Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.694
Filtrer
1.
Sci Rep ; 14(1): 15160, 2024 07 02.
Article de Anglais | MEDLINE | ID: mdl-38956132

RÉSUMÉ

In order to survive and replicate, Salmonella has evolved mechanisms to gain access to intestinal epithelial cells of the crypt. However, the impact of Salmonella Typhimurium on stem cells and progenitors, which are responsible for the ability of the intestinal epithelium to renew and protect itself, remains unclear. Given that intestinal organoids growth is sustained by stem cells and progenitors activity, we have used this model to document the effects of Salmonella Typhimurium infection on epithelial proliferation and differentiation, and compared it to an in vivo model of Salmonella infection in mice. Among gut segments, the caecum was preferentially targeted by Salmonella. Analysis of infected crypts and organoids demonstrated increased length and size, respectively. mRNA transcription profiles of infected crypts and organoids pointed to upregulated EGFR-dependent signals, associated with a decrease in secretory cell lineage differentiation. To conclude, we show that organoids are suited to mimic the impact of Salmonella on stem cells and progenitors cells, carrying a great potential to drastically reduce the use of animals for scientific studies on that topic. In both models, the EGFR pathway, crucial to stem cells and progenitors proliferation and differentiation, is dysregulated by Salmonella, suggesting that repeated infections might have consequences on crypt integrity and further oncogenesis.


Sujet(s)
Différenciation cellulaire , Récepteurs ErbB , Organoïdes , Salmonelloses , Salmonella typhimurium , Cellules souches , Animaux , Organoïdes/microbiologie , Cellules souches/métabolisme , Souris , Salmonella typhimurium/pathogénicité , Salmonella typhimurium/physiologie , Salmonelloses/microbiologie , Salmonelloses/anatomopathologie , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Muqueuse intestinale/microbiologie , Muqueuse intestinale/anatomopathologie , Prolifération cellulaire , Modèles animaux de maladie humaine , Souris de lignée C57BL
2.
Cell Host Microbe ; 32(7): 1041-1043, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38991499

RÉSUMÉ

Multiple host and microbial factors dictate whether Candida albicans can colonize the mammalian gastrointestinal tract. In this issue of Cell Host & Microbe, Savage et al. demonstrate that restoration of intestinal epithelial hypoxia is sufficient to restore Candida albicans colonization resistance, even when other Candida inhibitory effectors remain depleted.


Sujet(s)
Candida albicans , Candidose , Tube digestif , Candida albicans/croissance et développement , Candida albicans/physiologie , Humains , Tube digestif/microbiologie , Candidose/microbiologie , Animaux , Hypoxie/métabolisme , Muqueuse intestinale/microbiologie , Muqueuse intestinale/métabolisme , Souris , Interactions hôte-pathogène , Microbiome gastro-intestinal/physiologie
3.
Cells ; 13(13)2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38994991

RÉSUMÉ

Clostridium perfringens (C. perfringens), a Gram-positive bacterium, produces a variety of toxins and extracellular enzymes that can lead to disease in both humans and animals. Common symptoms include abdominal swelling, diarrhea, and intestinal inflammation. Severe cases can result in complications like intestinal hemorrhage, edema, and even death. The primary toxins contributing to morbidity in C. perfringens-infected intestines are CPA, CPB, CPB2, CPE, and PFO. Amongst these, CPB, CPB2, and CPE are implicated in apoptosis development, while CPA is associated with cell death, increased intracellular ROS levels, and the release of the inflammatory factor IL-18. However, the exact mechanism by which PFO toxins exert their effects in the infected gut is still unidentified. This study demonstrates that a C. perfringens PFO toxin infection disrupts the intestinal epithelial barrier function through in vitro and in vivo models. This study emphasizes the notable influence of PFO toxins on intestinal barrier integrity in the context of C. perfringens infections. It reveals that PFO toxins increase ROS production by causing mitochondrial damage, triggering pyroptosis in IPEC-J2 cells, and consequently resulting in compromised intestinal barrier function. These results offer a scientific foundation for developing preventive and therapeutic approaches against C. perfringens infections.


Sujet(s)
Toxines bactériennes , Clostridium perfringens , Cellules épithéliales , Hémolysines , Muqueuse intestinale , Pyroptose , Espèces réactives de l'oxygène , Clostridium perfringens/pathogénicité , Toxines bactériennes/toxicité , Toxines bactériennes/métabolisme , Pyroptose/effets des médicaments et des substances chimiques , Animaux , Hémolysines/métabolisme , Hémolysines/toxicité , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/microbiologie , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Espèces réactives de l'oxygène/métabolisme , Lignée cellulaire , Souris , Humains , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques
4.
Int J Mol Sci ; 25(13)2024 Jul 06.
Article de Anglais | MEDLINE | ID: mdl-39000530

RÉSUMÉ

The fish gut microbiome is well known for its role in degrading nutrients to improve the host's digestion and absorption efficiency. In this study, we focused on the core physiological adaptability during the various reproductive stages of the black Amur bream (Megalobrama terminalis) to explore the interaction mechanisms among the fish host gut mucosal structure, gut enzyme activity, and gut microbial metabolism in the course of the host's reproductive cycle. Our findings showed that M. terminalis exhibited locomotion metabolic type (aids in sporting) in the reproductive stage, and a change to visceral metabolic type (aids in digestion) during non-reproductive and post-reproductive stage phases. The impact of metabolic type selection and energy demand during various reproductive stages on fish nutrition strategy and digestive function was substantial. Our resulted showed that mitochondria in intestinal epithelial cells of reproductive M. terminalis appeared autophagy phenomenon, and the digestive enzyme activities in the intestines of reproductive M. terminalis were lower than those in the non-reproductive and post-reproductive individuals. Moreover, these differences in nutrition strategy have a prominent impact on the gut microbiome of reproductive M. terminalis, compared to non-reproductive and post-reproductive samples. Our findings showed that reproductive females had lower levels of alpha diversity compared to non-reproductive and post-reproductive females. Our results also showed a greater functional variety and an increase in functional genes related to carbohydrate, lipid, amino acid, cofactors, and vitamin metabolic pathways in the NRS and PRS group. It is noteworthy that an enrichment of genes encoding putative enzymes implicated in the metabolism of taurine and hypotaurine was observed in the RS samples. Our findings illustrated that the stability and resilience of the gut bacterial community could be shaped in the wild fish host-microbiome interactions during reproductive life history.


Sujet(s)
Microbiome gastro-intestinal , Reproduction , Animaux , Microbiome gastro-intestinal/physiologie , Femelle , Cyprinidae/microbiologie , Cyprinidae/physiologie , Cyprinidae/métabolisme , Eau douce/microbiologie , Muqueuse intestinale/métabolisme , Muqueuse intestinale/microbiologie
5.
World J Gastroenterol ; 30(23): 2964-2980, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38946874

RÉSUMÉ

Metabolic dysfunction-associated fatty liver disease (MAFLD) is a hepatic manifestation of the metabolic syndrome. It is one of the most common liver diseases worldwide and shows increasing prevalence rates in most countries. MAFLD is a progressive disease with the most severe cases presenting as advanced fibrosis or cirrhosis with an increased risk of hepatocellular carcinoma. Gut microbiota play a significant role in the pathogenesis and progression of MAFLD by disrupting the gut-liver axis. The mechanisms involved in maintaining gut-liver axis homeostasis are complex. One critical aspect involves preserving an appropriate intestinal barrier permeability and levels of intestinal lumen metabolites to ensure gut-liver axis functionality. An increase in intestinal barrier permeability induces metabolic endotoxemia that leads to steatohepatitis. Moreover, alterations in the absorption of various metabolites can affect liver metabolism and induce liver steatosis and fibrosis. Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) are a class of drugs developed for the treatment of type 2 diabetes mellitus. They are also commonly used to combat obesity and have been proven to be effective in reversing hepatic steatosis. The mechanisms reported to be involved in this effect include an improved regulation of glycemia, reduced lipid synthesis, ß-oxidation of free fatty acids, and induction of autophagy in hepatic cells. Recently, multiple peptide receptor agonists have been introduced and are expected to increase the effectiveness of the treatment. A modulation of gut microbiota has also been observed with the use of these drugs that may contribute to the amelioration of MAFLD. This review presents the current understanding of the role of the gut-liver axis in the development of MAFLD and use of members of the GLP-1 RA family as pleiotropic agents in the treatment of MAFLD.


Sujet(s)
Microbiome gastro-intestinal , Récepteur du peptide-1 similaire au glucagon , Foie , Humains , Récepteur du peptide-1 similaire au glucagon/agonistes , Récepteur du peptide-1 similaire au glucagon/métabolisme , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Stéatose hépatique non alcoolique/traitement médicamenteux , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/microbiologie , Animaux , Syndrome métabolique X/traitement médicamenteux , Syndrome métabolique X/métabolisme , Syndrome métabolique X/microbiologie , Hypoglycémiants/usage thérapeutique , Hypoglycémiants/pharmacologie , Diabète de type 2/traitement médicamenteux , Diabète de type 2/métabolisme , Diabète de type 2/microbiologie , Incrétines/usage thérapeutique , Incrétines/métabolisme , Muqueuse intestinale/métabolisme , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/microbiologie ,
6.
Nutrients ; 16(13)2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38999794

RÉSUMÉ

Enterohemorrhagic Escherichia coli (EHEC) is a major food-borne pathogen that causes human disease ranging from diarrhea to life-threatening complications. Accumulating evidence demonstrates that the Western diet enhances the susceptibility to enteric infection in mice, but the effect of diet on EHEC colonization and the role of human gut microbiota remains unknown. Our research aimed to investigate the effects of a Standard versus a Western diet on EHEC colonization in the human in vitro Mucosal ARtificial COLon (M-ARCOL) and the associated changes in the gut microbiota composition and activities. After donor selection using simplified fecal batch experiments, two M-ARCOL bioreactors were inoculated with a human fecal sample (n = 4) and were run in parallel, one receiving a Standard diet, the other a Western diet and infected with EHEC O157:H7 strain EDL933. EHEC colonization was dependent on the donor and diet in the luminal samples, but was maintained in the mucosal compartment without elimination, suggesting a favorable niche for the pathogen, and may act as a reservoir. The Western diet also impacted the bacterial short-chain fatty acid and bile acid profiles, with a possible link between high butyrate concentrations and prolonged EHEC colonization. The work demonstrates the application of a complex in vitro model to provide insights into diet, microbiota, and pathogen interactions in the human gut.


Sujet(s)
Côlon , Régime occidental , Escherichia coli entérohémorrhagique , Fèces , Microbiome gastro-intestinal , Humains , Microbiome gastro-intestinal/physiologie , Régime occidental/effets indésirables , Côlon/microbiologie , Fèces/microbiologie , Infections à Escherichia coli/microbiologie , Muqueuse intestinale/microbiologie , Muqueuse intestinale/métabolisme , Acides gras volatils/métabolisme , Acides et sels biliaires/métabolisme , Escherichia coli O157
7.
Nutrients ; 16(13)2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38999800

RÉSUMÉ

In this study, we investigated the effect of monobutyrin (MB) on the gut microbiota and intestinal health of weaned mice. MB was administered via gavage to 21-day-old weaned mice. Samples of small intestinal and ileal contents were collected on day 1, day 7, and day 21 post-administration. Seven days of MB administration enhanced the mucin layer and morphological structure of the intestine and the integrity of the intestinal brush border. Both MB and sodium butyrate (SB) accelerated tight junction development. Compared to SB, MB modulated intestinal T cells in a distinct manner. MB increased the ratio of Treg cells in the small intestine upon the cessation of weaning. After 21 days of MB administration, enhancement of the villus structure of the ileum was observed. MB increased the proportion of Th17 cells in the ileum. MB facilitated the transition of the small intestinal microbiota toward an adult microbial community structure and enhanced the complexity of the microbial community structure. An increase in Th17 cells enhanced intestinal barrier function. The regulatory effect of MB on Th17 cells may occur through the intestinal microbiota. Therefore, MB can potentially be used to promote intestinal barrier function, especially for weaning animals, with promising application prospects.


Sujet(s)
Microbiome gastro-intestinal , Muqueuse intestinale , Cellules Th17 , Sevrage , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Souris , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/microbiologie , Mâle , Souris de lignée C57BL , Iléum/microbiologie , Intestin grêle/microbiologie , Intestin grêle/effets des médicaments et des substances chimiques , Acide butyrique/pharmacologie , Acide butyrique/métabolisme , Jonctions serrées/métabolisme , Jonctions serrées/effets des médicaments et des substances chimiques , Lymphocytes T régulateurs ,
8.
Sci Rep ; 14(1): 15798, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38982226

RÉSUMÉ

The present study aimed to explore the underlying mechanism of bile reflux-inducing chronic atrophic gastritis (CAG) with colonic mucosal lesion. The rat model of CAG with colonic mucosal lesion was induced by free-drinking 20 mmol/L sodium deoxycholate to simulate bile reflux and 2% cold sodium salicylate for 12 weeks. In comparison to the control group, the model rats had increased abundances of Bacteroidetes and Firmicutes but had decreased abundances of Proteobacteria and Fusobacterium. Several gut bacteria with bile acids transformation ability were enriched in the model group, such as Blautia, Phascolarctobacter, and Enterococcus. The cytotoxic deoxycholic acid and lithocholic acid were significantly increased in the model group. Transcriptome analysis of colonic tissues presented that the down-regulated genes enriched in T cell receptor signaling pathway, antigen processing and presentation, Th17 cell differentiation, Th1 and Th2 cell differentiation, and intestinal immune network for IgA production in the model group. These results suggest that bile reflux-inducing CAG with colonic mucosal lesion accompanied by gut dysbacteriosis, mucosal immunocompromise, and increased gene expressions related to repair of intestinal mucosal injury.


Sujet(s)
Côlon , Acide désoxycholique , Gastrite atrophique , Microbiome gastro-intestinal , Muqueuse intestinale , Animaux , Gastrite atrophique/microbiologie , Gastrite atrophique/immunologie , Gastrite atrophique/anatomopathologie , Gastrite atrophique/induit chimiquement , Rats , Muqueuse intestinale/anatomopathologie , Muqueuse intestinale/immunologie , Muqueuse intestinale/microbiologie , Muqueuse intestinale/effets des médicaments et des substances chimiques , Mâle , Côlon/anatomopathologie , Côlon/effets des médicaments et des substances chimiques , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Immunité muqueuse/effets des médicaments et des substances chimiques , Rat Sprague-Dawley , Maladie chronique
9.
Nat Commun ; 15(1): 5778, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38987259

RÉSUMÉ

Antimicrobial proteins contribute to host-microbiota interactions and are associated with inflammatory bowel disease (IBD), but our understanding on antimicrobial protein diversity and functions remains incomplete. Ribonuclease 4 (Rnase4) is a potential antimicrobial protein with no known function in the intestines. Here we find that RNASE4 is expressed in intestinal epithelial cells (IEC) including Paneth and goblet cells, and is detectable in human and mouse stool. Results from Rnase4-deficient mice and recombinant protein suggest that Rnase4 kills Parasutterella to modulate intestinal microbiome, thereby enhancing indoleamine-2,3-dioxygenase 1 (IDO1) expression and subsequently kynurenic and xanthurenic acid production in IECs to reduce colitis susceptibility. Furthermore, deceased RNASE4 levels are observed in the intestinal tissues and stool from patients with IBD, correlating with increased stool Parasutterella. Our results thus implicate Rnase4 as an intestinal antimicrobial protein regulating gut microbiota and metabolite homeostasis, and as a potential diagnostic biomarker and therapeutic target for IBD.


Sujet(s)
Microbiome gastro-intestinal , Homéostasie , Maladies inflammatoires intestinales , Souris de lignée C57BL , Microbiome gastro-intestinal/physiologie , Animaux , Humains , Souris , Maladies inflammatoires intestinales/microbiologie , Maladies inflammatoires intestinales/métabolisme , Colite/microbiologie , Colite/métabolisme , Colite/induit chimiquement , Muqueuse intestinale/métabolisme , Muqueuse intestinale/microbiologie , Souris knockout , Ribonucléases/métabolisme , Mâle , Fèces/microbiologie , Femelle , Intestins/microbiologie , Peptides antimicrobiens/métabolisme
10.
Semin Immunopathol ; 46(1-2): 2, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38990345

RÉSUMÉ

The gut microbiota, housing trillions of microorganisms within the gastrointestinal tract, has emerged as a critical regulator of host health and homeostasis. Through complex metabolic interactions, these microorganisms produce a diverse range of metabolites that substantially impact various physiological processes within the host. This review aims to delve into the intricate relationships of gut microbiota-derived metabolites and their influence on the host homeostasis. We will explore how these metabolites affect crucial aspects of host physiology, including metabolism, mucosal integrity, and communication among gut tissues. Moreover, we will spotlight the potential therapeutic applications of targeting these metabolites to restore and sustain host equilibrium. Understanding the intricate interplay between gut microbiota and their metabolites is crucial for developing innovative strategies to promote wellbeing and improve outcomes of chronic diseases.


Sujet(s)
Microbiome gastro-intestinal , Homéostasie , Humains , Microbiome gastro-intestinal/immunologie , Animaux , Muqueuse intestinale/métabolisme , Muqueuse intestinale/microbiologie , Muqueuse intestinale/immunologie , Métabolome , Interactions hôte-microbes/immunologie
11.
Gut Microbes ; 16(1): 2361493, 2024.
Article de Anglais | MEDLINE | ID: mdl-38958039

RÉSUMÉ

The juxtaposition of well-oxygenated intestinal colonic tissue with an anerobic luminal environment supports a fundamentally important relationship that is altered in the setting of intestinal injury, a process likely to be relevant to diseases such as inflammatory bowel disease. Herein, using two-color phosphorometry to non-invasively quantify both intestinal tissue and luminal oxygenation in real time, we show that intestinal injury induced by DSS colitis reduces intestinal tissue oxygenation in a spatially defined manner and increases the flux of oxygen from the tissue into the gut lumen. By characterizing the composition of the microbiome in both DSS colitis-affected gut and in a bioreactor containing a stable human fecal community exposed to microaerobic conditions, we provide evidence that the increased flux of oxygen into the gut lumen augments glycan degrading bacterial taxa rich in glycoside hydrolases which are known to inhabit gut mucosal surface. Continued disruption of the intestinal mucus barrier through such a mechanism may play a role in the perpetuation of the intestinal inflammatory process.


Sujet(s)
Bactéries , Colite , Microbiome gastro-intestinal , Muqueuse intestinale , Oxygène , Colite/microbiologie , Colite/induit chimiquement , Colite/métabolisme , Animaux , Humains , Oxygène/métabolisme , Bactéries/métabolisme , Bactéries/classification , Bactéries/isolement et purification , Bactéries/génétique , Souris , Muqueuse intestinale/métabolisme , Muqueuse intestinale/microbiologie , Fèces/microbiologie , Souris de lignée C57BL , Sulfate dextran , Côlon/microbiologie , Côlon/métabolisme , Mâle
12.
Sci Rep ; 14(1): 15460, 2024 07 04.
Article de Anglais | MEDLINE | ID: mdl-38965287

RÉSUMÉ

The gut microbiota plays a crucial role in neural development and progression of neural disorders like Parkinson's disease (PD). Probiotics have been suggested to impact neurodegenerative diseases via gut-brain axis. This study aims to investigate the therapeutic potential of Lacticaseibacillus rhamnosus E9, a high exopolysaccharide producer, on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine(MPTP)-induced mouse model of PD. C57BL/6 mice subjected to MPTP were fed L. rhamnosus E9 for fifteen days and sacrificed after the last administration. Motor functions were determined by open-field, catalepsy, and wire-hanging tests. The ileum and the brain tissues were collected for ELISA, qPCR, and immunohistochemistry analyses. The cecum content was obtained for microbiota analysis. E9 supplementation alleviated MPTP-induced motor dysfunctions accompanied by decreased levels of striatal TH and dopamine. E9 also reduced the level of ROS in the striatum and decreased the DAT expression while increasing the DR1. Furthermore, E9 improved intestinal integrity by enhancing ZO-1 and Occludin levels and reversed the dysbiosis of the gut microbiota induced by MPTP. In conclusion, E9 supplementation improved the MPTP-induced motor deficits and neural damage as well as intestinal barrier by modulating the gut microbiota in PD mice. These findings suggest that E9 supplementation holds therapeutic potential in managing PD through the gut-brain axis.


Sujet(s)
1-Méthyl-4-phényl-1,2,3,6-tétrahydropyridine , Modèles animaux de maladie humaine , Microbiome gastro-intestinal , Lacticaseibacillus rhamnosus , Souris de lignée C57BL , Probiotiques , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Souris , Lacticaseibacillus rhamnosus/physiologie , Mâle , Probiotiques/pharmacologie , Probiotiques/administration et posologie , Maladie de Parkinson/traitement médicamenteux , Maladie de Parkinson/métabolisme , Maladie de Parkinson/microbiologie , Corps strié/métabolisme , Intoxication au MPTP/microbiologie , Intoxication au MPTP/métabolisme , Intoxication au MPTP/traitement médicamenteux , Muqueuse intestinale/métabolisme , Muqueuse intestinale/microbiologie , Muqueuse intestinale/effets des médicaments et des substances chimiques , Dopamine/métabolisme
13.
Sci Rep ; 14(1): 15442, 2024 07 04.
Article de Anglais | MEDLINE | ID: mdl-38965312

RÉSUMÉ

The human intestinal tract is colonized with microorganisms, which present a diverse array of immunological challenges. A number of antimicrobial mechanisms have evolved to cope with these challenges. A key defense mechanism is the expression of inducible antimicrobial peptides (AMPs), such as beta-defensins, which rapidly inactivate microorganisms. We currently have a limited knowledge of mechanisms regulating the inducible expression of AMP genes, especially factors from the host required in these regulatory mechanisms. To identify the host factors required for expression of the beta-defensin-2 gene (HBD2) in intestinal epithelial cells upon a bacterial challenge, we performed a RNAi screen using a siRNA library spanning the whole human genome. The screening was performed in duplicate to select the strongest 79 and 110 hit genes whose silencing promoted or inhibited HBD2 expression, respectively. A set of 57 hits selected among the two groups of genes was subjected to a counter-screening and a subset was subsequently validated for its impact onto HBD2 expression. Among the 57 confirmed hits, we brought out the TLR5-MYD88 signaling pathway, but above all new signaling proteins, epigenetic regulators and transcription factors so far unrevealed in the HBD2 regulatory circuits, like the GATA6 transcription factor involved in inflammatory bowel diseases. This study represents a significant step toward unveiling the key molecular requirements to promote AMP expression in human intestinal epithelial cells, and revealing new potential targets for the development of an innovative therapeutic strategy aiming at stimulating the host AMP expression, at the era of antimicrobial resistance.


Sujet(s)
Cellules épithéliales , Muqueuse intestinale , bêta-Défensines , Humains , bêta-Défensines/métabolisme , bêta-Défensines/génétique , Muqueuse intestinale/métabolisme , Muqueuse intestinale/microbiologie , Cellules épithéliales/métabolisme , Cellules épithéliales/microbiologie , Transduction du signal , Régulation de l'expression des gènes , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme , Facteur de différenciation myéloïde-88/métabolisme , Facteur de différenciation myéloïde-88/génétique , Interférence par ARN
14.
Sci Rep ; 14(1): 15335, 2024 07 03.
Article de Anglais | MEDLINE | ID: mdl-38961176

RÉSUMÉ

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.


Sujet(s)
Désunion anastomotique , Microbiome gastro-intestinal , Humains , Mâle , Femelle , Sujet âgé , Désunion anastomotique/étiologie , Désunion anastomotique/microbiologie , Adulte d'âge moyen , Microbiome gastro-intestinal/génétique , Tumeurs colorectales/chirurgie , Tumeurs colorectales/microbiologie , ARN ribosomique 16S/génétique , Chirurgie colorectale/effets indésirables , Muqueuse intestinale/microbiologie , Muqueuse intestinale/anatomopathologie , Bactéries/classification , Bactéries/génétique , Bactéries/isolement et purification , Côlon/microbiologie , Côlon/chirurgie , Côlon/anatomopathologie , Étude de validation de principe
15.
Gut Microbes ; 16(1): 2379633, 2024.
Article de Anglais | MEDLINE | ID: mdl-39024479

RÉSUMÉ

Gut microbiota-derived extracellular vesicles (mEVs) are reported to regulate inflammatory response by delivering bacterial products into host cells. The complement receptor of the immunoglobulin superfamily macrophages (CRIg+ Mφ) could clear invading bacteria and their derivatives. Here, we investigate the role of CRIg+ Mφ and the mechanism by which mEVs regulate intestinal inflammation. We found that it is exacerbated in IBD patients and colitis mice by mEVs' leakage from disturbed gut microbiota, enriching microbial DNA in the intestinal mucosa. CRIg+ Mφ significantly decrease in IBD patients, allowing the spread of mEVs into the mucosa. The microbial DNA within mEVs is the key trigger for inflammation and barrier function damage. The cGAS/STING pathway is crucial in mEVs-mediated inflammatory injury. Blocking cGAS/STING signaling effectively alleviates inflammation caused by mEVs leakage and CRIg+ Mφ deficiency. Microbial DNA-containing mEVs, along with CRIg+ Mφ deficiency, stimulate inflammation in IBD, with the cGAS/STING pathway playing a crucial role.


Sujet(s)
ADN bactérien , Vésicules extracellulaires , Microbiome gastro-intestinal , Inflammation , Maladies inflammatoires intestinales , Muqueuse intestinale , Macrophages , Protéines membranaires , Nucleotidyltransferases , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/immunologie , Animaux , Souris , Macrophages/immunologie , Macrophages/microbiologie , Macrophages/métabolisme , Maladies inflammatoires intestinales/microbiologie , Maladies inflammatoires intestinales/anatomopathologie , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/immunologie , Humains , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Nucleotidyltransferases/métabolisme , Nucleotidyltransferases/génétique , Muqueuse intestinale/microbiologie , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Muqueuse intestinale/immunologie , Inflammation/microbiologie , Inflammation/métabolisme , ADN bactérien/génétique , Souris de lignée C57BL , Mâle , Femelle , Transduction du signal , Colite/microbiologie , Colite/anatomopathologie
16.
Gut Microbes ; 16(1): 2377570, 2024.
Article de Anglais | MEDLINE | ID: mdl-39034613

RÉSUMÉ

Recent evidence indicates that repeated antibiotic usage lowers microbial diversity and ultimately changes the gut microbiota community. However, the physiological effects of repeated - but not recent - antibiotic usage on microbiota-mediated mucosal barrier function are largely unknown. By selecting human individuals from the deeply phenotyped Estonian Microbiome Cohort (EstMB), we here utilized human-to-mouse fecal microbiota transplantation to explore long-term impacts of repeated antibiotic use on intestinal mucus function. While a healthy mucus layer protects the intestinal epithelium against infection and inflammation, using ex vivo mucus function analyses of viable colonic tissue explants, we show that microbiota from humans with a history of repeated antibiotic use causes reduced mucus growth rate and increased mucus penetrability compared to healthy controls in the transplanted mice. Moreover, shotgun metagenomic sequencing identified a significantly altered microbiota composition in the antibiotic-shaped microbial community, with known mucus-utilizing bacteria, including Akkermansia muciniphila and Bacteroides fragilis, dominating in the gut. The altered microbiota composition was further characterized by a distinct metabolite profile, which may be caused by differential mucus degradation capacity. Consequently, our proof-of-concept study suggests that long-term antibiotic use in humans can result in an altered microbial community that has reduced capacity to maintain proper mucus function in the gut.


Sujet(s)
Antibactériens , Bactéries , Transplantation de microbiote fécal , Microbiome gastro-intestinal , Mucus , Humains , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Animaux , Antibactériens/pharmacologie , Souris , Mucus/métabolisme , Mucus/microbiologie , Bactéries/classification , Bactéries/génétique , Bactéries/effets des médicaments et des substances chimiques , Bactéries/isolement et purification , Bactéries/métabolisme , Muqueuse intestinale/microbiologie , Muqueuse intestinale/métabolisme , Muqueuse intestinale/effets des médicaments et des substances chimiques , Mâle , Femelle , Fèces/microbiologie , Adulte , Adulte d'âge moyen , Akkermansia (genre) , Souris de lignée C57BL , Côlon/microbiologie , Bacteroides fragilis/effets des médicaments et des substances chimiques
17.
Immun Inflamm Dis ; 12(7): e1316, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39023417

RÉSUMÉ

BACKGROUND: The gastrointestinal tract contains a wide range of microorganisms that have evolved alongside the immune system of the host. The intestinal mucosa maintains balance within the intestines by utilizing the mucosal immune system, which is controlled by the complex gut mucosal immune network. OBJECTIVE: This review aims to comprehensively introduce current knowledge of the gut mucosal immune system, focusing on its interaction with commensal bacteria. RESULTS: The gut mucosal immune network includes gut-associated lymphoid tissue, mucosal immune cells, cytokines, and chemokines. The connection between microbiota and the immune system occurs through the engagement of bacterial components with pattern recognition receptors found in the intestinal epithelium and antigen-presenting cells. This interaction leads to the activation of both innate and adaptive immune responses. The interaction between the microbial community and the host is vital for maintaining the balance and health of the host's mucosal system. CONCLUSION: The gut mucosal immune network maintains a delicate equilibrium between active immunity, which defends against infections and damaging non-self antigens, and immunological tolerance, which allows for the presence of commensal microbiota and dietary antigens. This balance is crucial for the maintenance of intestinal health and homeostasis. Disturbance of gut homeostasis leads to enduring or severe gastrointestinal ailments, such as colorectal cancer and inflammatory bowel disease. Utilizing these factors can aid in the development of cutting-edge mucosal vaccines that have the ability to elicit strong protective immune responses at the primary sites of pathogen invasion.


Sujet(s)
Microbiome gastro-intestinal , Immunité muqueuse , Muqueuse intestinale , Humains , Microbiome gastro-intestinal/immunologie , Immunité muqueuse/immunologie , Muqueuse intestinale/immunologie , Muqueuse intestinale/microbiologie , Animaux , Symbiose/immunologie , Homéostasie/immunologie
18.
Front Immunol ; 15: 1424332, 2024.
Article de Anglais | MEDLINE | ID: mdl-39026673

RÉSUMÉ

Background: The protective role of gut microbiota and its metabolites against intestinal damage in sepsis patients remain unclear. Methods: Fecal samples were acquired from patients categorized into sepsis and non-sepsis groups for analysis of microbial composition via 16S rRNA sequencing and untargeted metabolomics analysis. We assessed the impact of gut microbiota from sepsis patients on intestinal barriers in antibiotic-treated mice. Furthermore, We conducted spearman's correlation analysis to examine the relationship between metabolites and the severity of sepsis. Additionally, we performed animal experiments to validate the functionality of identified metabolites. Results: The diversity of intestinal flora is decreased in patients with sepsis compared to the control group. Through fecal microbiota transplantation experiments, it was discovered that the gut microbiota derived from sepsis patients could induce intestinal damage in antibiotic-treated mice. Metabolomics analysis of the microbiota revealed a significant enrichment of the Valine, leucine, and isoleucine biosynthesis pathway. Further analysis showed a significant decrease in the abundance of L-valine in sepsis patients, which was negatively correlated with APACHE-II and SOFA scores. In sepsis mouse experiments, it was found that L-valine could alleviate sepsis-induced intestinal damage. Conclusion: Alterations in microbial and metabolic features in the gut can affect the severity of sepsis. Furthermore, L-valine can protect against sepsis-induced intestinal injury.


Sujet(s)
Microbiome gastro-intestinal , Sepsie , Valine , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Sepsie/microbiologie , Animaux , Souris , Humains , Valine/pharmacologie , Valine/usage thérapeutique , Mâle , Femelle , Adulte d'âge moyen , Transplantation de microbiote fécal , Indice de gravité de la maladie , Métabolomique/méthodes , Sujet âgé , Fèces/microbiologie , Modèles animaux de maladie humaine , Souris de lignée C57BL , Muqueuse intestinale/métabolisme , Muqueuse intestinale/microbiologie , Intestins/microbiologie , ARN ribosomique 16S/génétique
19.
Front Cell Infect Microbiol ; 14: 1389925, 2024.
Article de Anglais | MEDLINE | ID: mdl-39027133

RÉSUMÉ

Gut barrier is not only part of the digestive organ but also an important immunological organ for the hosts. The disruption of gut barrier can lead to various diseases such as obesity and colitis. In recent years, traditional Chinese medicine (TCM) has gained much attention for its rich clinical experiences enriched in thousands of years. After orally taken, TCM can interplay with gut microbiota. On one hand, TCM can modulate the composition and function of gut microbiota. On the other hand, gut microbiota can transform TCM compounds. The gut microbiota metabolites produced during the actions of these interplays exert noticeable pharmacological effects on the host especially gut barrier. Recently, a large number of studies have investigated the repairing and fortifying effects of TCM on gut barriers from the perspective of gut microbiota and its metabolites. However, no review has summarized the mechanism behand this beneficiary effects of TCM. In this review, we first briefly introduce the unique structure and specific function of gut barrier. Then, we summarize the interactions and relationship amidst gut microbiota, gut microbiota metabolites and TCM. Further, we summarize the regulative effects and mechanisms of TCM on gut barrier including physical barrier, chemical barrier, immunological barrier, and microbial barrier. At last, we discuss the effects of TCM on diseases that are associated gut barrier destruction such as ulcerative colitis and type 2 diabetes. Our review can provide insights into TCM, gut barrier and gut microbiota.


Sujet(s)
Médicaments issus de plantes chinoises , Microbiome gastro-intestinal , Médecine traditionnelle chinoise , Microbiome gastro-intestinal/physiologie , Humains , Médicaments issus de plantes chinoises/pharmacologie , Animaux , Muqueuse intestinale/microbiologie , Muqueuse intestinale/métabolisme , Diabète de type 2/microbiologie , Diabète de type 2/métabolisme , Rectocolite hémorragique/microbiologie , Rectocolite hémorragique/traitement médicamenteux
20.
Int J Mol Sci ; 25(13)2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-39000116

RÉSUMÉ

Metabolic endotoxemia is a severe health problem for residents in developed countries who follow a Western diet, disrupting intestinal microbiota and the whole organism's homeostasis. Although the effect of endotoxin on the human immune system is well known, its long-term impact on the human body, lasting many months or even years, is unknown. This is due to the difficulty of conducting in vitro and in vivo studies on the prolonged effect of endotoxin on the central nervous system. In this article, based on the available literature, we traced the path of endotoxin from the intestines to the blood through the intestinal epithelium and factors promoting the development of metabolic endotoxemia. The presence of endotoxin in the bloodstream and the inflammation it induces may contribute to lowering the blood-brain barrier, potentially allowing its penetration into the central nervous system; although, the theory is still controversial. Microglia, guarding the central nervous system, are the first line of defense and respond to endotoxin with activation, which may contribute to the development of neurodegenerative diseases. We traced the pro-inflammatory role of endotoxin in neurodegenerative diseases and its impact on the epigenetic regulation of microglial phenotypes.


Sujet(s)
Endotoxémie , Endotoxines , Microbiome gastro-intestinal , Maladies neurodégénératives , Endotoxémie/métabolisme , Endotoxémie/étiologie , Humains , Maladies neurodégénératives/métabolisme , Maladies neurodégénératives/étiologie , Animaux , Endotoxines/métabolisme , Microglie/métabolisme , Microglie/anatomopathologie , Barrière hémato-encéphalique/métabolisme , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Muqueuse intestinale/microbiologie , Inflammation/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE