Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 5.472
Filtrer
1.
Front Immunol ; 15: 1435180, 2024.
Article de Anglais | MEDLINE | ID: mdl-39114658

RÉSUMÉ

Introduction: Introduction: The influenza virus primarily targets the respiratory tract, yet both the respiratory and intestinal systems suffer damage during infection. The connection between lung and intestinal damage remains unclear. Methods: Our experiment employs 16S rRNA technology and Liquid Chromatography-Mass Spectrometry (LC-MS) to detect the impact of influenza virus infection on the fecal content and metabolites in mice. Additionally, it investigates the effect of influenza virus infection on intestinal damage and its underlying mechanisms through HE staining, Western blot, Q-PCR, and flow cytometry. Results: Our study found that influenza virus infection caused significant damage to both the lungs and intestines, with the virus detected exclusively in the lungs. Antibiotic treatment worsened the severity of lung and intestinal damage. Moreover, mRNA levels of Toll-like receptor 7 (TLR7) and Interferon-b (IFN-b) significantly increased in the lungs post-infection. Analysis of intestinal microbiota revealed notable shifts in composition after influenza infection, including increased Enterobacteriaceae and decreased Lactobacillaceae. Conversely, antibiotic treatment reduced microbial diversity, notably affecting Firmicutes, Proteobacteria, and Bacteroidetes. Metabolomics showed altered amino acid metabolism pathways due to influenza infection and antibiotics. Abnormal expression of indoleamine 2,3-dioxygenase 1 (IDO1) in the colon disrupted the balance between helper T17 cells (Th17) and regulatory T cells (Treg cells) in the intestine. Mice infected with the influenza virus and supplemented with tryptophan and Lactobacillus showed reduced lung and intestinal damage, decreased Enterobacteriaceae levels in the intestine, and decreased IDO1 activity. Discussion: Overall, influenza infection caused damage to lung and intestinal tissues, disrupted intestinal microbiota and metabolites, and affected Th17/Treg balance. Antibiotic treatment exacerbated these effects. Supplementation with tryptophan and Lactobacillus improved lung and intestinal health, highlighting a new understanding of the lung-intestine connection in influenza-induced intestinal disease.


Sujet(s)
Modèles animaux de maladie humaine , Microbiome gastro-intestinal , Poumon , Infections à Orthomyxoviridae , Animaux , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/métabolisme , Souris , Poumon/immunologie , Poumon/microbiologie , Poumon/métabolisme , Poumon/virologie , Récepteur de type Toll-7/métabolisme , Indoleamine-pyrrole 2,3,-dioxygenase/métabolisme , Souris de lignée C57BL , Intestins/immunologie , Intestins/microbiologie , Intestins/virologie , Femelle , Antibactériens/pharmacologie , Antibactériens/usage thérapeutique , Transduction du signal , ARN ribosomique 16S/génétique , Glycoprotéines membranaires
2.
Front Immunol ; 15: 1414869, 2024.
Article de Anglais | MEDLINE | ID: mdl-39100674

RÉSUMÉ

Introduction: The prevention and mitigation of intestinal immune challenge is crucial for poultry production. This study investigated the effects of dietary Macleaya cordata extract (MCE) supplementation on the prevention of intestinal injury in broiler chickens challenged with lipopolysaccharide (LPS). Methods: A total of 256 one-day-old male Arbor Acres broilers were randomly divided into 4 treatment groups using a 2×2 factorial design with 2 MCE supplemental levels (0 and 400 mg/kg) and 2 LPS challenge levels (0 and 1 mg/kg body weight). The experiment lasted for 21 d. Results and discussion: The results showed that MCE supplementation increased the average daily feed intake during days 0-14. MCE supplementation and LPS challenge have an interaction on the average daily gain during days 15-21. MCE supplementation significantly alleviated the decreased average daily gain of broiler chickens induced by LPS. MCE supplementation increased the total antioxidant capacity and the activity of catalase and reduced the level of malondialdehyde in jejunal mucosa. MCE addition elevated the villus height and the ratio of villus height to crypt depth of the ileum. MCE supplementation decreased the mRNA expression of pro-inflammatory cytokines interleukin (IL)-6 and IL-8 in the jejunum. MCE addition mitigated LPS-induced mRNA up-expression of pro-inflammatory factors IL-1ß and IL-17 in the jejunum. MCE supplementation increased the abundance of probiotic bacteria (such as Lactobacillus and Blautia) and reduced the abundance of pathogenic bacteria (such as Actinobacteriota, Peptostretococcaceae, and Rhodococcus), leading to alterations in gut microbiota composition. MCE addition altered several metabolic pathways such as Amino acid metabolism, Nucleotide metabolism, Energy metabolism, Carbohydrate metabolism, and Lipid metabolism in broilers. In these pathways, MCE supplementation increased the levels of L-aspartic acid, L-Glutamate, L-serine, etc., and reduced the levels of phosphatidylcholine, phosphatidylethanolamine, thromboxane B2, 13-(S)-HODPE, etc. In conclusion, dietary supplementation of 400 mg/kg MCE effectively improved the growth performance and intestinal function in LPS-challenged broiler chickens, probably due to the modulation of gut microbiota and plasma metabolites.


Sujet(s)
Poulets , Compléments alimentaires , Microbiome gastro-intestinal , Lipopolysaccharides , Extraits de plantes , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Extraits de plantes/pharmacologie , Extraits de plantes/administration et posologie , Mâle , Papaveraceae/composition chimique , Aliment pour animaux , Maladies de la volaille/microbiologie , Maladies de la volaille/immunologie , Cytokines/métabolisme , Cytokines/sang , Intestins/effets des médicaments et des substances chimiques , Intestins/microbiologie , Intestins/immunologie
3.
Science ; 385(6708): eadk1679, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39088603

RÉSUMÉ

Neuroimmune cross-talk participates in intestinal tissue homeostasis and host defense. However, the matrix of interactions between arrays of molecularly defined neuron subsets and of immunocyte lineages remains unclear. We used a chemogenetic approach to activate eight distinct neuronal subsets, assessing effects by deep immunophenotyping, microbiome profiling, and immunocyte transcriptomics in intestinal organs. Distinct immune perturbations followed neuronal activation: Nitrergic neurons regulated T helper 17 (TH17)-like cells, and cholinergic neurons regulated neutrophils. Nociceptor neurons, expressing Trpv1, elicited the broadest immunomodulation, inducing changes in innate lymphocytes, macrophages, and RORγ+ regulatory T (Treg) cells. Neuroanatomical, genetic, and pharmacological follow-up showed that Trpv1+ neurons in dorsal root ganglia decreased Treg cell numbers via the neuropeptide calcitonin gene-related peptide (CGRP). Given the role of these neurons in nociception, these data potentially link pain signaling with gut Treg cell function.


Sujet(s)
Peptide relié au gène de la calcitonine , Ganglions sensitifs des nerfs spinaux , Neuro-immunomodulation , Nocicepteurs , Lymphocytes T régulateurs , Canaux cationiques TRPV , Cellules Th17 , Animaux , Souris , Peptide relié au gène de la calcitonine/métabolisme , Peptide relié au gène de la calcitonine/génétique , Neurones cholinergiques/métabolisme , Ganglions sensitifs des nerfs spinaux/métabolisme , Ganglions sensitifs des nerfs spinaux/cytologie , Microbiome gastro-intestinal , Intestins/immunologie , Intestins/cytologie , Macrophages/immunologie , Macrophages/métabolisme , Souris de lignée C57BL , Nociception , Nocicepteurs/métabolisme , Membre-3 du groupe F de la sous-famille-1 de récepteurs nucléaires/métabolisme , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Cellules Th17/immunologie , Canaux cationiques TRPV/métabolisme , Canaux cationiques TRPV/génétique
4.
Gastroenterol Clin North Am ; 53(3): 413-430, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39068003

RÉSUMÉ

In this review, the authors outlined concepts and strategies to achieve immune tolerance through inducing hematopoietic chimerism after solid organ transplantation and introduced challenges and opportunities in harnessing two-way alloresponses to improve outcomes after intestinal transplantation (ITx). Next, the authors discussed the dynamics and phenotypes of peripheral blood and intestinal graft T-cell subset chimerism and their association with outcomes. The authors also summarized studies on other types of immune cells after ITx and their potential participation in chimerism-mediated tolerance. The authors further discussed strategies and future directions to promote chimerism-associated tolerance after ITx to overcome rejection and minimize immunosuppression.


Sujet(s)
Intestins , Chimère obtenue par transplantation , Humains , Intestins/transplantation , Intestins/immunologie , Chimère obtenue par transplantation/immunologie , Tolérance à la transplantation/immunologie , Chimérisme , Transplantation d'organe/méthodes , Rejet du greffon/immunologie , Rejet du greffon/prévention et contrôle , Tolérance immunitaire
5.
Front Immunol ; 15: 1401949, 2024.
Article de Anglais | MEDLINE | ID: mdl-39076972

RÉSUMÉ

Introduction: Ubiquitin-specific proteases (USPs), a large subset of more than 50 deubiquitinase proteins, have recently emerged as promising targets in cancer. However, their role in immune cell regulation, particularly in T cell activation, differentiation, and effector functions, remains largely unexplored. Methods: We utilized a USP28 knockout mouse line to study the effect of USP28 on T cell activation and function, and its role in intestinal inflammation using the dextran sulfate sodium (DSS)-induced colitis model and a series of in vitro assays. Results: Our results show that USP28 exerts protective effects in acute intestinal inflammation. Mechanistically, USP28 knockout mice (USP28-/-) exhibited an increase in total T cells mainly due to an increased CD8+ T cell content. Additionally, USP28 deficiency resulted in early defects in T cell activation and functional changes. Specifically, we observed a reduced expression of IL17 and an increase in inducible regulatory T (iTreg) suppressive functions. Importantly, activated T cells lacking USP28 showed increased STAT5 phosphorylation. Consistent with these findings, these mice exhibited increased susceptibility to acute DSS-induced intestinal inflammation, accompanied by elevated IL22 cytokine levels. Conclusions: Our findings demonstrate that USP28 is essential for T cell functionality and protects mice from acute DSS-induced colitis by regulating STAT5 signaling and IL22 production. As a T cell regulator, USP28 plays a crucial role in immune responses and intestinal health.


Sujet(s)
Colite , , Interleukines , Facteur de transcription STAT-5 , Ubiquitin thiolesterase , Animaux , Souris , Colite/induit chimiquement , Colite/immunologie , Colite/métabolisme , Sulfate dextran , Modèles animaux de maladie humaine , Inflammation/immunologie , Inflammation/métabolisme , Interleukines/métabolisme , Interleukines/génétique , Intestins/immunologie , Intestins/anatomopathologie , Activation des lymphocytes/immunologie , Souris de lignée C57BL , Souris knockout , Phosphorylation , Facteur de transcription STAT-5/métabolisme , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Ubiquitin thiolesterase/génétique , Ubiquitin thiolesterase/métabolisme , Ubiquitin thiolesterase/déficit
6.
Cells ; 13(14)2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-39056760

RÉSUMÉ

Thousands struggle with acute and chronic intestinal injury due to various causes. Epithelial intestinal healing is dependent on phenotypic transitions to a mobile phenotype. Focal adhesion kinase (FAK) is a ubiquitous protein that is essential for cell mobility. This phenotype change is mediated by FAK activation and proves to be a promising target for pharmaceutical intervention. While FAK is crucial for intestinal healing, new evidence connects FAK with innate immunity and the importance it plays in macrophage/monocyte chemotaxis, as well as other intracellular signaling cascades. These cascades play a part in macrophage/monocyte polarization, maturation, and inflammation that is associated with intestinal injury. Colony stimulating factors (CSFs) such as macrophage colony stimulating factor (M-CSF/CSF-1) and granulocyte macrophage colony stimulating factor (GM-CSF/CSF-2) play a critical role in maintaining homeostasis within intestinal mucosa by crosstalk capabilities between macrophages and epithelial cells. The communication between these cells is imperative in orchestrating healing upon injury. Diving deeper into these connections may allow us a greater insight into the role that our immune system plays in healing, as well as a better comprehension of inflammatory diseases of the gut.


Sujet(s)
Homéostasie , Immunité innée , Animaux , Humains , Focal adhesion protein-tyrosine kinases/métabolisme , Muqueuse intestinale/métabolisme , Muqueuse intestinale/immunologie , Intestins/immunologie , Macrophages/métabolisme , Macrophages/immunologie , Transduction du signal
7.
Front Immunol ; 15: 1375486, 2024.
Article de Anglais | MEDLINE | ID: mdl-39007142

RÉSUMÉ

Introduction: It is unknown how intestinal B cell populations and B cell receptor (BCR) repertoires are established and maintained over time in humans. Following intestinal transplantation (ITx), surveillance ileal mucosal biopsies provide a unique opportunity to map the dynamic establishment of recipient gut lymphocyte populations in immunosuppressed conditions. Methods: Using polychromatic flow cytometry that includes HLA allele group-specific antibodies distinguishing donor from recipient cells along with high throughput BCR sequencing, we tracked the establishment of recipient B cell populations and BCR repertoire in the allograft mucosa of ITx recipients. Results: We confirm the early presence of naïve donor B cells in the circulation (donor age range: 1-14 years, median: 3 years) and, for the first time, document the establishment of recipient B cell populations, including B resident memory cells, in the intestinal allograft mucosa (recipient age range at the time of transplant: 1-44 years, median: 3 years). Recipient B cell repopulation of the allograft was most rapid in infant (<1 year old)-derived allografts and, unlike T cell repopulation, did not correlate with rejection rates. While recipient memory B cell populations were increased in graft mucosa compared to circulation, naïve recipient B cells remained detectable in the graft mucosa for years. Comparisons of peripheral and intra-mucosal B cell repertoires in the absence of rejection (recipient age range at the time of transplant: 1-9 years, median: 2 years) revealed increased BCR mutation rates and clonal expansion in graft mucosa compared to circulating B cells, but these parameters did not increase markedly after the first year post-transplant. Furthermore, clonal mixing between the allograft mucosa and the circulation was significantly greater in ITx recipients, even years after transplantation, than in deceased adult donors. In available pan-scope biopsies from pediatric recipients, we observed higher percentages of naïve recipient B cells in colon allograft compared to small bowel allograft and increased BCR overlap between native colon vs colon allograft compared to that between native colon vs ileum allograft in most cases, suggesting differential clonal distribution in large intestine vs small intestine. Discussion: Collectively, our data demonstrate intestinal mucosal B cell repertoire establishment from a circulating pool, a process that continues for years without evidence of stabilization of the mucosal B cell repertoire in pediatric ITx patients.


Sujet(s)
Muqueuse intestinale , Récepteurs pour l'antigène des lymphocytes B , Humains , Enfant , Enfant d'âge préscolaire , Adolescent , Nourrisson , Muqueuse intestinale/immunologie , Mâle , Femelle , Récepteurs pour l'antigène des lymphocytes B/génétique , Récepteurs pour l'antigène des lymphocytes B/immunologie , Adulte , Lymphocytes B/immunologie , Jeune adulte , Intestins/immunologie , Intestins/transplantation , Transplantation d'organe , Rejet du greffon/immunologie
8.
Gastroenterol Clin North Am ; 53(3): 493-507, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39068010

RÉSUMÉ

Outcomes in intestinal transplantation remain hampered by higher rates of rejection than any other solid organs. However, maintenance immunosuppression regimens have largely remained unchanged despite advances in therapies for induction and treatment of rejection and graft-versus-host disease. Recently, there have been a small number of new maintenance therapies attempted, and older agents have been used in new ways to achieve better outcomes. The authors herein review the traditional maintenance therapies and their mechanisms and then consider updates in new therapies and new ways of using old therapies for maintenance immunosuppression after intestinal transplantation.


Sujet(s)
Rejet du greffon , Immunosuppression thérapeutique , Immunosuppresseurs , Intestins , Humains , Immunosuppresseurs/usage thérapeutique , Intestins/transplantation , Intestins/immunologie , Rejet du greffon/prévention et contrôle , Rejet du greffon/immunologie , Immunosuppression thérapeutique/méthodes , Maladie du greffon contre l'hôte/prévention et contrôle
9.
Cell Metab ; 36(8): 1726-1744.e10, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-38986617

RÉSUMÉ

The intestinal tract generates significant reactive oxygen species (ROS), but the role of T cell antioxidant mechanisms in maintaining intestinal homeostasis is poorly understood. We used T cell-specific ablation of the catalytic subunit of glutamate cysteine ligase (Gclc), which impaired glutathione (GSH) production, crucially reducing IL-22 production by Th17 cells in the lamina propria, which is critical for gut protection. Under steady-state conditions, Gclc deficiency did not alter cytokine secretion; however, C. rodentium infection induced increased ROS and disrupted mitochondrial function and TFAM-driven mitochondrial gene expression, resulting in decreased cellular ATP. These changes impaired the PI3K/AKT/mTOR pathway, reducing phosphorylation of 4E-BP1 and consequently limiting IL-22 translation. The resultant low IL-22 levels led to poor bacterial clearance, severe intestinal damage, and high mortality. Our findings highlight a previously unrecognized, essential role of Th17 cell-intrinsic GSH in promoting mitochondrial function and cellular signaling for IL-22 protein synthesis, which is critical for intestinal integrity and defense against gastrointestinal infections.


Sujet(s)
Glutathion , , Interleukines , Mitochondries , Cellules Th17 , Animaux , Interleukines/métabolisme , Mitochondries/métabolisme , Glutathion/métabolisme , Cellules Th17/métabolisme , Cellules Th17/immunologie , Souris , Transduction du signal , Espèces réactives de l'oxygène/métabolisme , Souris de lignée C57BL , Citrobacter rodentium , Intestins/anatomopathologie , Intestins/immunologie , Inflammation/métabolisme , Inflammation/anatomopathologie , Infections à Enterobacteriaceae/immunologie , Infections à Enterobacteriaceae/métabolisme , Infections à Enterobacteriaceae/anatomopathologie , Souris knockout , Sérine-thréonine kinases TOR/métabolisme , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie
10.
Sci Rep ; 14(1): 16973, 2024 07 23.
Article de Anglais | MEDLINE | ID: mdl-39043769

RÉSUMÉ

Our previous research found that fecal microbiota transplantation (FMT) and inulin synergistically affected the intestinal barrier and immune system function in chicks. However, does it promote the early immunity of the poultry gut-associated lymphoid tissue (GALT)? How does it regulate the immunity? We evaluated immune-related indicators in the serum, cecal tonsil, and intestine to determine whether FMT synergistic inulin had a stronger impact on gut health and which gene expression regulation was affected. The results showed that FMT synergistic inulin increased TGF-ß secretion and intestinal goblet cell number and MUC2 expression on day 14. Expression of BAFFR, PAX5, CXCL12, and IL-2 on day 7 and expression of CXCR4 and IL-2 on day 14 in the cecal tonsils significantly increased. The transcriptome indicated that CD28 and CTLA4 were important regulatory factors in intestinal immunity. Correlation analysis showed that differential genes were related to the immunity and development of the gut and cecal tonsil. FMT synergistic inulin promoted the development of GALT, which improved the early-stage immunity of the intestine by regulating CD28 and CTLA4. This provided new measures for replacing antibiotic use and reducing the use of therapeutic drugs while laying a technical foundation for achieving anti-antibiotic production of poultry products.


Sujet(s)
Poulets , Transplantation de microbiote fécal , Inuline , Animaux , Inuline/pharmacologie , Poulets/microbiologie , Poulets/immunologie , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Intestins/immunologie , Intestins/microbiologie , Muqueuse intestinale/métabolisme , Muqueuse intestinale/immunologie , Muqueuse intestinale/microbiologie , Caecum/microbiologie
11.
Fish Shellfish Immunol ; 151: 109739, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38960108

RÉSUMÉ

Lauric acid (LA), a saturated fatty acid with 12 carbon atoms, is widely regarded as a healthy fatty acid that plays an important role in disease resistance and improving immune physiological function. The objective of this study was to determine the effects of dietary lauric acid on the growth performance, antioxidant capacity, non-specific immunity and intestinal microbiology, and evaluate the potential of lauric acids an environmentally friendly additive in swimming crab (Portunus trituberculatus) culture. A total of 192 swimming crabs with an initial body weight of 11.68 ± 0.02 g were fed six different dietary lauric acid levels, the analytical values of lauric acid were 0.09, 0.44, 0.80, 1.00, 1.53, 2.91 mg/g, respectively. There were four replicates per treatment and 8 juvenile swimming crabs per replicate. The results indicated that final weight, percent weight gain, specific growth rate, survival and feed intake were not significantly affected by dietary lauric acid levels; however, crabs fed diets with 0.80 and 1.00 mg/g lauric acid showed the lowest feed efficiency among all treatments. Proximate composition in hepatopancreas and muscle were not significantly affected by dietary lauric acid levels. The highest activities of amylase and lipase in hepatopancreas and intestine were found at crabs fed diet with 0.80 mg/g lauric acid (P < 0.05), the activity of carnitine palmityl transferase (CPT) in hepatopancreas and intestine significantly decreased with dietary lauric acid levels increasing from 0.09 to 2.91 mg/g (P < 0.05). The lowest concentration of glucose and total protein and the activity of alkaline phosphatase in hemolymph were observed at crabs fed diets with 0.80 and 1.00 mg/g lauric acid among all treatments. The activity of GSH-Px in hepatopancreas significantly increased with dietary lauric acid increasing from 0.09 to 1.53 mg/g, MDA in hepatopancreas and hemolymph was not significantly influenced by dietary lauric acid levels. The highest expression of cat and gpx in hepatopancreas were exhibited in crabs fed diet with 1.00 mg/g lauric acid, however, the expression of genes related to the inflammatory signaling pathway (relish, myd88, traf6, nf-κB) were up-regulated in the hepatopancreas with dietary lauric acid levels increasing from 0.09 to 1.00 mg/g, moreover, the expression of genes related to intestinal inflammatory, immune and antioxidant were significantly affected by dietary lauric acid levels (P < 0.05). Crabs fed diet without lauric acid supplementation exhibited higher lipid drop area in hepatopancreas than those fed the other diets (P < 0.05). The expression of genes related to lipid catabolism was up-regulated, however, and the expression of genes related to lipid synthesis was down-regulated in the hepatopancreas of crabs fed with 0.80 mg/g lauric acid. Lauric acid improved hepatic tubular integrity, and enhanced intestinal barrier function by increasing peritrophic membrane (PM) thickness and upregulating the expression of structural factors (per44, zo-1) and intestinal immunity-related genes. In addition, dietary 1.00 mg/g lauric acid significantly improved the microbiota composition of the intestinal, increased the abundance of Actinobacteria and Rhodobacteraceae, and decreased the abundance of Vibrio, thus maintaining the microbiota balance of the intestine. The correlation analysis showed that there was a relationship between intestinal microbiota and immune-antioxidant function. In conclusion, the dietary 1.00 mg/g lauric acid is beneficial to improve the antioxidant capacity and intestinal health of swimming crab.


Sujet(s)
Aliment pour animaux , Antioxydants , Brachyura , Régime alimentaire , Compléments alimentaires , Microbiome gastro-intestinal , Acides lauriques , Animaux , Brachyura/immunologie , Brachyura/effets des médicaments et des substances chimiques , Brachyura/croissance et développement , Brachyura/microbiologie , Acides lauriques/pharmacologie , Acides lauriques/administration et posologie , Aliment pour animaux/analyse , Antioxydants/métabolisme , Régime alimentaire/médecine vétérinaire , Compléments alimentaires/analyse , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Immunité innée/effets des médicaments et des substances chimiques , Intestins/effets des médicaments et des substances chimiques , Intestins/immunologie , Répartition aléatoire , Relation dose-effet des médicaments
12.
Fish Shellfish Immunol ; 151: 109750, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38969153

RÉSUMÉ

The largemouth bass has become one of the economically fish in China, according to the latest China Fishery Statistical Yearbook. The farming scale is constantly increasing. Salidroside has been found in past studies to have oxidative stress reducing and immune boosting properties. In this study, the addition of six different levels of salidroside supplements were 0、40、80、120、160 and 200 mg/kg. A 56-day feeding trial was conducted to investigate the effects of salidroside on the intestinal health, immune parameters and intestinal microbiota composition of largemouth bass. Dietary addition of salidroside significantly affected the Keap-1ß/Nrf-2 pathway as well as significantly increased antioxidant enzyme activities resulting in a significant increase in antioxidant capacity of largemouth bass. Dietary SLR significantly reduced feed coefficients. The genes related to tight junction proteins (Occludin, ZO-1, Claudin-4, Claudin-5) were found to be significantly upregulated in the diet supplemented with salidroside, indicating that salidroside can improve the intestinal barrier function (p < 0.05). The dietary administration of salidroside was found to significantly reduce the transcription levels of intestinal tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß) (p < 0.05). Furthermore, salidroside was observed to reduce the transcription levels of intestinal apoptosis factor Bcl-2 associated death promoter (BAD) and recombinant Tumor Protein p53 (P53) (p < 0.05). Concomitantly, the beneficial bacteria, Fusobacteriota and Cetobacterium, was significantly increased in the SLR12 group, while that of pathogenic bacteria, Proteobacteria, was significantly decreased (p < 0.05). In conclusion, the medium-sized largemouth bass optimal dosage of salidroside in the diet is 120mg/kg-1.


Sujet(s)
Aliment pour animaux , Serran , Régime alimentaire , Compléments alimentaires , Microbiome gastro-intestinal , Glucosides , Phénols , Animaux , Serran/immunologie , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Aliment pour animaux/analyse , Régime alimentaire/médecine vétérinaire , Compléments alimentaires/analyse , Glucosides/administration et posologie , Glucosides/pharmacologie , Phénols/administration et posologie , Phénols/pharmacologie , Intestins/effets des médicaments et des substances chimiques , Intestins/immunologie , Intestins/microbiologie , Immunité innée/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Répartition aléatoire
13.
Proc Natl Acad Sci U S A ; 121(29): e2402126121, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38980902

RÉSUMÉ

Upon sensing viral RNA, mammalian RIG-I-like receptors (RLRs) activate downstream signals using caspase activation and recruitment domains (CARDs), which ultimately promote transcriptional immune responses that have been well studied. In contrast, the downstream signaling mechanisms for invertebrate RLRs are much less clear. For example, the Caenorhabditis elegans RLR DRH-1 lacks annotated CARDs and up-regulates the distinct output of RNA interference. Here, we found that similar to mammal RLRs, DRH-1 signals through two tandem CARDs (2CARD) to induce a transcriptional immune response. Expression of DRH-1(2CARD) alone in the intestine was sufficient to induce immune gene expression, increase viral resistance, and promote thermotolerance, a phenotype previously associated with immune activation in C. elegans. We also found that DRH-1 is required in the intestine to induce immune gene expression, and we demonstrate subcellular colocalization of DRH-1 puncta with double-stranded RNA inside the cytoplasm of intestinal cells upon viral infection. Altogether, our results reveal mechanistic and spatial insights into antiviral signaling in C. elegans, highlighting unexpected parallels in RLR signaling between C. elegans and mammals.


Sujet(s)
Protéines de Caenorhabditis elegans , Caenorhabditis elegans , Transduction du signal , Animaux , Caenorhabditis elegans/immunologie , Caenorhabditis elegans/métabolisme , Protéines de Caenorhabditis elegans/métabolisme , Protéines de Caenorhabditis elegans/génétique , Protéines de Caenorhabditis elegans/immunologie , Transduction du signal/immunologie , Intestins/immunologie , Intestins/virologie , DEAD-box RNA helicases/métabolisme , DEAD-box RNA helicases/génétique , ARN double brin/métabolisme , ARN double brin/immunologie , Immunité innée , Muqueuse intestinale/immunologie , Muqueuse intestinale/métabolisme , ARN viral/immunologie , ARN viral/métabolisme , ARN viral/génétique
14.
Int Immunopharmacol ; 138: 112592, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-38955024

RÉSUMÉ

Cerebral ischemia-induced systemic inflammation and inflammasome-dependent pyroptotic cell death in ileum, causing serious intestinal injury. Glucocorticoid receptor (GR) mediates the effects of glucocorticoids and participates in inflammation. Escin has corticosteroid-like, neuroprotective, and anti-intestinal dysfunction effects. This study aimed to investigate the effect of Escin on the intestinal barrier injury in rats subjected to middle cerebral artery occlusion (MCAO) and on Caco-2 cells exposed to lipopolysaccharides. The MCAO-caused brain injury was evaluated by assessing neurological function, cerebral infarct volume, and plasma corticosterone (Cort) levels. Intestinal injury was evaluated by observing the histopathological changes, assessing the intestinal barrier function, and determining blood FD4, endotoxin and IL-1ß levels. The levels of the tight-junction proteins such as claudin-1, occludin, and ZO-1, and proteins involved in the GR/p38 MAPK/NF-κB pathway and NLRP3-inflammasome activation were evaluated using western blotting or immunofluorescence. Administration of Escin suppressed the cerebral ischemia-induced increases in Garcia-test scores and infarct volume, alleviated the injury to the intestinal barrier, and decreased the levels of Cort, endotoxin, and IL-1ß. Additionally, Escin upregulated GR and downregulated phospho(p)-p65, p-p38MAPK, NLRP3, GSDMD-N, and cleaved-caspase-1 in the intestine. The effects of Escin could be suppressed by the GR antagonist RU486 or enhanced by the p38 MAPK antagonist SB203580. We revealed details how Escin improves cerebral ischemia-induced intestinal barrier injury by upregulating GR and thereby inhibiting the pyroptosis induced by NF-κB-mediated NLRP3 activation. This study will provide a experimental foundation for the features of glucocorticoid-like activity and the discovery of new clinical application for Escin.


Sujet(s)
Encéphalopathie ischémique , Aescine , Inflammasomes , Pyroptose , Récepteurs aux glucocorticoïdes , Transduction du signal , Animaux , Humains , Mâle , Rats , Encéphalopathie ischémique/traitement médicamenteux , Encéphalopathie ischémique/métabolisme , Cellules Caco-2 , Modèles animaux de maladie humaine , Aescine/pharmacologie , Aescine/usage thérapeutique , Infarctus du territoire de l'artère cérébrale moyenne/traitement médicamenteux , Infarctus du territoire de l'artère cérébrale moyenne/anatomopathologie , Infarctus du territoire de l'artère cérébrale moyenne/immunologie , Inflammasomes/métabolisme , Interleukine-1 bêta/métabolisme , Intestins/anatomopathologie , Intestins/effets des médicaments et des substances chimiques , Intestins/immunologie , Lipopolysaccharides , Facteur de transcription NF-kappa B/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , p38 Mitogen-Activated Protein Kinases/métabolisme , Pyroptose/effets des médicaments et des substances chimiques , Rat Sprague-Dawley , Récepteurs aux glucocorticoïdes/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
15.
Cell Rep ; 43(7): 114490, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38990720

RÉSUMÉ

Although oral tolerance is a critical system in regulating allergic disorders, the mechanisms by which dietary factors regulate the induction and maintenance of oral tolerance remain unclear. To address this, we explored the differentiation and function of various immune cells in the intestinal immune system under fasting and ad libitum-fed conditions before oral ovalbumin (OVA) administration. Fasting mitigated OVA-specific Treg expansion, which is essential for oral tolerance induction. This abnormality mainly resulted from functional defects in the CX3CR1+ cells responsible for the uptake of luminal OVA and reduction of tolerogenic CD103+ dendritic cells. Eventually, fasting impaired the preventive effect of oral OVA administration on asthma and allergic rhinitis development. Specific food ingredients, namely carbohydrates and arginine, were indispensable for oral tolerance induction by activating glycolysis and mTOR signaling. Overall, prior food intake and nutritional signals are critical for maintaining immune homeostasis by inducing tolerance to ingested food antigens.


Sujet(s)
Arginine , Cellules dendritiques , Tolérance immunitaire , Ovalbumine , Lymphocytes T régulateurs , Sérine-thréonine kinases TOR , Animaux , Arginine/métabolisme , Lymphocytes T régulateurs/immunologie , Ovalbumine/immunologie , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Souris , Sérine-thréonine kinases TOR/métabolisme , Souris de lignée C57BL , Administration par voie orale , Récepteur-1 de la chimiokine CX3C/métabolisme , Intestins/immunologie , Antigènes CD/métabolisme , Intégrines alpha/métabolisme , Sucres/métabolisme , Glycolyse , Jeûne , Transduction du signal , Muqueuse intestinale/immunologie , Muqueuse intestinale/métabolisme , Femelle
17.
Parasit Vectors ; 17(1): 284, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956725

RÉSUMÉ

BACKGROUND: Toxoplasma gondii infection affects a significant portion of the global population, leading to severe toxoplasmosis and, in immunocompromised patients, even death. During T. gondii infection, disruption of gut microbiota further exacerbates the damage to intestinal and brain barriers. Therefore, identifying imbalanced probiotics during infection and restoring their equilibrium can regulate the balance of gut microbiota metabolites, thereby alleviating tissue damage. METHODS: Vimentin gene knockout (vim-/-) mice were employed as an immunocompromised model to evaluate the influence of host immune responses on gut microbiota balance during T. gondii infection. Behavioral experiments were performed to assess changes in cognitive levels and depressive tendencies between chronically infected vim-/- and wild-type (WT) mice. Fecal samples were subjected to 16S ribosomal RNA (rRNA) sequencing, and serum metabolites were analyzed to identify potential gut probiotics and their metabolites for the treatment of T. gondii infection. RESULTS: Compared to the immunocompetent WT sv129 mice, the immunocompromised mice exhibited lower levels of neuronal apoptosis and fewer neurobehavioral abnormalities during chronic infection. 16S rRNA sequencing revealed a significant decrease in the abundance of probiotics, including several species of Lactobacillus, in WT mice. Restoring this balance through the administration of Lactobacillus murinus and Lactobacillus gasseri significantly suppressed the T. gondii burden in the intestine, liver, and brain. Moreover, transplantation of these two Lactobacillus spp. significantly improved intestinal barrier damage and alleviated inflammation and neuronal apoptosis in the central nervous system. Metabolite detection studies revealed that the levels of various Lactobacillus-related metabolites, including indole-3-lactic acid (ILA) in serum, decreased significantly after T. gondii infection. We confirmed that L. gasseri secreted much more ILA than L. murinus. Notably, ILA can activate the aromatic hydrocarbon receptor signaling pathway in intestinal epithelial cells, promoting the activation of CD8+ T cells and the secretion of interferon-gamma. CONCLUSION: Our study revealed that host immune responses against T. gondii infection severely disrupted the balance of gut microbiota, resulting in intestinal and brain damage. Lactobacillus spp. play a crucial role in immune regulation, and the metabolite ILA is a promising therapeutic compound for efficient and safe treatment of T. gondii infection.


Sujet(s)
Lésions encéphaliques , Microbiome gastro-intestinal , Souris knockout , Toxoplasma , Animaux , Souris , Toxoplasma/immunologie , Lésions encéphaliques/immunologie , Probiotiques/administration et posologie , Encéphale/immunologie , Lactobacillus , Modèles animaux de maladie humaine , Sujet immunodéprimé , Toxoplasmose/immunologie , ARN ribosomique 16S/génétique , Mâle , Intestins/immunologie
18.
Fish Shellfish Immunol ; 151: 109741, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38964436

RÉSUMÉ

Decay-accelerating factor (DAF) is an essential member of the complement regulatory protein family that plays an important role in immune response and host homeostasis in mammals. However, the immune function of DAF has not been well characterized in bony fish. In this study, a complement regulatory protein named CiDAF was firstly characterized from Ctenopharyngodon idella and its potential roles were investigated in intestine following bacterial infection. Similar to mammalian DAFs, CiDAF has multiple complement control protein (CCP) functional domains, suggesting the evolutionary conservation of DAFs. CiDAF was broadly expressed in all tested tissues, with a relatively high expression level detected in the spleen and kidney. In vivo immune challenge experiments revealed that CiDAF strongly responded to bacterial pathogens (Aeromonas hydrophila and Aeromonas veronii) and PAMPs (lipopolysaccharide (LPS) or muramyl dipeptide (MDP)) challenges. In vitro RNAi experiments indicated that knockdown of CiDAF could upregulate the expression of complement genes (C4b, C5 and C7) and inflammatory cytokines (TNF-α, IL-1ß and IL-8). Moreover, 2000 ng/mL of CiDAF agonist progesterone effectively alleviated LPS- or MDP-induced intestinal inflammation by regulating expression of complement factors, TLR/PepT1 pathway genes and inflammatory cytokines. Overall, these findings revealed that CiDAF may act as a negative regulator of intestinal complement pathway and immune response to bacterial challenge in grass carp.


Sujet(s)
Carpes (poisson) , Maladies des poissons , Protéines de poisson , Infections bactériennes à Gram négatif , Immunité innée , Intestins , Animaux , Carpes (poisson)/immunologie , Protéines de poisson/génétique , Protéines de poisson/immunologie , Maladies des poissons/immunologie , Immunité innée/génétique , Infections bactériennes à Gram négatif/immunologie , Infections bactériennes à Gram négatif/médecine vétérinaire , Intestins/immunologie , Régulation de l'expression des gènes/immunologie , Phylogenèse , Analyse de profil d'expression de gènes/médecine vétérinaire , Aeromonas hydrophila/physiologie , Séquence d'acides aminés , Alignement de séquences/médecine vétérinaire , Protéines du système du complément/immunologie
20.
Gastroenterol Clin North Am ; 53(3): 359-382, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39068000

RÉSUMÉ

Intestinal transplantation is a life-saving procedure utilized for patients failing total parenteral nutrition. However, intestinal transplantattion remains plagued with low survival rates and high risk of allograft rejection. The authors explore roles of innate (macrophages, natural killer cells, innate lymphoid cells) and adaptive immune cells (Th1, Th2, Th17, Tregs) in inflammatory responses, particularly inflammatory bowel disease and graft versus host disease, and correlate these findings to intestinal allograft rejection, highlighting which effectors exacerbate or suppress intestinal rejection. Better understanding of this immunology can open further investigation into potential biomolecular targets to develop improved therapeutic treatment options and immunomonitoring techniques to combat allograft rejection and enhance patient lives.


Sujet(s)
Immunité acquise , Rejet du greffon , Maladie du greffon contre l'hôte , Immunité innée , Maladies inflammatoires intestinales , Intestins , Humains , Rejet du greffon/immunologie , Maladie du greffon contre l'hôte/immunologie , Maladie du greffon contre l'hôte/étiologie , Maladies inflammatoires intestinales/immunologie , Intestins/immunologie , Intestins/transplantation , Cellules tueuses naturelles/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE