Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.068
Filtrer
1.
FASEB J ; 38(13): e23775, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38967223

RÉSUMÉ

Inflammatory bowel disease (IBD) is a chronic disease of the gastrointestinal tract affecting millions of people. Here, we investigated the expression and functions of poly(ADP-ribose) polymerase 14 (Parp14), an important regulatory protein in immune cells, with an IBD patient cohort as well as two mouse colitis models, that is, IBD-mimicking oral dextran sulfate sodium (DSS) exposure and oral Salmonella infection. Parp14 was expressed in the human colon by cells in the lamina propria, but, in particular, by the epithelial cells with a granular staining pattern in the cytosol. The same expression pattern was evidenced in both mouse models. Parp14-deficiency caused increased rectal bleeding as well as stronger epithelial erosion, Goblet cell loss, and immune cell infiltration in DSS-exposed mice. The absence of Parp14 did not affect the mouse colon bacterial microbiota. Also, the colon leukocyte populations of Parp14-deficient mice were normal. In contrast, bulk tissue RNA-Seq demonstrated that the colon transcriptomes of Parp14-deficient mice were dominated by abnormalities in inflammation and infection responses both prior and after the DSS exposure. Overall, the data indicate that Parp14 has an important role in the maintenance of colon epithelial barrier integrity. The prognostic and predictive biomarker potential of Parp14 in IBD merits further investigation.


Sujet(s)
Colite , Sulfate dextran , Souris de lignée C57BL , Poly(ADP-ribose) polymerases , Animaux , Femelle , Humains , Mâle , Souris , Colite/génétique , Colite/induit chimiquement , Colite/anatomopathologie , Côlon/anatomopathologie , Côlon/métabolisme , Sulfate dextran/toxicité , Modèles animaux de maladie humaine , Microbiome gastro-intestinal , Maladies inflammatoires intestinales/génétique , Maladies inflammatoires intestinales/anatomopathologie , Maladies inflammatoires intestinales/métabolisme , Souris knockout , Poly(ADP-ribose) polymerases/métabolisme , Poly(ADP-ribose) polymerases/génétique , Poly(ADP-ribose) polymerases/déficit
2.
Curr Protoc ; 4(7): e1092, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39007482

RÉSUMÉ

The intestinal inflammation induced by injection of naïve CD4+ T cells into lymphocyte-deficient hosts (more commonly known as the T cell transfer model of colitis) shares many features of idiopathic inflammatory bowel disease (IBD) in humans, such as epithelial cell hyperplasia, crypt abscess formation, and dense lamina propria lymphocyte infiltration. As such, it provides a useful tool for studying mucosal immune regulation as it relates to the pathogenesis and treatment of IBD in humans. In the IBD model described here, colitis is induced in Rag (recombination-activating gene)-deficient mice by reconstitution of these mice with naïve CD4+CD45RBhi T cells through adoptive T cell transfer. Although different recipient hosts of cell transfer can be used, Rag-deficient mice are the best characterized and support studies that are both flexible and reproduceable. As described in the Basic Protocol, in most studies the transferred cells consist of naïve CD4+ T cells (CD45RBhi T cells) derived by fluorescence-activated cell sorting from total CD4+ T cells previously purified using immunomagnetic negative selection beads. In a Support Protocol, methods to characterize colonic disease progression are described, including the monitoring of weight loss and diarrhea and the histological assessment of colon pathology. © 2024 The Author(s). Current Protocols published by Wiley Periodicals LLC. Basic Protocol: Induction of IBD in Rag-deficient mice by the transfer of naïve CD4+CD45RBhi T cells Support Protocol: Monitoring development of colitis.


Sujet(s)
Lymphocytes T CD4+ , Modèles animaux de maladie humaine , Maladies inflammatoires intestinales , Animaux , Souris , Lymphocytes T CD4+/immunologie , Maladies inflammatoires intestinales/immunologie , Maladies inflammatoires intestinales/anatomopathologie , Colite/immunologie , Colite/induit chimiquement , Colite/anatomopathologie , Transfert adoptif
3.
Stem Cell Res Ther ; 15(1): 190, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956621

RÉSUMÉ

BACKGROUND: Mesenchymal stem cells (MSCs) demonstrate a wide range of therapeutic capabilities in the treatment of inflammatory bowel disease (IBD). The intraperitoneal injection of MSCs has exhibited superior therapeutic efficacy on IBD than intravenous injection. Nevertheless, the precise in vivo distribution of MSCs and their biological consequences following intraperitoneal injection remain inadequately understood. Additional studies are required to explore the correlation between MSCs distribution and their biological effects. METHODS: First, the distribution of human umbilical cord MSCs (hUC-MSCs) and the numbers of Treg and Th17 cells in mesenteric lymph nodes (MLNs) were analyzed after intraperitoneal injection of hUC-MSCs. Subsequently, the investigation focused on the levels of transforming growth factor beta1 (TGF-ß1), a key cytokine to the biology of both Treg and Th17 cells, in tissues of mice with colitis, particularly in MLNs. The study also delved into the impact of hUC-MSCs therapy on Treg cell counts in MLNs, as well as the consequence of TGFB1 knockdown hUC-MSCs on the differentiation of Treg cells and the treatment of IBD. RESULTS: The therapeutic effectiveness of intraperitoneally administered hUC-MSCs in the treatment of colitis was found to be significant, which was closely related to their quick migration to MLNs and secretion of TGF-ß1. The abundance of hUC-MSCs in MLNs of colitis mice is much higher than that in other organs even the inflamed sites of colon. Intraperitoneal injection of hUC-MSCs led to a significant increase in the number of Treg cells and a decrease in Th17 cells especially in MLNs. Furthermore, the concentration of TGF-ß1, the key cytokine for Treg differentiation, were also found to be significantly elevated in MLNs after hUC-MSCs treatment. Knockdown of TGFB1 in hUC-MSCs resulted in a noticeable reduction of Treg cells in MLNs and the eventually failure of hUC-MSCs therapy in colitis. CONCLUSIONS: MLNs may be a critical site for the regulatory effect of hUC-MSCs on Treg/Th17 cells and the therapeutic effect on colitis. TGF-ß1 derived from hUC-MSCs promotes local Treg differentiation in MLNs. This study will provide new ideas for the development of MSC-based therapeutic strategies in IBD patients.


Sujet(s)
Différenciation cellulaire , Colite , Noeuds lymphatiques , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Lymphocytes T régulateurs , Cellules Th17 , Facteur de croissance transformant bêta-1 , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Facteur de croissance transformant bêta-1/métabolisme , Animaux , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Humains , Colite/thérapie , Colite/induit chimiquement , Colite/métabolisme , Colite/anatomopathologie , Transplantation de cellules souches mésenchymateuses/méthodes , Souris , Noeuds lymphatiques/métabolisme , Cellules Th17/métabolisme , Cellules Th17/immunologie , Cordon ombilical/cytologie , Mésentère/métabolisme , Souris de lignée C57BL , Souris de lignée BALB C , Mâle , Maladies inflammatoires intestinales/thérapie , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/anatomopathologie
4.
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
5.
World J Gastroenterol ; 30(24): 3022-3035, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38983953

RÉSUMÉ

Managing inflammatory bowel disease (IBD) is becoming increasingly complex and personalized, considering the advent of new advanced therapies with distinct mechanisms of action. Achieving mucosal healing (MH) is a pivotal therapeutic goal in IBD management and can prevent IBD progression and reduce flares, hospitalization, surgery, intestinal damage, and colorectal cancer. Employing proactive disease and therapy assessment is essential to achieve better control of intestinal inflammation, even if subclinical, to alter the natural course of IBD. Periodic monitoring of fecal calprotectin (FC) levels and interval endoscopic evaluations are cornerstones for evaluating response/remission to advanced therapies targeting IBD, assessing MH, and detecting subclinical recurrence. Here, we comment on the article by Ishida et al Moreover, this editorial aimed to review the role of FC and endoscopic scores in predicting MH in patients with IBD. Furthermore, we intend to present some evidence on the role of these markers in future targets, such as histological and transmural healing. Additional prospective multicenter studies with a stricter MH criterion, standardized endoscopic and histopathological analyses, and virtual chromoscopy, potentially including artificial intelligence and other biomarkers, are desired.


Sujet(s)
Marqueurs biologiques , Fèces , Maladies inflammatoires intestinales , Muqueuse intestinale , Complexe antigénique L1 leucocytaire , Humains , Complexe antigénique L1 leucocytaire/analyse , Fèces/composition chimique , Muqueuse intestinale/anatomopathologie , Muqueuse intestinale/métabolisme , Marqueurs biologiques/analyse , Marqueurs biologiques/métabolisme , Maladies inflammatoires intestinales/anatomopathologie , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/thérapie , Indice de gravité de la maladie , Cicatrisation de plaie , Coloscopie , Évolution de la maladie , Récidive , Endoscopie gastrointestinale/méthodes
6.
BMC Med Genomics ; 17(1): 187, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-39014344

RÉSUMÉ

BACKGROUND: With recent advances in gene sequencing technology, more than 60 genetic mutations associated with very early onset inflammatory bowel disease (VEO-IBD) have been reported. Most of the genes are associated with immune deficiencies. The Myosin 5B (MYO5B) gene is primarily involved in cell motility and material transport which is associated with congenital intractable diarrhea and cholestasis. No studies have examined the relationship between the MYO5B gene and VEO-IBD. We report a case of a child with a mutation in the MYO5B gene who was diagnosed with VEO-IBD, then we investigated the association between the MYO5B gene and VEO-IBD. CASE PRESENTATION: A 7-month-old baby girl with a chief complaint of "blood in the stool for more than 4 months and vaginal pus and blood discharge for 3 weeks" was diagnosed with VEO-IBD, and her symptoms improved after treatment with mesalazine. The whole-exome sequencing was performed with peripheral blood. Immunohistochemistry was performed on the terminal ileal tissue. Western blotting, quantitative polymerase chain reaction (Q-PCR) and immunofluorescence were performed with cultured organoid tissue from the terminal ileum. Whole-exome sequencing identified heterozygous missense of MYO5B variant of unknown significance (p. [I769N]; [T1546M]). Immunohistochemistry revealed a significant decrease in the expression of MYO5B protein in the terminal ileum of the child with MYO5B mutation; Q-PCR revealed a decrease in the mRNA levels of occludin and ZO-1 and both the mRNA levels and protein levels of MYO5B was downregulated in the patient. Immunofluorescence images showed that MYO5B gene mutation disrupted the apical delivery of transporters SGLT1, NHE3 and AQP7. CONCLUSIONS: MYO5B gene mutation leading to the downregulation of MYO5B protein may promote the occurrence of VEO-IBD by decreasing mRNA and protein levels of intestinal tight junction genes and dislocating the apical transporters.


Sujet(s)
Maladies inflammatoires intestinales , Mutation , Myosine de type V , Humains , Femelle , Maladies inflammatoires intestinales/génétique , Maladies inflammatoires intestinales/anatomopathologie , Myosine de type V/génétique , Nourrisson , Âge de début , , Chaînes lourdes de myosine
7.
Sci Data ; 11(1): 761, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38992012

RÉSUMÉ

The incidence of inflammatory bowel disease (IBD) is increasing annually. Children with IBD often suffer significant morbidity due to physical and emotional effects of the disease and treatment. Corticosteroids, often a component of therapy, carry undesirable side effects with long term use. Steroid-free remission has become a standard for care-quality improvement. Anticipating therapeutic outcomes is difficult, with treatments often leveraged in a trial-and-error fashion. Artificial intelligence (AI) has demonstrated success in medical imaging classification tasks. Predicting patients who will attain remission will help inform treatment decisions. The provided dataset comprises 951 tissue section scans (167 whole-slides) obtained from 18 pediatric IBD patients. Patient level structured data include IBD diagnosis, 12- and 52-week steroid use and name, and remission status. Each slide is labelled with biopsy site and normal or abnormal classification per the surgical pathology report. Each tissue section scan from an abnormal slide is further classified by an experienced pathologist. Researchers utilizing this dataset may select from the provided outcomes or add labels and annotations from their own institutions.


Sujet(s)
Maladies inflammatoires intestinales , Humains , Enfant , Maladies inflammatoires intestinales/imagerie diagnostique , Maladies inflammatoires intestinales/anatomopathologie , Adolescent , Induction de rémission , Intelligence artificielle
8.
ACS Appl Mater Interfaces ; 16(26): 33081-33092, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38888094

RÉSUMÉ

Inflammatory bowel disease (IBD) is a chronic and recurrent inflammatory disease that affects the gastrointestinal tract. The major hurdles impeding IBD treatment are the low targeting efficiency and short retention time of drugs in IBD sites. Nanoparticles with specific shapes have demonstrated the ability to improve mucus retention and cellular uptake. Herein, mesoporous silica nanoparticles (MSNs) with various morphologies were used to deliver budesonide (BUD) for the treatment of IBD. The therapeutic efficacy is strongly dependent on their shapes. The system comprises different shapes of MSNs as carriers for budesonide (BUD), along with Eudragit S100 as the enteric release shell. The encapsulation of Eudragit S100 not only improved the stability of MSNs-BUD in the gastrointestinal tract but also conferred pH-responsive drug release properties. Then, MSNs efficiently deliver BUD to the colon site, and the special shape of MSNs plays a critical role in enhancing their permeability and retention in the mucus layer. Among them, dendritic MSNs (MSND) effectively reduced myeloperoxidase (MPO) activity and levels of inflammatory cytokines in the colon due to long retention time and rapid release in IBD sites, thereby enhancing the therapeutic efficacy against colitis. Given the special shapes of MSNs and pH-responsivity of Eudragit S100, BUD loaded in the voids of MSND (E@MSNs-BUD) could penetrate the mucous layer and be accurately delivered to the colon with minor side effects. This system is expected to complement current treatment strategies for the IBD.


Sujet(s)
Budésonide , Vecteurs de médicaments , Maladies inflammatoires intestinales , Nanoparticules , Silice , Budésonide/composition chimique , Budésonide/administration et posologie , Budésonide/usage thérapeutique , Budésonide/pharmacocinétique , Nanoparticules/composition chimique , Nanoparticules/usage thérapeutique , Animaux , Silice/composition chimique , Maladies inflammatoires intestinales/traitement médicamenteux , Maladies inflammatoires intestinales/anatomopathologie , Vecteurs de médicaments/composition chimique , Souris , Poly(acides méthacryliques)/composition chimique , Libération de médicament , Humains , Anti-inflammatoires/composition chimique , Anti-inflammatoires/usage thérapeutique , Anti-inflammatoires/administration et posologie , Porosité , Concentration en ions d'hydrogène
9.
Int J Mol Sci ; 25(12)2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38928122

RÉSUMÉ

Inflammatory Bowel Disease (IBD) is a complex and challenging health problem that exerts a significant impact on the quality of life of millions of individuals worldwide [...].


Sujet(s)
Fibrose , Maladies inflammatoires intestinales , Humains , Maladies inflammatoires intestinales/immunologie , Maladies inflammatoires intestinales/thérapie , Maladies inflammatoires intestinales/anatomopathologie , Animaux
10.
Int J Mol Sci ; 25(12)2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38928243

RÉSUMÉ

Creatine transporter (CrT1) mediates cellular uptake of creatine (Cr), a nutrient pivotal in maintaining energy homeostasis in various tissues including intestinal epithelial cells (IECs). The impact of CrT1 deficiency on the pathogenesis of various psychiatric and neurological disorders has been extensively investigated. However, there are no studies on its regulation in IECs in health and disease. Current studies have determined differential expression of CrT1 along the length of the mammalian intestine and its dysregulation in inflammatory bowel disease (IBD)-associated inflammation and Adherent Invasive E. coli (AIEC) infection. CrT1 mRNA and protein levels in normal intestines and their alterations in inflammation and following AIEC infection were determined in vitro in model IECs (Caco-2/IEC-6) and in vivo in SAMP1/YitFc mice, a model of spontaneous ileitis resembling human IBD. CrT1 is differentially expressed in different regions of mammalian intestines with its highest expression in jejunum. In vitro, CrT1 function (Na+-dependent 14C-Cr uptake), expression and promoter activity significantly decreased following TNFα/IL1ß treatments and AIEC infection. SAMP1 mice and ileal organoids generated from SAMP1 mice also showed decreased CrT1 mRNA and protein compared to AKR controls. Our studies suggest that Cr deficiency in IECs secondary to CrT1 dysregulation could be a key factor contributing to IBD pathogenesis.


Sujet(s)
Infections à Escherichia coli , Muqueuse intestinale , Animaux , Infections à Escherichia coli/métabolisme , Infections à Escherichia coli/microbiologie , Souris , Humains , Muqueuse intestinale/métabolisme , Muqueuse intestinale/microbiologie , Muqueuse intestinale/anatomopathologie , Cellules Caco-2 , Transporteurs plasmiques de neurotransmetteurs/métabolisme , Transporteurs plasmiques de neurotransmetteurs/génétique , Transporteurs plasmiques de neurotransmetteurs/déficit , Inflammation/métabolisme , Inflammation/génétique , Inflammation/anatomopathologie , Escherichia coli , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/génétique , Maladies inflammatoires intestinales/anatomopathologie , Protéines de transport membranaire/métabolisme , Protéines de transport membranaire/génétique , Cellules épithéliales/métabolisme , Cellules épithéliales/microbiologie , Créatine/métabolisme
11.
Int J Biol Macromol ; 273(Pt 2): 132909, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38848832

RÉSUMÉ

The pathological changes in inflammatory bowel disease (IBD) include the disruption of intestinal barrier function and the infiltration of pathogenic microbes. The application of an artificial protective barrier at the site of inflammation can prevent bacterial infiltration, promote epithelial cell migration, and accelerate wound healing. In this study, dopamine-modified hyaluronic acid (HA-DA) was developed as a bioadhesive self-cross-linkable hydrogel, which acted as an enteroprotective agent to promote the healing of inflamed intestinal tissue. The adhesion strength HA-DA to mouse colon was 3.81-fold higher than HA. Moreover, HA-DA promoted Caco-2 cell proliferation and migration as well as had a strong physical barrier effect after gelation. After oral administration, the HA-DA reduced weight loss and attenuated impaired goblet cell function in mice with dextran sodium sulfate-induced IBD. In addition, HA-DA promoted restoration of the epithelial barrier by the upregulation of tight junction proteins. The results reported herein substantiated that self-cross-linkable hydrogel-based enteroprotective agents are a promising approach for the treatment of IBD.


Sujet(s)
Acide hyaluronique , Hydrogels , Maladies inflammatoires intestinales , Acide hyaluronique/composition chimique , Acide hyaluronique/pharmacologie , Animaux , Humains , Maladies inflammatoires intestinales/traitement médicamenteux , Maladies inflammatoires intestinales/anatomopathologie , Souris , Cellules Caco-2 , Hydrogels/composition chimique , Hydrogels/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Agents protecteurs/pharmacologie , Agents protecteurs/composition chimique , Mouvement cellulaire/effets des médicaments et des substances chimiques , Mâle , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/métabolisme , Sulfate dextran
12.
Cell Rep ; 43(6): 114311, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38848214

RÉSUMÉ

The lymphatic fluid is the conduit by which part of the tissue "omics" is transported to the draining lymph node for immunosurveillance. Following cannulation of the pre-nodal cervical and mesenteric afferent lymphatics, herein we investigate the lymph proteomic composition, uncovering that its composition varies according to the tissue of origin. Tissue specificity is also reflected in the dendritic cell-major histocompatibility complex class II-eluted immunopeptidome harvested from the cervical and mesenteric nodes. Following inflammatory disruption of the gut barrier, the lymph antigenic and inflammatory loads are analyzed in both mice and subjects with inflammatory bowel diseases. Gastrointestinal tissue damage reflects the lymph inflammatory and damage-associated molecular pattern signatures, microbiome-derived by-products, and immunomodulatory molecules, including metabolites of the gut-brain axis, mapped in the afferent mesenteric lymph. Our data point to the relevance of the lymphatic fluid to probe the tissue-specific antigenic and inflammatory load transported to the draining lymph node for immunosurveillance.


Sujet(s)
Antigènes , Inflammation , Noeuds lymphatiques , Lymphe , Souris de lignée C57BL , Animaux , Souris , Lymphe/métabolisme , Lymphe/immunologie , Inflammation/immunologie , Inflammation/anatomopathologie , Inflammation/métabolisme , Noeuds lymphatiques/immunologie , Noeuds lymphatiques/métabolisme , Humains , Antigènes/métabolisme , Antigènes/immunologie , Mâle , Femelle , Maladies inflammatoires intestinales/immunologie , Maladies inflammatoires intestinales/anatomopathologie , Maladies inflammatoires intestinales/métabolisme , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme
13.
Int J Med Sci ; 21(8): 1385-1398, 2024.
Article de Anglais | MEDLINE | ID: mdl-38903915

RÉSUMÉ

Inflammatory bowel disease (IBD) is a chronic inflammatory intestinal disease, characterized by dysregulated immune response. HDAC3 is reported to be an epigenetic brake in inflammation, playing critical roles in macrophages. However, its role in IBD is unclear. In our study, we found HDAC3 was upregulated in CX3CR1-positive cells in the mucosa from IBD mice. Conditional knockout (cKO) of Hdac3 in CX3CR1 positive cells attenuated the disease severity of Dextran Sulfate Sodium (DSS)-induced colitis. In addition, inhibition of HDAC3 with RGFP966 could also alleviate the DSS-induced tissue injury and inflammation in IBD. The RNA sequencing results revealed that Hdac3 cKO restrained DSS-induced upregulation of genes in the pathways of cytokine-cytokine receptor interaction, complement and coagulation cascades, chemokine signaling, and extracellular matrix receptor interaction. We also identified that Guanylate-Binding Protein 5 (GBP5) was transcriptionally regulated by HDAC3 in monocytes by RNA sequencing. Inhibition of HDAC3 resulted in decreased transcriptional activity of interferon-gamma-induced expression of GBP5 in CX3CR1-positive cells, such as macrophages and microglia. Overexpression of HDAC3 upregulated the transcriptional activity of GBP5 reporter. Lastly, conditional knockout of Hdac3 in macrophages (Hdac3 mKO) attenuated the disease severity of DSS-induced colitis. In conclusion, inhibition of HDAC3 in macrophages could ameliorate the disease severity and inflammatory response in colitis by regulating GBP5-NLRP3 axis, identifying a new therapeutic avenue for the treatment of colitis.


Sujet(s)
Colite , Sulfate dextran , Histone deacetylases , Macrophages , Souris knockout , Protéine-3 de la famille des NLR contenant un domaine pyrine , Transduction du signal , Animaux , Sulfate dextran/toxicité , Sulfate dextran/effets indésirables , Histone deacetylases/métabolisme , Histone deacetylases/génétique , Souris , Macrophages/métabolisme , Macrophages/immunologie , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Colite/induit chimiquement , Colite/génétique , Colite/anatomopathologie , Colite/métabolisme , Humains , Transduction du signal/effets des médicaments et des substances chimiques , Maladies inflammatoires intestinales/induit chimiquement , Maladies inflammatoires intestinales/génétique , Maladies inflammatoires intestinales/anatomopathologie , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/traitement médicamenteux , Protéines G/génétique , Protéines G/métabolisme , Protéines G/antagonistes et inhibiteurs , Modèles animaux de maladie humaine , Récepteur-1 de la chimiokine CX3C/métabolisme , Récepteur-1 de la chimiokine CX3C/génétique , Souris de lignée C57BL , Inhibiteurs de désacétylase d'histone/pharmacologie , Inhibiteurs de désacétylase d'histone/usage thérapeutique , Muqueuse intestinale/anatomopathologie , Muqueuse intestinale/métabolisme , Muqueuse intestinale/effets des médicaments et des substances chimiques , Acrylamides , Phénylènediamines
14.
Int J Mol Sci ; 25(11)2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38891786

RÉSUMÉ

Inflammatory bowel disease (IBD) is a nonspecific chronic inflammatory disease resulting from an immune disorder in the intestine that is prone to relapse and incurable. The understanding of the pathogenesis of IBD remains unclear. In this study, we found that ace (angiotensin-converting enzyme), expressed abundantly in the intestine, plays an important role in IBD. The deletion of ace in zebrafish caused intestinal inflammation with increased expression of the inflammatory marker genes interleukin 1 beta (il1b), matrix metallopeptidase 9 (mmp9), myeloid-specific peroxidase (mpx), leukocyte cell-derived chemotaxin-2-like (lect2l), and chemokine (C-X-C motif) ligand 8b (cxcl8b). Moreover, the secretion of mucus in the ace-/- mutants was significantly higher than that in the wild-type zebrafish, validating the phenotype of intestinal inflammation. This was further confirmed by the IBD model constructed using dextran sodium sulfate (DSS), in which the mutant zebrafish had a higher susceptibility to enteritis. Our study reveals the role of ace in intestinal homeostasis, providing a new target for potential therapeutic interventions.


Sujet(s)
Peptidyl-Dipeptidase A , Danio zébré , Animaux , Danio zébré/génétique , Peptidyl-Dipeptidase A/génétique , Peptidyl-Dipeptidase A/métabolisme , Modèles animaux de maladie humaine , Sulfate dextran , Inflammation/génétique , Inflammation/métabolisme , Inflammation/anatomopathologie , Protéines de poisson-zèbre/génétique , Protéines de poisson-zèbre/métabolisme , Intestins/anatomopathologie , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/génétique , Maladies inflammatoires intestinales/anatomopathologie , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/génétique , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie
15.
Int J Mol Sci ; 25(11)2024 May 27.
Article de Anglais | MEDLINE | ID: mdl-38892004

RÉSUMÉ

Vedolizumab (VDZ) is used for treating inflammatory bowel disease (IBD) patients. A study investigating colonic epithelial barrier function ex vivo following VDZ is lacking. This work aims to evaluate ex vivo the colonic epithelial barrier function in IBD patients at baseline and during VDZ treatment, and to investigate the relationships between barrier function and clinical parameters. Colonic specimens were obtained from 23 IBD patients before, and at 24 and 52 weeks after VDZ treatment, and from 26 healthy volunteers (HV). Transepithelial electrical resistance (TEER, permeability to ions) and paracellular permeability were measured in Ussing chambers. IBD patients showed increased epithelial permeability to ions (TEER, 13.80 ± 1.04 Ω × cm2 vs. HV 20.70 ± 1.52 Ω × cm2, p < 0.001) without changes in paracellular permeability of a 4 kDa probe. VDZ increased TEER (18.09 ± 1.44 Ω × cm2, p < 0.001) after 52 weeks. A clinical response was observed in 58% and 25% of patients at week 24, and in 62% and 50% at week 52, in ulcerative colitis and Crohn's disease, respectively. Clinical and endoscopic scores were strongly associated with TEER. TEER < 14.65 Ω × cm2 predicted response to VDZ (OR 11; CI 2-59). VDZ reduces the increased permeability to ions observed in the colonic epithelium of IBD patients before treatment, in parallel to a clinical, histological (inflammatory infiltrate), and endoscopic improvement. A low TEER predicts clinical response to VDZ therapy.


Sujet(s)
Anticorps monoclonaux humanisés , Côlon , Maladies inflammatoires intestinales , Muqueuse intestinale , Perméabilité , Humains , Anticorps monoclonaux humanisés/pharmacologie , Anticorps monoclonaux humanisés/usage thérapeutique , Mâle , Femelle , Adulte , Adulte d'âge moyen , Perméabilité/effets des médicaments et des substances chimiques , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Maladies inflammatoires intestinales/traitement médicamenteux , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/anatomopathologie , Côlon/effets des médicaments et des substances chimiques , Côlon/métabolisme , Côlon/anatomopathologie , Ions/métabolisme , Agents gastro-intestinaux/pharmacologie , Agents gastro-intestinaux/usage thérapeutique , Impédance électrique , Rectocolite hémorragique/traitement médicamenteux , Rectocolite hémorragique/métabolisme , Rectocolite hémorragique/anatomopathologie , Maladie de Crohn/traitement médicamenteux , Maladie de Crohn/métabolisme , Maladie de Crohn/anatomopathologie , Sujet âgé
16.
Int J Mol Sci ; 25(11)2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38892399

RÉSUMÉ

Tuft cells are more than guardian chemosensory elements of the digestive tract. They produce a variety of immunological effector molecules in response to stimulation; moreover, they are essential for defense against protozoa and nematodes. Beyond the description of their characteristics, this review aims to elucidate the potential pathogenic and therapeutic roles of colonic tuft cells in inflammatory bowel disease and colorectal cancer, focusing on their primarily immunomodulatory action. Regarding inflammatory bowel disease, tuft cells are implicated in both maintaining the integrity of the intestinal epithelial barrier and in tissue repair and regeneration processes. In addition to maintaining intestinal homeostasis, they display complex immune-regulatory functions. During the development of colorectal cancer, tuft cells can promote the epithelial-to-mesenchymal transition, alter the gastrointestinal microenvironment, and modulate both the anti-tumor immune response and the tumor microenvironment. A wide variety of their biological functions can be targeted for anti-inflammatory or anti-tumor therapies; however, the adverse side effects of immunomodulatory actions must be strictly considered.


Sujet(s)
Tumeurs colorectales , Maladies inflammatoires intestinales , Microenvironnement tumoral , Humains , Tumeurs colorectales/immunologie , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Tumeurs colorectales/thérapie , Maladies inflammatoires intestinales/immunologie , Maladies inflammatoires intestinales/thérapie , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/anatomopathologie , Animaux , Microenvironnement tumoral/immunologie , Muqueuse intestinale/immunologie , Muqueuse intestinale/métabolisme , Transition épithélio-mésenchymateuse , Côlon/immunologie , Côlon/anatomopathologie , Côlon/métabolisme ,
17.
Nat Commun ; 15(1): 4775, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38839750

RÉSUMÉ

The metal ion transporter SLC39A8 is associated with physiological traits and diseases, including blood manganese (Mn) levels and inflammatory bowel diseases (IBD). The mechanisms by which SLC39A8 controls Mn homeostasis and epithelial integrity remain elusive. Here, we generate Slc39a8 intestinal epithelial cell-specific-knockout (Slc39a8-IEC KO) mice, which display markedly decreased Mn levels in blood and most organs. Radiotracer studies reveal impaired intestinal absorption of dietary Mn in Slc39a8-IEC KO mice. SLC39A8 is localized to the apical membrane and mediates 54Mn uptake in intestinal organoid monolayer cultures. Unbiased transcriptomic analysis identifies alkaline ceramidase 1 (ACER1), a key enzyme in sphingolipid metabolism, as a potential therapeutic target for SLC39A8-associated IBDs. Importantly, treatment with an ACER1 inhibitor attenuates colitis in Slc39a8-IEC KO mice by remedying barrier dysfunction. Our results highlight the essential roles of SLC39A8 in intestinal Mn absorption and epithelial integrity and offer a therapeutic target for IBD associated with impaired Mn homeostasis.


Sujet(s)
Alkaline Ceramidase , Transporteurs de cations , Maladies inflammatoires intestinales , Muqueuse intestinale , Manganèse , Souris knockout , Animaux , Transporteurs de cations/métabolisme , Transporteurs de cations/génétique , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/génétique , Maladies inflammatoires intestinales/anatomopathologie , Manganèse/métabolisme , Souris , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Alkaline Ceramidase/métabolisme , Alkaline Ceramidase/génétique , Humains , Souris de lignée C57BL , Homéostasie , Mâle , Colite/métabolisme , Colite/génétique , Colite/anatomopathologie , Absorption intestinale , Cellules épithéliales/métabolisme
18.
Eur J Med Res ; 29(1): 335, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38890719

RÉSUMÉ

Intestinal fibrosis is a prevalent complication of IBD that that can frequently be triggered by prolonged inflammation. Fibrosis in the gut can cause a number of issues, which continue as an ongoing challenge to healthcare systems worldwide. The primary causes of intestinal fibrosis are soluble molecules, G protein-coupled receptors, epithelial-to-mesenchymal or endothelial-to-mesenchymal transition, and the gut microbiota. Fresh perspectives coming from in vivo and in vitro experimental models demonstrate that fibrogenic pathways might be different, at least to some extent, independent of the ones that influence inflammation. Understanding the distinctive procedures of intestinal fibrogenesis should provide a realistic foundation for targeting and blocking specific fibrogenic pathways, estimating the risk of fibrotic consequences, detecting early fibrotic alterations, and eventually allowing therapy development. Here, we first summarize the inflammatory and non-inflammatory components of fibrosis, and then we elaborate on the underlying mechanism associated with multiple cytokines in fibrosis, providing the framework for future clinical practice. Following that, we discuss the relationship between modernization and disease, as well as the shortcomings of current studies. We outline fibrosis diagnosis and therapy, as well as our recommendations for the future treatment of intestinal fibrosis. We anticipate that the global review will provides a wealth of fresh knowledge and suggestions for future fibrosis clinical practice.


Sujet(s)
Fibrose , Inflammation , Humains , Inflammation/anatomopathologie , Maladies inflammatoires intestinales/complications , Maladies inflammatoires intestinales/anatomopathologie , Intestins/anatomopathologie , Animaux , Microbiome gastro-intestinal , Transition épithélio-mésenchymateuse , Cytokines/métabolisme
19.
Nat Genet ; 56(7): 1456-1467, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38902475

RÉSUMÉ

According to conventional views, colon cancer originates from stem cells. However, inflammation, a key risk factor for colon cancer, has been shown to suppress intestinal stemness. Here, we used Paneth cells as a model to assess the capacity of differentiated lineages to trigger tumorigenesis in the context of inflammation in mice. Upon inflammation, Paneth cell-specific Apc mutations led to intestinal tumors reminiscent not only of those arising in patients with inflammatory bowel disease, but also of a larger fraction of human sporadic colon cancers. The latter is possibly because of the inflammatory consequences of western-style dietary habits, a major colon cancer risk factor. Machine learning methods designed to predict the cell-of-origin of cancer from patient-derived tumor samples confirmed that, in a substantial fraction of sporadic cases, the origins of colon cancer reside in secretory lineages and not in stem cells.


Sujet(s)
Carcinogenèse , Lignage cellulaire , Tumeurs du côlon , Inflammation , Cellules de Paneth , Animaux , Souris , Lignage cellulaire/génétique , Cellules de Paneth/anatomopathologie , Humains , Inflammation/génétique , Inflammation/anatomopathologie , Carcinogenèse/génétique , Tumeurs du côlon/génétique , Tumeurs du côlon/anatomopathologie , Mutation , Cellules souches/anatomopathologie , Différenciation cellulaire/génétique , Transformation cellulaire néoplasique/génétique , Protéine de la polypose adénomateuse colique/génétique , Souris de lignée C57BL , Maladies inflammatoires intestinales/génétique , Maladies inflammatoires intestinales/anatomopathologie
20.
Lancet Gastroenterol Hepatol ; 9(8): 758-772, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38759661

RÉSUMÉ

Integrating artificial intelligence into inflammatory bowel disease (IBD) has the potential to revolutionise clinical practice and research. Artificial intelligence harnesses advanced algorithms to deliver accurate assessments of IBD endoscopy and histology, offering precise evaluations of disease activity, standardised scoring, and outcome prediction. Furthermore, artificial intelligence offers the potential for a holistic endo-histo-omics approach by interlacing and harmonising endoscopy, histology, and omics data towards precision medicine. The emerging applications of artificial intelligence could pave the way for personalised medicine in IBD, offering patient stratification for the most beneficial therapy with minimal risk. Although artificial intelligence holds promise, challenges remain, including data quality, standardisation, reproducibility, scarcity of randomised controlled trials, clinical implementation, ethical concerns, legal liability, and regulatory issues. The development of standardised guidelines and interdisciplinary collaboration, including policy makers and regulatory agencies, is crucial for addressing these challenges and advancing artificial intelligence in IBD clinical practice and trials.


Sujet(s)
Intelligence artificielle , Maladies inflammatoires intestinales , Médecine de précision , Humains , Maladies inflammatoires intestinales/anatomopathologie , Médecine de précision/méthodes , Endoscopie gastrointestinale/méthodes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE