACT

BACKGROUND: Ulcerative colitis (UC) is an intestinal disorder marked by chronic, recurring inflammation, yet its underlying mechanisms have not been fully elucidated. METHODS: The current research dealt with examining the biological impacts of toll-like receptor 2 (TLR2) on dextran sulfate sodium (DSS)-triggered inflammation in the intestines of wild-type (WT) and TLR2-knockout (TLR2-KO) colitis mouse models. To elucidate the protective function of TLR2 in DSS-triggered colitis, RNA-sequencing (RNA-Seq) was carried out to compare the global gene expression data in the gut of WT and TLR2-KO mice. Further, 16S rRNA gene sequencing revealed notable variations in gut microbiota composition between WT and TLR2-KO colitis mice. RESULTS: It was revealed that TLR2-KO mice exhibited increased susceptibility to DSS-triggered colitis. RNA-Seq results demonstrated that cell cycle pathway-related genes were notably downregulated in TLR2-KO colitis mice (enrichment score = 30, p < 0.001). 16S rRNA gene sequencing revealed that in comparison to the WT colitis mice, the relative abundance of Marinifilacea (p = 0.006), Rikenellacea (p = 0.005), Desulfovibrionaceae (p = 0.045), Tannerellaceae (p = 0.038), Ruminococcaceae (p = 0.003), Clostridia (p = 0.027), and Mycoplasmataceae (p = 0.0009) was significantly increased at the family level in the gut of TLR2-KO colitis mice. In addition, microbiome diversity-transcriptome collaboration analysis highlighted that the relative abundance of Marinifilaceae was negatively linked to the expression of cell cycle signaling-related genes (p values were all less than 0.001). CONCLUSION: Based on these findings, we concluded that TLR2-KO exacerbates DSS-triggered intestinal injury by mitigating cell cycle signaling in a Marinifilaceae-dependent manner.


Subject(s)
Cell Cycle , Dextran Sulfate , Gastrointestinal Microbiome , Mice, Knockout , Signal Transduction , Toll-Like Receptor 2 , Animals , Dextran Sulfate/toxicity , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Cell Cycle/genetics , Mice , Mice, Inbred C57BL , Colitis/chemically induced , Colitis/genetics , Colitis/microbiology , Colitis/metabolism , RNA, Ribosomal, 16S/genetics , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/genetics , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/metabolism , Disease Models, Animal , Male
3.
Biochim Biophys Acta Mol Basis Dis ; 1870(8): 167489, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39233260

ABSTRACT

Inflammatory Bowel Disease (IBD) is a chronic inflammatory disorder of the gastrointestinal tract characterized by disrupted immune function. Indeed, gut microbiota dysbiosis and metabolomic profile alterations, are hallmarks of IBD. In this scenario, metabolite-sensing G-protein coupled receptors (GPCRs), involved in several biological processes, have emerged as pivotal players in the pathophysiology of IBD. The aim of this study was to characterize the axis microbiota-metabolite-GPCR in intestinal surgical resections from IBD patients. Results showed that UC patients had a lower microbiota richness and bacterial load, with a higher proportion of the genus Cellulosimicrobium and a reduced proportion of Escherichia, whereas CD patients showed a decreased abundance of Enterococcus. Furthermore, metabolomic analysis revealed alterations in carboxylic acids, fatty acids, and amino acids in UC and CD samples. These patients also exhibited upregulated expression of most metabolite-sensing GPCRs analysed, which positively correlated with pro-inflammatory and pro-fibrotic markers. The role of GPR109A was studied in depth and increased expression of this receptor was detected in epithelial cells and cells from lamina propria, including CD68+ macrophages, in IBD patients. The treatment with ß-hydroxybutyrate increased gene expression of GPR109A, CD86, IL1B and NOS2 in U937-derived macrophages. Besides, when GPR109A was transiently silenced, the mRNA expression and secretion of IL-1ß, IL-6 and TNF-α were impaired in M1 macrophages. Finally, the secretome from siGPR109A M1 macrophages reduced the gene and protein expression of COL1A1 and COL3A1 in intestinal fibroblasts. A better understanding of metabolite-sensing GPCRs, such as GPR109A, could establish their potential as therapeutic targets for managing IBD.


Subject(s)
Dysbiosis , Gastrointestinal Microbiome , Macrophages , Receptors, G-Protein-Coupled , Receptors, Nicotinic , Humans , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Dysbiosis/microbiology , Dysbiosis/metabolism , Receptors, Nicotinic/metabolism , Receptors, Nicotinic/genetics , Male , Macrophages/metabolism , Macrophages/microbiology , Female , Adult , Middle Aged , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Collagen Type I, alpha 1 Chain , Collagen Type I/metabolism , Collagen Type I/genetics , Crohn Disease/microbiology , Crohn Disease/metabolism , Crohn Disease/pathology
4.
Virulence ; 15(1): 2399217, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39221673

ABSTRACT

Fusobacterium nucleatum (F. nucleatum), an anaerobic resident of the oral cavity, is increasingly recognized as a contributing factor to ulcerative colitis (UC). The adhesive properties of F. nucleatum are mediated by its key virulence protein, FadA adhesin. However, further investigations are needed to understand the pathogenic mechanisms of this oral pathogen in UC. The present study aimed to explore the role of the FadA adhesin in the colonization and invasion of oral F. nucleatum in dextran sulphate sodium (DSS)-induced colitis mice via molecular techniques. In this study, we found that oral inoculation of F. nucleatum strain carrying the FadA adhesin further exacerbated DSS-induced colitis, leading to elevated alveolar bone loss, disease severity, and mortality. Additionally, CDH1 gene knockout mice treated with DSS presented increases in body weight and alveolar bone density, as well as a reduction in disease severity. Furthermore, FadA adhesin adhered to its mucosal receptor E-cadherin, leading to the phosphorylation of ß-catenin and the degradation of IκBα, the activation of the NF-κB signalling pathway and the upregulation of downstream cytokines. In conclusion, this research revealed that oral inoculation with F. nucleatum facilitates experimental colitis via the secretion of the virulence adhesin FadA. Targeting the oral pathogen F. nucleatum and its virulence factor FadA may represent a promising therapeutic approach for a portion of UC patients.


Subject(s)
Adhesins, Bacterial , Colitis, Ulcerative , Fusobacterium Infections , Fusobacterium nucleatum , Animals , Humans , Mice , Adhesins, Bacterial/metabolism , Adhesins, Bacterial/genetics , Bacterial Adhesion , Cadherins/metabolism , Colitis, Ulcerative/microbiology , Dextran Sulfate , Disease Models, Animal , Fusobacterium Infections/microbiology , Fusobacterium nucleatum/pathogenicity , Mice, Inbred C57BL , Mice, Knockout , Virulence , Virulence Factors/genetics , Virulence Factors/metabolism
5.
Sci Rep ; 14(1): 21504, 2024 09 14.
Article in English | MEDLINE | ID: mdl-39277703

ABSTRACT

WSGP has demonstrated significant potential for various bioactive effects. However, limited research has explored their anti-ulcerative colitis (UC) effects and mechanism on the colonic system and gut microbial metabolites. We evaluated the ameliorative effects of WSGP on the UC mice model. Using H&E to assess histological injury of colon morphology, AB-PAS staining to detect mucin secretion from goblet cells and the mucous layer, IF to evaluate the expression of intercellular tight junction proteins, ELISA to measure inflammatory factors, WB analysis to measure protein expression of inflammatory signaling pathways, RT-qPCR to quantify gene transcription of inflammatory factors, and LC-MS to analyze metabolites in mouse cecum contents. WSGP supplementation increased food intake, body weight, and colon length while reducing disease activity and histological scores in colitis-afflicted mice. WSGP mitigated colonic tissue damage and restored intestinal barrier integrity by suppressing NF-κB/STAT3 signaling, thereby decreasing gene transcription, protein expression of proinflammatory factors, and nitric oxide production. Additionally, WSGP improved UC by altering the variety of intestinal microbial metabolites. This study demonstrates that WSGP supplementation attenuates UC mice by suppressing the NF-κB/STAT3 signaling pathway, enhancing mucosal barrier function, reducing pro-inflammatory cytokines, and modulating gut microbial metabolites.


Subject(s)
Colitis, Ulcerative , Garlic , Gastrointestinal Microbiome , Intestinal Mucosa , Polysaccharides , Animals , Gastrointestinal Microbiome/drug effects , Mice , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Polysaccharides/pharmacology , Garlic/chemistry , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Disease Models, Animal , Male , Colon/metabolism , Colon/pathology , Colon/drug effects , Colon/microbiology , Signal Transduction/drug effects , NF-kappa B/metabolism , STAT3 Transcription Factor/metabolism , Water , Mice, Inbred C57BL
7.
Molecules ; 29(17)2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39274878

ABSTRACT

Ulcerative colitis (UC) is a typical inflammatory bowel disease (IBD), impairing the quality of life of patients. Dehydroevodiamine (DHE) is an active alkaloid isolated from Tetradium ruticarpum that exerts significant anti-inflammatory effects in gastrointestinal diseases. However, the effect and mechanisms of DHE on UC remain unclear. We performed a DSS-induced experimental UC rat model to reveal the efficacy and potential mechanisms of DHE on UC. HE and AB-PAS staining were used for the evaluation of pathologies, and 16S rRNA sequencing was used to detect changes in gut microbes. Metabolomics was used to detect changes in serum metabolites. Network pharmacology and transcriptomics were conducted to reveal the underlying mechanisms of DHE for UC. HuProt proteome microarrays, molecular docking, and SPR were used to reveal the targets of action of DHE. WB, RT-qPCR, and IHC were used to assess the action effects of DHE. DHE demonstrated significant alleviation of DSS-induced colitis symptoms in rats by suppressing inflammatory and oxidative stress responses, amending colonic barrier injury, and inhibiting apoptosis. In terms of gut microbial modulation, DHE decreased the abundance of Allobaculum, Clostridium, Escherichia, Enterococcus, and Barnesiella and increased the abundance of Lactobacillus, Bifidobacterium, and SMB5. Moreover, metabolomics suggested that the regulation of DHE in DSS-induced UC rats mainly involved aminoacyl-tRNA biosynthesis, vitamin B6 metabolism, phenylalanine, tyrosine, and so on. Mechanically, DHE alleviated UC in rats by targeting AKT1, thereby inhibiting the PI3K/AKT/NF-κB signaling pathway.


Subject(s)
Alkaloids , Colitis, Ulcerative , Gastrointestinal Microbiome , Signal Transduction , Animals , Male , Rats , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/microbiology , Dextran Sulfate , Disease Models, Animal , Gastrointestinal Microbiome/drug effects , Molecular Docking Simulation , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Alkaloids/administration & dosage
8.
Korean J Intern Med ; 39(5): 783-792, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39252488

ABSTRACT

BACKGROUND/AIMS: Fecal microbiota transplantation (FMT) is a promising therapy for inducing and maintaining remission in patients with ulcerative colitis (UC). However, FMT has not been approved for UC treatment in Korea. Our study aimed to investigate patient perceptions of FMT under the national medical policy. METHODS: This was a prospective, multicenter study. Patients with UC ≥ 19 years of age were included. Patients were surveyed using 22 questions on FMT. Changes in perceptions of FMT before and after education were also compared. RESULTS: A total of 210 patients with UC were enrolled. We found that 51.4% of the patients were unaware that FMT was an alternative treatment option for UC. After reading the educational materials on FMT, more patients were willing to undergo this procedure (27.1% vs. 46.7%; p < 0.001). The preferred fecal donor was the one recommended by a physician (41.0%), and the preferred transplantation method was the oral capsule (30.4%). A large proportion of patients (50.0%) reported that the national medical policy influenced their choice of FMT treatment. When patients felt severe disease activity, their willingness to undergo FMT increased (92.3% vs. 43.1%; p = 0.001). CONCLUSION: Education can increase preference for FMT in patients with UC. When patients have severe disease symptoms or their quality of life decreases their willingness to undergo FMT increases. Moreover, national medical policies may influence patient choices regarding FMT.


Subject(s)
Colitis, Ulcerative , Fecal Microbiota Transplantation , Health Knowledge, Attitudes, Practice , Humans , Colitis, Ulcerative/therapy , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/diagnosis , Male , Female , Adult , Republic of Korea , Middle Aged , Prospective Studies , Patient Education as Topic , Patient Preference , Treatment Outcome , Patient Acceptance of Health Care , Young Adult , Aged , Perception
9.
J Agric Food Chem ; 72(37): 20419-20431, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39249130

ABSTRACT

Flavonoid natural products are emerging as a promising approach for treating Ulcerative Colitis (UC) due to their natural origin and minimal toxicity. This study investigates the effects of Neohesperidin (NEO), a natural flavonoid, on Dextran Sodium Sulfate (DSS)-induced UC in mice, focusing on the underlying molecular mechanisms. Early intervention with NEO (25 and 50 mg/kg) mitigated colon shortening, restored damaged barrier proteins, and significantly reduced the inflammatory cytokine levels. Moreover, NEO inhibited the MAPK/NF-κB signaling pathway and enhanced the levels of intestinal barrier proteins (Claudin-3 and ZO-1). Additionally, NEO increased beneficial intestinal probiotics (S24-7 and Lactobacillaceae) while reducing harmful bacteria (Erysipelotrichi, Enterobacteriaceae). Fecal microbial transplantation (FMT) results demonstrated that NEO (50 mg/kg) markedly improved UC symptoms. In conclusion, early NEO intervention may alleviate DSS-induced UC by inhibiting inflammatory responses, preserving intestinal barrier integrity and modulating gut microbiota.


Subject(s)
Colitis, Ulcerative , Dextran Sulfate , Gastrointestinal Microbiome , Hesperidin , Intestinal Mucosa , Mice, Inbred C57BL , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/immunology , Animals , Gastrointestinal Microbiome/drug effects , Mice , Dextran Sulfate/adverse effects , Male , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Hesperidin/pharmacology , Hesperidin/administration & dosage , Hesperidin/analogs & derivatives , Humans , Bacteria/classification , Bacteria/drug effects , Bacteria/isolation & purification , Bacteria/genetics , NF-kappa B/metabolism , NF-kappa B/genetics , Colon/microbiology , Colon/drug effects , Colon/immunology , Colon/metabolism , Colon/pathology , Disease Models, Animal , Inflammation/drug therapy
10.
Int J Mol Sci ; 25(17)2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39273567

ABSTRACT

Recent evidence indicates that the gut microbiota (GM) has a significant impact on the inflammatory bowel disease (IBD) progression. Our aim was to investigate the GM profiles, the Microbial Dysbiosis Index (MDI) and the intestinal microbiota-associated markers in relation to IBD clinical characteristics and disease state. We performed 16S rRNA metataxonomy on both stools and ileal biopsies, metabolic dysbiosis tests on urine and intestinal permeability and mucosal immunity activation tests on the stools of 35 IBD paediatric patients. On the GM profile, we assigned the MDI to each patient. In the statistical analyses, the MDI was correlated with clinical parameters and intestinal microbial-associated markers. In IBD patients with high MDI, Gemellaceae and Enterobacteriaceae were increased in stools, and Fusobacterium, Haemophilus and Veillonella were increased in ileal biopsies. Ruminococcaceae and WAL_1855D were enriched in active disease condition; the last one was also positively correlated to MDI. Furthermore, the MDI results correlated with PUCAI and Matts scores in ulcerative colitis patients (UC). Finally, in our patients, we detected metabolic dysbiosis, intestinal permeability and mucosal immunity activation. In conclusion, the MDI showed a strong association with both severity and activity of IBD and a positive correlation with clinical scores, especially in UC. Thus, this evidence could be a useful tool for the diagnosis and prognosis of IBD.


Subject(s)
Biomarkers , Dysbiosis , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Precision Medicine , Humans , Dysbiosis/microbiology , Child , Female , Male , Inflammatory Bowel Diseases/microbiology , Adolescent , Precision Medicine/methods , RNA, Ribosomal, 16S/genetics , Feces/microbiology , Child, Preschool , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Intestinal Mucosa/metabolism , Ileum/microbiology , Ileum/pathology , Colitis, Ulcerative/microbiology
11.
Nutrients ; 16(17)2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39275234

ABSTRACT

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.


Subject(s)
Bacteroides , Colon , Dextran Sulfate , Disease Models, Animal , Feces , Gastrointestinal Microbiome , Mice, Inbred C57BL , Probiotics , Animals , Probiotics/pharmacology , Humans , Colon/microbiology , Gastrointestinal Microbiome/drug effects , Mice , Bacteroides/isolation & purification , Feces/microbiology , Male , Colitis/microbiology , Colitis/chemically induced , Dysbiosis/microbiology , Colitis, Ulcerative/microbiology
12.
Int Immunopharmacol ; 142(Pt B): 113230, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39321700

ABSTRACT

BACKGROUND AND AIM: Ulcerative colitis (UC) is a growing global health concern, with current treatments facing challenges like drug dependence and side effects. Fresh bamboo juice (FBJ), known for its antimicrobial and potential immune-modulating properties, has shown promise as a natural therapeutic agent. The present study aimed to explore the protective effects of FBJ against colitis and further analyze the changes of gut microbiota composition, metabolite profiles, and underlying immune mechanisms. MATERIALS AND METHODS: A colitis model in mice was established using DSS to investigate the effectiveness of FBJ. Intestinal tissue and fecal samples were also collected for 16S rRNA gene sequencing and liquid chromatography-mass spectrometry (LC-MS) analysis. Additionally, immunofluorescence and flow cytometry were employed to detect the proliferation and function of group 2 innate lymphoid cells (ILC2). Enzyme-linked immunosorbent assay (ELISA) was used to measure the cytokines secreted by immune cells. RESULTS: FBJ demonstrated significant therapeutic effects against DSS-induced colitis in mice. At the genus level, the abundance of Bacteroides, Akkermansia and unassigned bacteria in the bamboo juice group increased compared with the DSS group. In contrast, the abundance of Alloprevotella, Lactobacillus, Lachnospiraceae_NK4A136_group and Ruminococcaceae_UCG-014 significantly decreased. FBJ partially restored the balance of gut microbiota, as evidenced by the increased levels of beneficial bacteria. Metabolome analysis revealed significant alterations in fecal metabolites, including 3-Hydroxypyridine, Pyridoxine, SM(d18:1/16:0), and DL-Methionine sulfoxide were remarkably altered. Dysregulation of pathways such as Vitamin B6 metabolism, sphingolipid metabolism, and tyrosine metabolism was observed, which may contribute to protection against colitis. Flow cytometry and immunofluorescence showed a significant reduction in the proportion of ILC2 cells following FBJ treatment in the DSS group (1.82 % v.s. 3.18 %, P < 0.05). ELISA showed that the FBJ group had lower levels of IL-5, IL-6, IL-10, IL-13, IL-33, TNF-α, IFN-γ in intestinal tissue. CONCLUSIONS: Our findings demonstrate that FBJ exerts a protective effect against colitis, primarily by modulating the intestinal flora and metabolite profiles in mice with colitis. Furthermore, the observed alterations in bacterial flora and metabolites likely affect ILC2 function and cytokine production, thereby mediating the protective effects against colitis through modulation of the immune system.


Subject(s)
Cytokines , Gastrointestinal Microbiome , Sasa , Animals , Gastrointestinal Microbiome/drug effects , Mice , Cytokines/metabolism , Disease Models, Animal , Mice, Inbred C57BL , Lymphocytes/immunology , Lymphocytes/drug effects , Dextran Sulfate , Immunity, Innate/drug effects , Male , Colitis, Ulcerative/immunology , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Bacteria/classification , Feces/microbiology , Colitis/immunology , Colitis/chemically induced , Colitis/microbiology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism
13.
NPJ Biofilms Microbiomes ; 10(1): 84, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39266529

ABSTRACT

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.


Subject(s)
Dextran Sulfate , Gastrointestinal Microbiome , Receptors, IgE , Animals , Mice , Colitis/prevention & control , Colitis/microbiology , Colitis/chemically induced , Colitis, Ulcerative/microbiology , Disease Models, Animal , Inflammasomes/metabolism , Lactobacillus , Lactobacillus plantarum/genetics , Lactobacillus plantarum/physiology , Mast Cells/immunology , Mice, Inbred C57BL , Mice, Knockout , Probiotics , Receptors, IgE/genetics
14.
Sci Rep ; 14(1): 18558, 2024 08 09.
Article in English | MEDLINE | ID: mdl-39122767

ABSTRACT

Fecal microbial transplantation (FMT) offers promise for treating ulcerative colitis (UC), though the mechanisms underlying treatment failure are unknown. This study harnessed longitudinally collected colonic biopsies (n = 38) and fecal samples (n = 179) from 19 adults with mild-to-moderate UC undergoing serial FMT in which antimicrobial pre-treatment and delivery mode (capsules versus enema) were assessed for clinical response (≥ 3 points decrease from the pre-treatment Mayo score). Colonic biopsies underwent dual RNA-Seq; fecal samples underwent parallel 16S rRNA and shotgun metagenomic sequencing as well as untargeted metabolomic analyses. Pre-FMT, the colonic mucosa of non-responsive (NR) patients harbored an increased burden of bacteria, including Bacteroides, that expressed more antimicrobial resistance genes compared to responsive (R) patients. NR patients also exhibited muted mucosal expression of innate immune antimicrobial response genes. Post-FMT, NR and R fecal microbiomes and metabolomes exhibited significant divergence. NR metabolomes had elevated concentrations of immunostimulatory compounds including sphingomyelins, lysophospholipids and taurine. NR fecal microbiomes were enriched for Bacteroides fragilis and Bacteroides salyersiae strains that encoded genes capable of taurine production. These findings suggest that both effective mucosal microbial clearance and reintroduction of bacteria that reshape luminal metabolism associate with FMT success and that persistent mucosal and fecal colonization by antimicrobial-resistant Bacteroides species may contribute to FMT failure.


Subject(s)
Bacteroides , Colitis, Ulcerative , Fecal Microbiota Transplantation , Feces , Intestinal Mucosa , Humans , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/therapy , Colitis, Ulcerative/metabolism , Male , Female , Feces/microbiology , Bacteroides/genetics , Adult , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism , Middle Aged , Gastrointestinal Microbiome , Treatment Failure , RNA, Ribosomal, 16S/genetics , Metabolome
15.
Cell Host Microbe ; 32(9): 1502-1518.e9, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39197456

ABSTRACT

Probiotics are potential treatments for ulcerative colitis (UC), but their efficacy is frequently compromised by gastrointestinal conditions that limit adhesion and activity. Here, we use machine learning and bioinformatics to confirm that patients with UC have decreased prevalence of Lactobacillus genus and increased oxidative stress, which correlate with inflammation severity. Accordingly, we developed a probiotic-based therapeutic that synergistically restores intestinal redox and microbiota homeostasis. Lactobacillus casei (Lac) were induced to form a pericellular film, providing a polysaccharide network for spatially confined crystallization of ultrasmall but highly active selenium dots (Se-Lac). Upon oral administration, the selenium dot-embedded pericellular film efficiently enhanced gastric acid resistance and intestinal mucoadhesion of Lac cells. At the lesion site, the selenium dots scavenged reactive oxygen species, while Lac modulated the gut microbiota. In multiple mouse models and non-human primates, this therapeutic effectively relieved inflammation and reduced colonic damage, thus showing promise as a UC treatment.


Subject(s)
Colitis, Ulcerative , Gastrointestinal Microbiome , Homeostasis , Lacticaseibacillus casei , Oxidation-Reduction , Oxidative Stress , Probiotics , Colitis, Ulcerative/therapy , Colitis, Ulcerative/microbiology , Probiotics/pharmacology , Probiotics/administration & dosage , Animals , Gastrointestinal Microbiome/drug effects , Mice , Lacticaseibacillus casei/metabolism , Lacticaseibacillus casei/physiology , Humans , Oxidative Stress/drug effects , Disease Models, Animal , Selenium/pharmacology , Selenium/metabolism , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Male , Colon/microbiology , Colon/pathology , Female
16.
Int J Mol Sci ; 25(16)2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39201260

ABSTRACT

Ulcerative colitis (UC) is a persistent inflammatory intestinal disease that consistently affects the colon and rectum. Its exact cause remains unknown. UC causes a considerable challenge in healthcare, prompting research for novel therapeutic strategies. Although probiotics have gained popularity as possible candidates for managing UC, studies are still ongoing to identify the best probiotics or probiotic mixtures for clinical applications. This study aimed to determine the efficacy of a multi-strain probiotic mixture in mitigating intestinal inflammation in a colitis mouse model induced by dextran sulfate sodium. Specifically, a multi-strain probiotic mixture consisting of Tetragenococcus halophilus and Eubacterium rectale was used to study its impact on colitis symptoms. Anti-inflammatory effects were evaluated using ELISA and flow cytometry. The configuration of gut microbial communities was determined using 16S rRNA metagenomic analysis. According to this study, colitis mice treated with the probiotic mixture experienced reduced weight loss and significantly less colonic shortening compared to untreated mice. Additionally, the treated mice exhibited increased levels of forkhead box P3 (Foxp3) and interleukin 10, along with decreased expression of dendritic cell activation markers, such as CD40+, CD80+, and CD83+, in peripheral blood leukocytes and intraepithelial lymphocytes. Furthermore, there was a significant decrease in the frequencies of CD8+N.K1.1+ cells and CD11b+Ly6G+ cells. In terms of the gut microbiota, probiotic-mixture treatment of colitis mice significantly increased the abundance of the phyla Actinobacteria and Verrucomicrobia (p < 0.05). These results provide valuable insights into the therapeutic promise of multi-strain probiotics, shedding light on their potential to alleviate colitis symptoms. This research contributes to the ongoing exploration of effective probiotic interventions for managing inflammatory bowel disease.


Subject(s)
Colitis , Dextran Sulfate , Disease Models, Animal , Gastrointestinal Microbiome , Probiotics , Animals , Probiotics/pharmacology , Gastrointestinal Microbiome/drug effects , Mice , Colitis/microbiology , Colitis/therapy , Colitis/diet therapy , Colitis/chemically induced , Dextran Sulfate/toxicity , RNA, Ribosomal, 16S/genetics , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/therapy , Colitis, Ulcerative/immunology , Colitis, Ulcerative/diet therapy , Biomarkers , Mice, Inbred C57BL , Colon/microbiology , Colon/pathology , Colon/metabolism
17.
Int Immunopharmacol ; 140: 112846, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39121607

ABSTRACT

Ulcerative colitis (UC) is a chronic inflammatory condition with recurrent and challenging symptoms. Effective treatments are lacking, making UC management a critical research area. Morin (MO), a flavonoid from the Moraceae family, shows potential as an anti-UC agent, but its mechanisms are not fully understood. Using a dextran sulfate sodium (DSS)-induced UC mouse model, we employed network pharmacology to predict MO's therapeutic effects. Assessments included changes in body weight, disease activity index (DAI), and colon length. Immunofluorescence, hematoxylin and eosin (H&E), and PAS staining evaluated colon damage. ELISA and western blot analyzed inflammatory factors, tight junction (TJ)-associated proteins (Claudin-3, Occludin, ZO-1), and Mitogen-Activated Protein Kinase (MAPK)/ Nuclear Factor kappa B (NF-κB) pathways. 16S rRNA sequencing assessed gut microbiota diversity, confirmed by MO's modulation via Fecal Microbial Transplantation (FMT). Early MO intervention reduced UC severity by improving weight, DAI scores, and colon length, increasing goblet cells, enhancing barrier function, and inhibiting MAPK/NF-κB pathways. MO enriched gut microbiota, favoring beneficial bacteria like Muribaculaceae and Erysipelotrichaceae while reducing harmful Erysipelotrichaceae and Muribaculaceae. This study highlights MO's potential in UC management through inflammation control, mucosal integrity maintenance, and gut flora modulation.


Subject(s)
Colitis, Ulcerative , Dextran Sulfate , Flavonoids , Gastrointestinal Microbiome , Animals , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , Gastrointestinal Microbiome/drug effects , Flavonoids/pharmacology , Flavonoids/therapeutic use , Mice , Male , Disease Models, Animal , Mice, Inbred C57BL , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Colon/pathology , Colon/drug effects , Colon/microbiology , Colon/immunology , NF-kappa B/metabolism , Fecal Microbiota Transplantation , Humans , Flavones
18.
J Agric Food Chem ; 72(36): 19852-19868, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39197065

ABSTRACT

Alterations to the gut microbiota are associated with ulcerative colitis (UC), whereas restoration of normobiosis can effectively alleviate UC. l-Theanine has been shown to reshape the gut microbiota and regulate gut immunity. To investigate the mechanisms by which l-theanine alleviates UC, we used l-theanine and l-theanine fecal microbiota solution to treat UC mice. In this study, we used l-theanine and l-theanine fecal microbiota solution to treat UC mice to explore the mechanism by which l-theanine alleviates UC. By reducing inflammation in the colon, we demonstrated that l-theanine alleviates symptoms of UC. Meanwhile, l-theanine can improve the abundance of microbiota related to short-chain fatty acid, bile acid, and tryptophan production. Single-cell sequencing results indicated that l-theanine-mediated suppression of UC was associated with immune cell changes, especially regarding macrophages and T and B cells, and validated the immune cell responses to the gut microbiota. Further, flow cytometry results showed that the ability of dendritic cells, macrophages, and monocytes to present microbiota antigens to colonic T cells in an MHC-II-dependent manner was reduced after treating normal mouse fecal donors with l-theanine. These results demonstrate that l-theanine modulates colon adaptive and innate immunity by regulating the gut microbiota in an MHC-II-dependent manner, thereby alleviating UC.


Subject(s)
Colitis, Ulcerative , Colon , Gastrointestinal Microbiome , Glutamates , Mice, Inbred C57BL , Animals , Gastrointestinal Microbiome/drug effects , Mice , Glutamates/pharmacology , Glutamates/administration & dosage , Colitis, Ulcerative/immunology , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/microbiology , Colon/immunology , Colon/microbiology , Colon/drug effects , Male , Humans , Macrophages/immunology , Macrophages/drug effects , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/genetics , Bacteria/classification , Bacteria/drug effects , Bacteria/genetics , Bacteria/isolation & purification , Bacteria/immunology , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Feces/microbiology
19.
Int Immunopharmacol ; 141: 112995, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39191121

ABSTRACT

Zymogen granule 16 (ZG16) is a secretory glycoprotein found in zymogen granules, which also plays an important role in colorectal inflammation and cancer. Herein, a ZG16 gene knock-out (ZG16-/-) mouse line was established and we found that ZG16 deletion damaged the intestinal mucosal barrier and gut microbiota, which resulted in low-level inflammation and further promoted the development of ulcerative colitis and inflammation-related colorectal cancer. Meanwhile, a metabolomics analysis on mouse feces showed that the metabolites significantly differed between ZG16-/- and WT mice, which were important mediators of host-microbiota communication and may impact the pulmonary inflammation of mice. Indeed, ZG16-/- mice showed more severe inflammation in a bronchial asthma model. Taken together, the results demonstrate that ZG16 plays a pivotal role in inhibiting inflammation and regulating immune responses in colorectum and lung of experimental animals, which may provide a better understanding of the underlying mechanism of human inflammatory diseases associated with ZG16.


Subject(s)
Gastrointestinal Microbiome , Intestinal Mucosa , Mice, Inbred C57BL , Mice, Knockout , Animals , Gastrointestinal Microbiome/immunology , Mice , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Asthma/immunology , Asthma/metabolism , Asthma/microbiology , Colitis, Ulcerative/immunology , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/pathology , Colitis, Ulcerative/metabolism , Humans , Lung/immunology , Lung/pathology , Lung/metabolism , Disease Models, Animal , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Male , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Bacteria/metabolism , Bacteria/immunology , Glycoproteins/metabolism , Glycoproteins/genetics
20.
J Ethnopharmacol ; 335: 118645, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39089661

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Ulcerative colitis (UC) is a chronic inflammatory bowel condition that is frequently related with Spleen-Kidney Yang Deficiency Syndrome (SKYD) in Chinese medicine. Fuzi Lizhong Pill (FLZP), a traditional medicine for SKYD, has been utilized in China for generations, although the exact mechanism by which it treats UC is unknown. AIM OF THE STUDY: The goal of this study is to further understand FLZP's therapeutic mechanism in SKYD-associated UC. MATERIALS AND METHODS: To investigate the impact of FLZP on SKYD-associated UC, we used a comprehensive method that included serum metabolomics and gut microbiota profiling. The chemical composition of FLZP was determined using mass spectrometry. UC rats with SKYD were induced and treated with FLZP. Serum metabolomics and 16S rRNA microbial community analysis were used to evaluate FLZP's effects on endogenous metabolites and gut microbiota, respectively. Correlation analysis investigated the association between metabolites and intestinal flora. A metabolic pathway analysis was undertaken to discover putative FLZP action mechanisms. RESULTS: FLZP contains 109 components, including liquiritin (584.8176 µg/g), benzoylaconine (16.3087 µg/g), benzoylhypaconine (31.9583), and hypaconitine (8.1160 µg/g). FLZP predominantly regulated seven metabolites and eight metabolic pathways involved in amino acid and nucleotide metabolism, with an emphasis on energy metabolism and gastrointestinal digestion. FLZP also influenced intestinal flora variety, increasing probiotic abundance while decreasing pathogenic bacteria prevalence. An integrated investigation identified associations between changes in certain gut flora and energy metabolism, specifically the tricarboxylic acid (TCA) cycle. CONCLUSIONS: FLZP successfully cures UC in SKYD rats by regulating amino acid and energy metabolism. Its positive effects may include altering microbiota composition and metabolite profiles in UC rats with SKYD. These findings shed light on FLZP's mode of action and its implications for UC management.


Subject(s)
Colitis, Ulcerative , Drugs, Chinese Herbal , Gastrointestinal Microbiome , Rats, Sprague-Dawley , Yang Deficiency , Animals , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/chemically induced , Yang Deficiency/drug therapy , Drugs, Chinese Herbal/pharmacology , Gastrointestinal Microbiome/drug effects , Male , Rats , Metabolomics , RNA, Ribosomal, 16S/genetics , Spleen/drug effects , Spleen/metabolism , Kidney/drug effects , Kidney/metabolism , Metabolome/drug effects , Disease Models, Animal
SELECTION OF CITATIONS
SEARCH DETAIL