Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.398
Filtrer
1.
Food Chem ; 462: 140951, 2025 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-39213975

RÉSUMÉ

Inflammatory bowel disease is a multifaceted condition that is influenced by nutritional, microbial, environmental, genetic, psychological, and immunological factors. Polyphenols and polysaccharides have gained recognition for their therapeutic potential. This review emphasizes the biological effects of polyphenols and polysaccharides, and explores their antioxidant, anti-inflammatory, and microbiome-modulating properties in the management of inflammatory bowel disease (IBD). However, polyphenols encounter challenges, such as low stability and low bioavailability in the colon during IBD treatment. Hence, polysaccharide-based encapsulation is a promising solution to achieve targeted delivery, improved bioavailability, reduced toxicity, and enhanced stability. This review also discusses the significance of covalent and non-covalent interactions, and simple and complex encapsulation between polyphenols and polysaccharides. The administration of these compounds in appropriate quantities has proven beneficial in preventing the development of Crohn's disease and ulcerative colitis, ultimately leading to the management of IBD. The use of polyphenols and polysaccharides has been found to reduce histological scores and colon injury associated with IBD, increase the abundance of beneficial microbes, inhibit the development of colitis-associated cancer, promote the production of microbial end-products, such as short-chain fatty acids (SCFAs), and improve anti-inflammatory properties. Despite the combined effects of polyphenols and polysaccharides observed in both in vitro and in vivo studies, further human clinical trials are needed to comprehend their effectiveness on inflammatory bowel disease.


Sujet(s)
Anti-inflammatoires , Maladies inflammatoires intestinales , Polyphénols , Polyosides , Polyphénols/composition chimique , Polyphénols/pharmacologie , Polyphénols/administration et posologie , Humains , Polyosides/composition chimique , Polyosides/pharmacologie , Maladies inflammatoires intestinales/traitement médicamenteux , Maladies inflammatoires intestinales/métabolisme , Animaux , Anti-inflammatoires/composition chimique , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/administration et posologie , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Antioxydants/composition chimique , Antioxydants/pharmacologie
2.
Mymensingh Med J ; 33(4): 1149-1156, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39351737

RÉSUMÉ

Inflammatory bowel disease (IBD) is a chronic idiopathic inflammatory disorder of the gastrointestinal tract with relapsing and remitting course. Recurrent abdominal pain and discomfort in association with abnormal defecation in the absence of structural abnormality of the gut is the key feature of irritable bowel syndrome (IBS). Faecal biomarker may be used a precise tool in the differentiation of IBD and IBS. The aim of this study was to measure faecal calprotectin (FC) in patients with IBD and IBS and compare between them. This was a cross-sectional study done in the department of Gastroenterology, BSMMU, Bangladesh from May 2017 to August 2018. IBD patients were diagnosed on the basis of compatible history, clinical examination, laboratory, radiological and endoscopic findings. IBS patients were selected by using the Rome IV criteria. Quantitative faecal calprotectin ELISA (BUHLMANN Quantum Blue) test was done and compared between IBD and IBS patients. In this study, ninety (90) patients were enrolled, 45 patients with IBD and 45 patients with IBS. Mean age of the IBD patients was 32.24±9.76 years and IBS patients was 33.80±9.70 years. There were 28(62.2%) male and 17(37.8%) female patients with IBD and 30(66.7%) male and 15(33.3%) female patients with IBS. We found faecal calprotectin (FC) level was 445.68±237.35µg/gm in IBD patients and 39.16±17.31µg/gm in IBS patients. There was a significant difference of faecal calprotectin level between IBD and IBS patients (p-value <0.001). The sensitivity and specificity of faecal calprotectin to differentiate IBD from IBS was 91.1% and 86.7% respectively. The test accuracy was 88.9%. Area under ROC was 0.959 (95% CI, 0.909 to 1.0). This study showed that faecal calprotectin appears to be clinically useful, non-invasive, rapid and reliable marker to differentiate IBD from IBS.


Sujet(s)
Marqueurs biologiques , Fèces , Maladies inflammatoires intestinales , Syndrome du côlon irritable , Complexe antigénique L1 leucocytaire , Humains , Complexe antigénique L1 leucocytaire/analyse , Syndrome du côlon irritable/diagnostic , Femelle , Mâle , Fèces/composition chimique , Adulte , Maladies inflammatoires intestinales/diagnostic , Maladies inflammatoires intestinales/métabolisme , Études transversales , Diagnostic différentiel , Marqueurs biologiques/analyse , Marqueurs biologiques/métabolisme , Sensibilité et spécificité , Adulte d'âge moyen , Pertinence clinique
3.
Int J Biol Sci ; 20(12): 4618-4634, 2024.
Article de Anglais | MEDLINE | ID: mdl-39309424

RÉSUMÉ

Some colorectal cancer patients have experienced normal epithelial transformation into inflammatory and tumor states, but the molecular basis still needs to be further determined. The expression levels of SIX4 are gradually increased in dextran sodium sulfate (DSS) and azoxymethane (AOM)/DSS-induced colonic epithelial inflammation and tumors, respectively, in mice. Targeting SIX4 alleviates intestinal inflammation occurrence and reduces adenoma formation in mice. Clinical sample assays indicated that SIX4 is upregulated in inflammatory bowel disease (IBD) and colorectal cancer (CRC) tissues compared to normal colorectal tissues. In a subsequent study, we found that SIX4, transcriptionally activated by the proinflammatory IL-6/STAT3 signal, binds to c-Jun to transcribe IL-6, thus forming a positive IL-6/STAT3/SIX4/c-Jun feedback loop, which further induces intestinal inflammation occurrence. In addition, elevated SIX4 also induces the expression of DeltaNp63, rather than wild-type p63, by binding to its promoter and thus facilitates the activation of tumor stemness signals, which ultimately leads to the formation of colorectal cancer. Our study first observes that activated SIX4 in inflammation induction drives the transformation of colorectal epithelium into inflammation and tumor, which demonstrates SIX4 as a significant therapeutic target in IBD and colitis-associated colorectal cancer (CAC) and CRC pathogenesis.


Sujet(s)
Tumeurs colorectales , Inflammation , Transduction du signal , Animaux , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Souris , Inflammation/métabolisme , Humains , Interleukine-6/métabolisme , Protéines à homéodomaine/métabolisme , Protéines à homéodomaine/génétique , Sulfate dextran , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/anatomopathologie , Maladies inflammatoires intestinales/induit chimiquement , Souris de lignée C57BL , Muqueuse intestinale/métabolisme , Oxyde de diméthyl-diazène
4.
Cell Biol Toxicol ; 40(1): 74, 2024 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-39259386

RÉSUMÉ

Human umbilical cord mesenchymal stem cell-derived exosomes (hucMSC-Ex) have emerged as a new treatment strategy for inflammatory bowel disease (IBD) due to their immunoregulatory function. N6-methyladenosine (m6A) plays a crucial role in regulating intestinal immunity, especially in IBD where macrophages play an important role, although its mechanism is not yet fully understood. From this perspective, this research aimed to evaluate the effect of hucMSC-Ex on m6A modification of macrophages in IBD. In the process of alleviating inflammation, hucMSC-Ex promotes macrophage polarization toward the M2 type and regulates intracellular m6A levels by upregulating the expression of m6A "Writer" METTL3 and "Reader" YTHDF1. Solute Carrier Family 37 Member 2 (Slc37a2) was identified by Methylation RNA immunoprecipitation sequencing as the target molecule of the hucMSC-Ex. Mechanically, hucMSC-Ex promoted the binding of METTL3 to the Slc37a2 mRNA complex, and enhanced the binding of Slc37a2 to YTHDF1 to upregulate the intracellular expression of Slc37a2, thereby attenuating the pro-inflammatory function of macrophage. This study confirms the modulatory role of hucMSC-Ex on the m6A modification of macrophages in IBD, providing a new scientific basis for the treatment of IBD with hucMSC-Ex.


Sujet(s)
Maladies inflammatoires intestinales , Macrophages , Cellules souches mésenchymateuses , Methyltransferases , Protéines de liaison à l'ARN , Cordon ombilical , Animaux , Macrophages/métabolisme , Macrophages/immunologie , Souris , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/immunologie , Humains , Methyltransferases/métabolisme , Methyltransferases/génétique , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Cellules souches mésenchymateuses/métabolisme , Cordon ombilical/cytologie , Cordon ombilical/métabolisme , Exosomes/métabolisme , Adénosine/analogues et dérivés , Adénosine/métabolisme , Souris de lignée C57BL , Mâle , Activation des macrophages
5.
Int J Mol Sci ; 25(18)2024 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-39337509

RÉSUMÉ

Inflammatory bowel diseases (IBDs) involve chronic inflammation of the gastrointestinal tract, where effector CD4+ T-cells play a central role. Thereby, the recruitment of T-cells into the colonic mucosa represents a key process in IBD. We recently found that CCR9 and DRD5 might form a heteromeric complex on the T-cell surface. The increase in CCL25 production and the reduction in dopamine levels associated with colonic inflammation represent a dual signal stimulating the CCR9:DRD5 heteromer, which promotes the recruitment of CD4+ T-cells into the colonic lamina propria. Here, we aimed to analyse the molecular requirements involved in the heteromer assembly as well as to determine the underlying cellular mechanisms involved in the colonic tropism given by the stimulation of the CCR9:DRD5 complex. The results show that dual stimulation of the CCR9:DRD5 heteromer potentiates the phosphorylation of the myosin light chain 2 (MLC2) and the migration speed in confined microchannels. Accordingly, disrupting the CCR9:DRD5 assembly induced a sharp reduction in the pMLC2 in vitro, decreased the migratory speed in confined microchannels, and dampened the recruitment of CD4+ T-cells into the inflamed colonic mucosa. Furthermore, in silico analysis confirmed that the interface of interaction of CCR9:DRD5 is formed by the transmembrane segments 5 and 6 from each protomer. Our findings demonstrated that the CCR9:DRD5 heteromeric complex plays a fundamental role in the migration of CD4+ T-cells into the colonic mucosa upon inflammation. Thereby, the present study encourages the design of strategies for disassembling the formation of the CCR9:DRD5 as a therapeutic opportunity to treat IBD.


Sujet(s)
Lymphocytes T CD4+ , Muqueuse intestinale , Récepteurs CCR , Récepteur D5 de la dopamine , Transduction du signal , Récepteurs CCR/métabolisme , Récepteurs CCR/génétique , Humains , Lymphocytes T CD4+/métabolisme , Lymphocytes T CD4+/immunologie , Récepteur D5 de la dopamine/métabolisme , Récepteur D5 de la dopamine/génétique , Muqueuse intestinale/métabolisme , Côlon/métabolisme , Mouvement cellulaire , Dopamine/métabolisme , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/anatomopathologie , Maladies inflammatoires intestinales/immunologie
6.
J Transl Med ; 22(1): 813, 2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-39223577

RÉSUMÉ

Inflammatory bowel disease (IBD) represents a group of recurrent chronic inflammatory disorders associated with autoimmune dysregulation, typically characterized by neutrophil infiltration and mucosal inflammatory lesions. Neutrophils, as the earliest immune cells to arrive at inflamed tissues, play a dual role in the onset and progression of mucosal inflammation in IBD. Most of these cells specifically express CD177, a molecule increasingly recognized for its critical role in the pathogenesis of IBD. Under IBD-related inflammatory stimuli, CD177 is highly expressed on neutrophils and promotes their migration. CD177 + neutrophils activate bactericidal and barrier-protective functions at IBD mucosal inflammation sites and regulate the release of inflammatory mediators highly correlated with the severity of inflammation in IBD patients, thus playing a dual role. However, mitigating the detrimental effects of neutrophils in inflammatory bowel disease remains a challenge. Based on these data, we have summarized recent articles on the role of neutrophils in intestinal inflammation, with a particular emphasis on CD177, which mediates the recruitment, transepithelial migration, and activation of neutrophils, as well as their functional consequences. A better understanding of CD177 + neutrophils may contribute to the development of novel therapeutic targets to selectively modulate the protective role of this class of cells in IBD.


Sujet(s)
Protéines liées au GPI , Maladies inflammatoires intestinales , Granulocytes neutrophiles , Humains , Granulocytes neutrophiles/métabolisme , Granulocytes neutrophiles/immunologie , Maladies inflammatoires intestinales/immunologie , Maladies inflammatoires intestinales/anatomopathologie , Maladies inflammatoires intestinales/métabolisme , Protéines liées au GPI/métabolisme , Animaux , Récepteurs de surface cellulaire/métabolisme , Inflammation/anatomopathologie , Inflammation/immunologie , Isoantigènes/immunologie
7.
BMC Vet Res ; 20(1): 393, 2024 Sep 06.
Article de Anglais | MEDLINE | ID: mdl-39238011

RÉSUMÉ

BACKGROUND: Calprotectin, a damage-associated molecular pattern protein of the S100/calgranulin family, is a potential marker of gastrointestinal inflammation in dogs and mainly originates from activated macrophages and granulocytes. Increased calprotectin concentrations are reported in feces and serum samples from dogs with chronic inflammatory enteropathy (CIE), but mucosal calprotectin expression has not been extensively investigated in canine CIE. Thus, we aimed to evaluate gastrointestinal mucosal concentrations of calprotectin in 62 dogs (44 dogs with CIE compared to 18 healthy Beagles) using a particle-enhanced turbidimetric immunoassay method. Additionally, we assessed the relationship of gastric, duodenal, jejunal, ileal, and colonic mucosal calprotectin levels with the clinical disease severity (canine clinical inflammatory bowel disease activity index, CIBDAI), histopathologic findings, clinical outcome, and serum albumin concentrations to further evaluate the potential of calprotectin as a biomarker for CIE. RESULTS: Mucosal calprotectin concentrations in dogs with CIE were significantly higher in the duodenum (median: 276.2 µg/g) and colon (median: 298.2 µg/g) compared to healthy controls (median: 94.3 µg/g, P = 0.0039; and median: 112.0 µg/g, P = 0.0061). Similar numerical differences in the ileum and cecum were not statistically significant, and mucosal calprotectin concentrations correlated significantly among the different gastrointestinal segments. Histologic lesion severity was linked to mucosal calprotectin concentrations for inflammatory and structural histology criteria in the duodenum and colon (all P < 0.05). Higher mucosal calprotectin levels in the duodenum and across all segments correlated with lower serum albumin concentrations (both P < 0.05); duodenal mucosal calprotectin concentrations were more than sixfold higher in hypoalbuminemic dogs (median: 1441 µg/g, n = 4) than normoalbuminemic dogs (median: 227 µg/g, n = 40). There was no significant association of mucosal calprotectin levels with CIBDAI scores or individual clinical outcomes. CONCLUSIONS: These results show that duodenal and colonic mucosal calprotectin concentrations are increased in dogs with CIE, providing further supporting evidence for the diagnostic potential of fecal calprotectin (presumably reflecting mucosal) concentrations and in dogs with CIE. Further longitudinal research is needed to assess changes in mucosal calprotectin concentrations with clinical response to treatment vs. mucosal disease remission and to determine the clinical utility of fecal calprotectin concentrations to diagnose and monitor dogs with CIE in clinical practice.


Sujet(s)
Côlon , Maladies des chiens , Muqueuse intestinale , Complexe antigénique L1 leucocytaire , Animaux , Chiens , Maladies des chiens/métabolisme , Maladies des chiens/anatomopathologie , Complexe antigénique L1 leucocytaire/analyse , Muqueuse intestinale/anatomopathologie , Muqueuse intestinale/métabolisme , Mâle , Femelle , Côlon/anatomopathologie , Côlon/métabolisme , Duodénum/anatomopathologie , Duodénum/métabolisme , Marqueurs biologiques/métabolisme , Maladies inflammatoires intestinales/médecine vétérinaire , Maladies inflammatoires intestinales/anatomopathologie , Maladies inflammatoires intestinales/métabolisme , Indice de gravité de la maladie
8.
Sci Adv ; 10(37): eadp4119, 2024 Sep 13.
Article de Anglais | MEDLINE | ID: mdl-39259805

RÉSUMÉ

Antibiotic use is a risk factor for development of inflammatory bowel diseases (IBDs). IBDs are characterized by a damaged mucus layer, which does not separate the intestinal epithelium from the microbiota. Here, we hypothesized that antibiotics affect the integrity of the mucus barrier, which allows bacterial penetrance and predisposes to intestinal inflammation. We found that antibiotic treatment led to breakdown of the colonic mucus barrier and penetration of bacteria into the mucus layer. Using fecal microbiota transplant, RNA sequencing followed by machine learning, ex vivo mucus secretion measurements, and antibiotic treatment of germ-free mice, we determined that antibiotics induce endoplasmic reticulum stress in the colon that inhibits colonic mucus secretion in a microbiota-independent manner. This antibiotic-induced mucus secretion flaw led to penetration of bacteria into the colonic mucus layer, translocation of microbial antigens into circulation, and exacerbation of ulcerations in a mouse model of IBD. Thus, antibiotic use might predispose to intestinal inflammation by impeding mucus production.


Sujet(s)
Antibactériens , Côlon , Microbiome gastro-intestinal , Muqueuse intestinale , Mucus , Animaux , Antibactériens/pharmacologie , Antibactériens/effets indésirables , Souris , Muqueuse intestinale/métabolisme , Muqueuse intestinale/microbiologie , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/anatomopathologie , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Côlon/métabolisme , Côlon/effets des médicaments et des substances chimiques , Côlon/anatomopathologie , Côlon/microbiologie , Mucus/métabolisme , Maladies inflammatoires intestinales/induit chimiquement , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/anatomopathologie , Maladies inflammatoires intestinales/microbiologie , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Transplantation de microbiote fécal , Souris de lignée C57BL , Humains
9.
Cell Mol Life Sci ; 81(1): 397, 2024 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-39261351

RÉSUMÉ

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.


Sujet(s)
Sulfate dextran , Modèles animaux de maladie humaine , Microbiome gastro-intestinal , Facteur-1 induit par l'hypoxie , Muqueuse intestinale , Transduction du signal , Vitamine B12 , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Vitamine B12/pharmacologie , Vitamine B12/métabolisme , Souris , Muqueuse intestinale/métabolisme , Muqueuse intestinale/microbiologie , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques , Sulfate dextran/toxicité , Facteur-1 induit par l'hypoxie/métabolisme , Colite/métabolisme , Colite/induit chimiquement , Colite/microbiologie , Colite/anatomopathologie , Colite/traitement médicamenteux , Dysbiose/microbiologie , Dysbiose/métabolisme , Souris de lignée C57BL , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/microbiologie , Maladies inflammatoires intestinales/anatomopathologie , Maladies inflammatoires intestinales/traitement médicamenteux , Drosophila/métabolisme
10.
Nutrition ; 127: 112552, 2024 Nov.
Article de Anglais | MEDLINE | ID: mdl-39236524

RÉSUMÉ

OBJECTIVE: The present study aimed to compare measured and estimated resting metabolic rate (RMR) predicted by selected equations in patients with nonactive inflammatory bowel disease (IBD) on an outpatient university clinic regimen. RESEARCH METHODS & PROCEDURES: Seventy-two adult (≥20 years) IBD patients (45 with Crohn's disease-CD) had RMR measured (mRMR) by indirect calorimetry and also estimated by predictive equations (Cunningham, Henry, Anjos et al., and Marra et al.). Body composition was assessed by DXA. Absolute Bias (estimated - mRMR) and % Bias (Bias/mRMR) were calculated. Agreement was assessed as the limit of agreement (LoA) in the Bland & Altman approach. RESULTS: There was no difference in age, body composition and mRMR between individuals with CD (5414.2 ± 1023.7 kJ/day) and ulcerative colitis (5443.9 ± 1008.9 kJ/day). Among the equations, only the Anjos et al.'s population-specific equation (-52.1 [642.0] kJ/day, P = 0.493; LoA: -1311; 1206 kJ/d) accurately estimated RMR. The equations of Marra et al. produced the highest % Bias (24.1 ± 14.8%). The Bland & Altman plots showed that the range of the LoA was relatively similar for all equations. In the simple regression analysis, the model with FFM resulted in a higher coefficient of determination (R2 = 0.51 for DC 0.74 for UC) compared to the model that included BM (R2 = 0.35 for DC and 0.65 for UC). CONCLUSIONS: Among the equations analyzed, only Anjos et al.'s accurately estimated RMR in outpatients with nonactive IBD. However, caution is advised when applying it at the individual level, due to the wide observed LoA.


Sujet(s)
Métabolisme basal , Composition corporelle , Calorimétrie indirecte , Maladies inflammatoires intestinales , Humains , Métabolisme basal/physiologie , Mâle , Femelle , Adulte , Calorimétrie indirecte/méthodes , Adulte d'âge moyen , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/physiopathologie , Rectocolite hémorragique/physiopathologie , Rectocolite hémorragique/métabolisme , Maladie de Crohn/métabolisme , Maladie de Crohn/physiopathologie , Jeune adulte
11.
Int J Mol Sci ; 25(17)2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-39273457

RÉSUMÉ

Inflammatory bowel disease (IBD) is a chronic non-specific intestinal inflammatory disease that affects millions of people worldwide, and current treatment methods have certain limitations. This study aimed to explore the therapeutic potential and mechanism of action of lemairamin (Wgx-50) in inflammatory bowel disease (IBD). We used dextran sulfate sodium (DSS)-treated zebrafish as an inflammatory bowel disease model, and observed the effect of Wgx-50 on DSS-induced colitis inflammation. The results of the study showed that Wgx-50 could reduce the expression of pro-inflammatory cytokines induced by DSS and inhibit the recruitment of neutrophils to the site of intestinal injury. Further experiments revealed that Wgx-50 exerted its anti-inflammatory effect by regulating the activation of the Akt pathway. These research findings indicate that Wgx-50 possesses anti-inflammatory activity.


Sujet(s)
Anti-inflammatoires , Colite , Sulfate dextran , Modèles animaux de maladie humaine , Danio zébré , Animaux , Sulfate dextran/effets indésirables , Anti-inflammatoires/pharmacologie , Colite/induit chimiquement , Colite/traitement médicamenteux , Colite/métabolisme , Colite/anatomopathologie , Cytokines/métabolisme , Granulocytes neutrophiles/effets des médicaments et des substances chimiques , Granulocytes neutrophiles/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Maladies inflammatoires intestinales/traitement médicamenteux , Maladies inflammatoires intestinales/induit chimiquement , Maladies inflammatoires intestinales/anatomopathologie , Maladies inflammatoires intestinales/métabolisme , Intestins/effets des médicaments et des substances chimiques , Intestins/anatomopathologie , Protéines proto-oncogènes c-akt/métabolisme , Inflammation/traitement médicamenteux , Inflammation/anatomopathologie , Inflammation/induit chimiquement , Inflammation/métabolisme
12.
Int J Mol Sci ; 25(17)2024 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-39273536

RÉSUMÉ

Inflammatory bowel disease (IBD) is an incurable, chronic disorder of the gastrointestinal tract whose incidence increases every year. Scientific research constantly delivers new information about the disease and its multivariate, complex etiology. Nevertheless, full discovery and understanding of the complete mechanism of IBD pathogenesis still pose a significant challenge to today's science. Recent studies have unanimously confirmed the association of gut microbial dysbiosis with IBD and its contribution to the regulation of the inflammatory process. It transpires that the altered composition of pathogenic and commensal bacteria is not only characteristic of disturbed intestinal homeostasis in IBD, but also of viruses, parasites, and fungi, which are active in the intestine. The crucial function of the microbial metabolome in the human body is altered, which causes a wide range of effects on the host, thus providing a basis for the disease. On the other hand, human genomic and functional research has revealed more loci that play an essential role in gut homeostasis regulation, the immune response, and intestinal epithelial function. This review aims to organize and summarize the currently available knowledge concerning the role and interaction of crucial factors associated with IBD pathogenesis, notably, host genetic composition, intestinal microbiota and metabolome, and immune regulation.


Sujet(s)
Dysbiose , Microbiome gastro-intestinal , Maladies inflammatoires intestinales , Métabolome , Humains , Maladies inflammatoires intestinales/microbiologie , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/génétique , Dysbiose/microbiologie , Animaux
13.
Gut Microbes ; 16(1): 2394249, 2024.
Article de Anglais | MEDLINE | ID: mdl-39224018

RÉSUMÉ

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.


Sujet(s)
Colite , Fèces , Microbiome gastro-intestinal , Maladies inflammatoires intestinales , microARN , microARN/génétique , microARN/métabolisme , Animaux , Humains , Fèces/microbiologie , Souris , Maladies inflammatoires intestinales/microbiologie , Maladies inflammatoires intestinales/génétique , Maladies inflammatoires intestinales/métabolisme , Colite/microbiologie , Colite/induit chimiquement , Colite/génétique , Inflammation/microbiologie , Inflammation/métabolisme , Dysbiose/microbiologie , Souris de lignée C57BL , Femelle , Souris knockout , Bactéries/classification , Bactéries/génétique , Bactéries/isolement et purification , Mâle , Muqueuse intestinale/microbiologie , Muqueuse intestinale/métabolisme , Cytokines/métabolisme , Macrophages/immunologie , Macrophages/microbiologie , Macrophages/métabolisme , Modèles animaux de maladie humaine , Interleukine-10/génétique , Interleukine-10/métabolisme
14.
PLoS One ; 19(9): e0309921, 2024.
Article de Anglais | MEDLINE | ID: mdl-39250478

RÉSUMÉ

Multi-omics analysis offers a promising avenue to a better understanding of complex biological phenomena. In particular, untangling the pathophysiology of multifactorial health conditions such as the inflammatory bowel disease (IBD) could benefit from simultaneous consideration of several omics levels. However, taking full advantage of multi-omics data requires the adoption of suitable new tools. Multi-view learning, a machine learning technique that natively joins together heterogeneous data, is a natural source for such methods. Here we present a new approach to variable selection in unsupervised multi-view learning by applying stability selection to canonical correlation analysis (CCA). We apply our method, StabilityCCA, to simulated and real multi-omics data, and demonstrate its ability to find relevant variables and improve the stability of variable selection. In a case study on an IBD microbiome data set, we link together metagenomics and metabolomics, revealing a connection between their joint structure and the disease, and identifying potential biomarkers. Our results showcase the usefulness of multi-view learning in multi-omics analysis and demonstrate StabilityCCA as a powerful tool for biomarker discovery.


Sujet(s)
Marqueurs biologiques , Maladies inflammatoires intestinales , Métabolomique , Humains , Marqueurs biologiques/métabolisme , Maladies inflammatoires intestinales/métabolisme , Métabolomique/méthodes , Métagénomique/méthodes , Apprentissage machine , Microbiome gastro-intestinal , Multi-omique
15.
Adv Immunol ; 163: 1-20, 2024.
Article de Anglais | MEDLINE | ID: mdl-39271257

RÉSUMÉ

Autophagy, a highly conserved catabolic process that targets various types of cellular cargoes to lysosomal degradation, is one of the most important biological mechanisms critical for cellular homeostasis. Components of these cellular cargoes can range from individual proteins to invading pathogens, and degrading these materials is important for maintaining organismal health and survival. The process of autophagy is carried out by complex molecular mechanisms, and a growing body of evidence indicates that these mechanisms intersect with those involved in the cell death pathways. In this review, we examine several emerging studies elucidating the role of autophagy in RIP1-mediated cell death signaling, with particular emphasis on impaired autophagy caused by ATG16L1 deficiency. We also discuss how autophagy in RIP1-mediated cell death affects intestinal homeostasis in preclinical models, and the implications of the intersection between RIP1 and autophagy for understanding the intestinal pathologies associated with inflammatory bowel disease (IBD). Finally, we highlight the potential benefits of therapeutic targeting of RIP1 and autophagy proteins, while also proposing areas of research that will likely elucidate new links between autophagy and cell death signaling.


Sujet(s)
Autophagie , Complexe protéique du pore nucléaire , Protéines de liaison à l'ARN , Transduction du signal , Animaux , Humains , Mort cellulaire , Inflammation/immunologie , Inflammation/métabolisme , Maladies inflammatoires intestinales/immunologie , Maladies inflammatoires intestinales/métabolisme , Intestins/immunologie , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme , Protéines de liaison à l'ARN/métabolisme , Complexe protéique du pore nucléaire/métabolisme
16.
Biochim Biophys Acta Mol Basis Dis ; 1870(8): 167489, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39233260

RÉSUMÉ

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.


Sujet(s)
Dysbiose , Microbiome gastro-intestinal , Macrophages , Récepteurs couplés aux protéines G , Récepteurs nicotiniques , Humains , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/génétique , Dysbiose/microbiologie , Dysbiose/métabolisme , Récepteurs nicotiniques/métabolisme , Récepteurs nicotiniques/génétique , Mâle , Macrophages/métabolisme , Macrophages/microbiologie , Femelle , Adulte , Adulte d'âge moyen , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/microbiologie , Maladies inflammatoires intestinales/anatomopathologie , Rectocolite hémorragique/microbiologie , Rectocolite hémorragique/métabolisme , Rectocolite hémorragique/anatomopathologie , Chaine alpha-1 du collagène de type I , Collagène de type I/métabolisme , Collagène de type I/génétique , Maladie de Crohn/microbiologie , Maladie de Crohn/métabolisme , Maladie de Crohn/anatomopathologie
17.
ACS Appl Mater Interfaces ; 16(38): 50212-50228, 2024 Sep 25.
Article de Anglais | MEDLINE | ID: mdl-39266250

RÉSUMÉ

Excessive reactive oxygen species (ROS) is a hallmark of both the onset and progression of inflammatory bowel disease (IBD), where a continuous cycle of ROS and inflammation drives the progression of diseases. The design of oral antioxidant nanoenzymes for scavenging ROS has emerged as a promising strategy to intervene in IBD. However, the practical application of these nanoenzymes is limited due to their single catalytical property and significantly impacted by substantial leakage in the upper gastrointestinal tract. This study introduces a novel oral delivery system, SP@CS-SeNPs, combining natural microalgae Spirulina platensis (SP), which possesses superoxide dismutase (SOD)-like activity, with chitosan-functionalized selenium nanoparticles (CS-SeNPs) that exhibit catalase-like activity. The SP@CS-SeNPs system leverages the dual catalytic capabilities of these components to initiate a cascade reaction that first converts superoxide anion radicals (O2•-) into hydrogen peroxide (H2O2), and then catalyzes the decomposition of H2O2 into water and oxygen. This system not only utilizes the resistance of the microalgae carrier to gastric acid and its efficient capture by intestinal villi, thereby enhancing intestinal distribution and retention but also demonstrates significant anti-inflammatory effects and effective repair of the damaged intestinal barrier in a colitis mice model. These results demonstrate that this oral delivery system successfully combines the features of microalgae and nanozymes, exhibits excellent biocompatibility, and offers a novel approach for antioxidant nanozyme intervention in IBD.


Sujet(s)
Chitosane , Maladies inflammatoires intestinales , Microalgues , Sélénium , Spirulina , Animaux , Souris , Sélénium/composition chimique , Sélénium/pharmacologie , Maladies inflammatoires intestinales/traitement médicamenteux , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/anatomopathologie , Spirulina/composition chimique , Microalgues/composition chimique , Administration par voie orale , Chitosane/composition chimique , Vecteurs de médicaments/composition chimique , Nanoparticules/composition chimique , Antioxydants/composition chimique , Antioxydants/pharmacologie , Humains , Espèces réactives de l'oxygène/métabolisme
18.
Front Immunol ; 15: 1405790, 2024.
Article de Anglais | MEDLINE | ID: mdl-39119343

RÉSUMÉ

Background: Previous observational studies have revealed the strong relationship between fatty acids (FA) and inflammatory bowel disease (IBD). Nonetheless, due to the inherent limitations of retrospective research, the causality between the two has not been clearly established. Methods: Genetic variants associated with the 17 FA indicators were derived from genome-wide association studies. Summary statistics for the discovery cohort and testing cohort for IBD, including ulcerative colitis (UC) and Crohn's disease (CD), were available from IIBDGC and FinnGen, respectively. Bidirectional MR analysis and sensitivity analysis with multiple measures were applied to comprehensively investigate the causal link between FA and IBD. Results: Combining the results of various MR methods, the validation of testing cohort, and the merging of meta-analysis, we demonstrated that genetically predicted Omega-3 FA levels, Ratio of Omega-3 FA to total FA, Docosahexaenoic acid (DHA) levels, and Ratio of DHA to total FA reduced the risk of IBD, UC, and CD. Meanwhile, multivariate MR suggested that the risk effects of Omega-3 FA and DHA for UC and CD were mainly affected by Saturated FA and Monounsaturated fatty acid (MUFA). Furthermore, although there was the causal association between Ratio of MUFA to total FA as well as Ratio of Polyunsaturated fatty acid (PUFA) to MUFA and CD, sensitivity analysis prompted that the findings were not robust. None of the above results had a reverse causal effect. Conclusion: This MR investigation provided evidence of causality between diverse FA and IBD. These findings offered new insights into the treatment and prevention of IBD.


Sujet(s)
Acides gras , Étude d'association pangénomique , Maladies inflammatoires intestinales , Humains , Acides gras/métabolisme , Maladies inflammatoires intestinales/étiologie , Maladies inflammatoires intestinales/métabolisme , Prédisposition génétique à une maladie , Rectocolite hémorragique/génétique , Rectocolite hémorragique/métabolisme , Rectocolite hémorragique/immunologie , Maladie de Crohn/génétique , Maladie de Crohn/étiologie , Polymorphisme de nucléotide simple , Femelle , Mâle , Acide docosahexaénoïque
19.
mBio ; 15(9): e0204324, 2024 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-39162488

RÉSUMÉ

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.


Sujet(s)
Microbiome gastro-intestinal , Maladies inflammatoires intestinales , Souris knockout , Facteur-6 associé aux récepteurs de TNF , Animaux , Humains , Souris , Colite/microbiologie , Colite/induit chimiquement , Colite/génétique , Colite/métabolisme , Modèles animaux de maladie humaine , Maladies inflammatoires intestinales/microbiologie , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/génétique , Maladies inflammatoires intestinales/immunologie , Muqueuse intestinale/métabolisme , Muqueuse intestinale/microbiologie , Muqueuse intestinale/immunologie , Souris de lignée C57BL , Stabilité protéique , Protéines SOCS/génétique , Protéines SOCS/métabolisme , Facteur-6 associé aux récepteurs de TNF/métabolisme , Facteur-6 associé aux récepteurs de TNF/génétique , Ubiquitination
20.
Front Immunol ; 15: 1423069, 2024.
Article de Anglais | MEDLINE | ID: mdl-39185411

RÉSUMÉ

Inflammatory bowel disease (IBD), a condition of the digestive tract and one of the autoimmune diseases, is becoming a disease of significant global public health concern and substantial clinical burden. Various signaling pathways have been documented to modulate IBD, but the exact activation and regulatory mechanisms have not been fully clarified; thus, a need for constant exploration of the molecules and pathways that play key roles in the development of IBD. In recent years, several protein post-translational modification pathways, such as ubiquitination, phosphorylation, methylation, acetylation, and glycolysis, have been implicated in IBD. An aberrant ubiquitination in IBD is often associated with dysregulated immune responses and inflammation. Mesenchymal stem cells (MSCs) play a crucial role in regulating ubiquitination modifications through the ubiquitin-proteasome system, a cellular machinery responsible for protein degradation. Specifically, MSCs have been shown to influence the ubiquitination of key signaling molecules involved in inflammatory pathways. This paper reviews the recent research progress in MSC-regulated ubiquitination in IBD, highlighting their therapeutic potential in treating IBD and offering a promising avenue for developing targeted interventions to modulate the immune system and alleviate inflammatory conditions.


Sujet(s)
Maladies inflammatoires intestinales , Cellules souches mésenchymateuses , Ubiquitination , Humains , Maladies inflammatoires intestinales/immunologie , Maladies inflammatoires intestinales/thérapie , Maladies inflammatoires intestinales/métabolisme , Cellules souches mésenchymateuses/immunologie , Cellules souches mésenchymateuses/métabolisme , Animaux , Transplantation de cellules souches mésenchymateuses , Transduction du signal , Maturation post-traductionnelle des protéines
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE