Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41.805
Filter
1.
Gut Microbes ; 16(1): 2361493, 2024.
Article in English | MEDLINE | ID: mdl-38958039

ABSTRACT

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


Subject(s)
Bacteria , Colitis , Gastrointestinal Microbiome , Intestinal Mucosa , Oxygen , Colitis/microbiology , Colitis/chemically induced , Colitis/metabolism , Animals , Humans , Oxygen/metabolism , Bacteria/metabolism , Bacteria/classification , Bacteria/isolation & purification , Bacteria/genetics , Mice , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Feces/microbiology , Mice, Inbred C57BL , Dextran Sulfate , Colon/microbiology , Colon/metabolism , Male
2.
Sci Rep ; 14(1): 15460, 2024 07 04.
Article in English | MEDLINE | ID: mdl-38965287

ABSTRACT

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


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Disease Models, Animal , Gastrointestinal Microbiome , Lacticaseibacillus rhamnosus , Mice, Inbred C57BL , Probiotics , Animals , Gastrointestinal Microbiome/drug effects , Mice , Lacticaseibacillus rhamnosus/physiology , Male , Probiotics/pharmacology , Probiotics/administration & dosage , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Parkinson Disease/microbiology , Corpus Striatum/metabolism , MPTP Poisoning/microbiology , MPTP Poisoning/metabolism , MPTP Poisoning/drug therapy , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/drug effects , Dopamine/metabolism
3.
Sci Rep ; 14(1): 15442, 2024 07 04.
Article in English | MEDLINE | ID: mdl-38965312

ABSTRACT

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


Subject(s)
Epithelial Cells , Intestinal Mucosa , beta-Defensins , Humans , beta-Defensins/metabolism , beta-Defensins/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Signal Transduction , Gene Expression Regulation , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , RNA Interference
4.
Front Endocrinol (Lausanne) ; 15: 1398462, 2024.
Article in English | MEDLINE | ID: mdl-38957441

ABSTRACT

Background: Cannabidiol (CBD), a non-psychoactive phytocannabinoid of cannabis, is therapeutically used as an analgesic, anti-convulsant, anti-inflammatory, and anti-psychotic drug. There is a growing concern about the adverse side effects posed by CBD usage. Pregnane X receptor (PXR) is a nuclear receptor activated by a variety of dietary steroids, pharmaceutical agents, and environmental chemicals. In addition to the role in xenobiotic metabolism, the atherogenic and dyslipidemic effects of PXR have been revealed in animal models. CBD has a low affinity for cannabinoid receptors, thus it is important to elucidate the molecular mechanisms by which CBD activates cellular signaling and to assess the possible adverse impacts of CBD on pro-atherosclerotic events in cardiovascular system, such as dyslipidemia. Objective: Our study aims to explore the cellular and molecular mechanisms by which exposure to CBD activates human PXR and increases the risk of dyslipidemia. Methods: Both human hepatic and intestinal cells were used to test if CBD was a PXR agonist via cell-based transfection assay. The key residues within PXR's ligand-binding pocket that CBD interacted with were investigated using computational docking study together with site-directed mutagenesis assay. The C57BL/6 wildtype mice were orally fed CBD in the presence of PXR antagonist resveratrol (RES) to determine how CBD exposure could change the plasma lipid profiles in a PXR-dependent manner. Human intestinal cells were treated with CBD and/or RES to estimate the functions of CBD in cholesterol uptake. Results: CBD was a selective agonist of PXR with higher activities on human PXR than rodents PXRs and promoted the dissociation of human PXR from nuclear co-repressors. The key amino acid residues Met246, Ser247, Phe251, Phe288, Trp299, and Tyr306 within PXR's ligand binding pocket were identified to be necessary for the agonistic effects of CBD. Exposure to CBD increased the circulating total cholesterol levels in mice which was partially caused by the induced expression levels of the key intestinal PXR-regulated lipogenic genes. Mechanistically, CBD induced the gene expression of key intestinal cholesterol transporters, which led to the increased cholesterol uptake by intestinal cells. Conclusion: CBD was identified as a selective PXR agonist. Exposure to CBD activated PXR signaling and increased the atherogenic cholesterol levels in plasma, which partially resulted from the ascended cholesterol uptake by intestinal cells. Our study provides potential evidence for the future risk assessment of CBD on cardiovascular disease, such as dyslipidemia.


Subject(s)
Cannabidiol , Cholesterol , Mice, Inbred C57BL , Pregnane X Receptor , Pregnane X Receptor/metabolism , Animals , Humans , Mice , Cannabidiol/pharmacology , Cholesterol/metabolism , Male , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Molecular Docking Simulation
5.
Virol J ; 21(1): 157, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992629

ABSTRACT

Newborn piglets' health is seriously threatened by the porcine epidemic diarrhea virus (PEDV), which also has a significant effect on the pig industry. The gut microbiota produces butyrate, an abundant metabolite that modulates intestinal function through many methods to improve immunological and intestinal barrier function. The objective of this investigation was to ascertain how elevated butyrate concentrations impacted the host transcriptional profile of PEDV CV777 strain infection. Our findings showed that higher concentrations of butyrate have a stronger inhibitory effect on PEDV CV777 strain infection. According to RNA-seq data, higher concentrations of butyrate induced more significant transcriptional changes in IPEC-J2 cells, and signaling pathways such as PI3K-AKT may play a role in the inhibition of PEDV CV777 strain by high concentrations of butyrate. Ultimately, we offer a theoretical and experimental framework for future research and development of novel approaches to harness butyrate's antiviral infection properties.


Subject(s)
Butyrates , Epithelial Cells , Porcine epidemic diarrhea virus , Animals , Porcine epidemic diarrhea virus/drug effects , Porcine epidemic diarrhea virus/physiology , Swine , Butyrates/pharmacology , Butyrates/metabolism , Epithelial Cells/virology , Epithelial Cells/drug effects , Cell Line , Swine Diseases/virology , Coronavirus Infections/virology , Coronavirus Infections/drug therapy , Coronavirus Infections/veterinary , Antiviral Agents/pharmacology , Signal Transduction/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/virology , Intestinal Mucosa/drug effects , Virus Replication/drug effects , Intestines/virology
6.
Cells ; 13(13)2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38994934

ABSTRACT

The luminal surface of the intestinal epithelium is protected by a vital mucus layer, which is essential for lubrication, hydration, and fostering symbiotic bacterial relationships. Replicating and studying this complex mucus structure in vitro presents considerable challenges. To address this, we developed a hydrogel-integrated millifluidic tissue chamber capable of applying precise apical shear stress to intestinal models cultured on flat or 3D structured hydrogel scaffolds with adjustable stiffness. The chamber is designed to accommodate nine hydrogel scaffolds, 3D-printed as flat disks with a storage modulus matching the physiological range of intestinal tissue stiffness (~3.7 kPa) from bioactive decellularized and methacrylated small intestinal submucosa (dSIS-MA). Computational fluid dynamics simulations were conducted to confirm a laminar flow profile for both flat and 3D villi-comprising scaffolds in the physiologically relevant regime. The system was initially validated with HT29-MTX seeded hydrogel scaffolds, demonstrating accelerated differentiation, increased mucus production, and enhanced 3D organization under shear stress. These characteristic intestinal tissue features are essential for advanced in vitro models as they critically contribute to a functional barrier. Subsequently, the chamber was challenged with human intestinal stem cells (ISCs) from the terminal ileum. Our findings indicate that biomimicking hydrogel scaffolds, in combination with physiological shear stress, promote multi-lineage differentiation, as evidenced by a gene and protein expression analysis of basic markers and the 3D structural organization of ISCs in the absence of chemical differentiation triggers. The quantitative analysis of the alkaline phosphatase (ALP) activity and secreted mucus demonstrates the functional differentiation of the cells into enterocyte and goblet cell lineages. The millifluidic system, which has been developed and optimized for performance and cost efficiency, enables the creation and modulation of advanced intestinal models under biomimicking conditions, including tunable matrix stiffness and varying fluid shear stresses. Moreover, the readily accessible and scalable mucus-producing cellular tissue models permit comprehensive mucus analysis and the investigation of pathogen interactions and penetration, thereby offering the potential to advance our understanding of intestinal mucus in health and disease.


Subject(s)
Hydrogels , Mucus , Humans , Mucus/metabolism , Hydrogels/chemistry , Tissue Scaffolds/chemistry , Intestinal Mucosa/metabolism , HT29 Cells , Models, Biological , Stem Cells/metabolism , Stem Cells/cytology , Cell Differentiation/drug effects , Printing, Three-Dimensional , Tissue Engineering/methods
7.
Cells ; 13(13)2024 Jun 22.
Article in English | MEDLINE | ID: mdl-38994938

ABSTRACT

In Crohn's Disease (CD), intestinal fibrosis is a prevalent yet unresolved complication arising from chronic and transmural inflammation. The histological assessment of CD intestines shows changes in tissue morphology in all the layers, including the mucosa and muscularis. This study aimed to determine the differences in fibrogenesis between mucosa and muscularis. Human precision-cut intestinal slices (hPCIS) were prepared from human intestine mucosa and muscularis and treated with TGF-ß1 and/or PDGF-BB for 72 h. Gene and protein expression and matrix metalloproteinase (MMP) activity were determined. The basal gene expression of various fibrosis markers was higher in muscularis compared to mucosa hPCIS. During incubation, Pro-Collagen-1A1 secretion increased in muscularis but not in mucosa hPCIS. MMP gene expression increased during incubation in mucosa and muscularis hPCIS, except for MMP9, MMP12, and MMP13 in muscularis hPCIS. Incubation with TGF-ß1 caused increased COL1A1 expression in the mucosa but not in muscularis hPCIS. In muscularis hPCIS, TGF-ß1 treatment caused a decrease in MMP1 and CTSK expression, while MMP13 was increased. In the presence of TGF-ß1, protease inhibitor expression was stable, except for SERPINE1, which was increased in muscularis hPCIS. We conclude that fibrogenesis is more pronounced in muscularis hPCIS compared to mucosa hPCIS, especially when stimulated with TGF-ß1.


Subject(s)
Fibrosis , Intestinal Mucosa , Transforming Growth Factor beta1 , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/drug effects , Transforming Growth Factor beta1/metabolism , Collagen Type I, alpha 1 Chain , Matrix Metalloproteinases/metabolism , Matrix Metalloproteinases/genetics , Crohn Disease/pathology , Crohn Disease/metabolism , Crohn Disease/genetics , Collagen Type I/metabolism , Collagen Type I/genetics , Muscle, Smooth/metabolism , Muscle, Smooth/pathology , Muscle, Smooth/drug effects , Male , Female , Adult
8.
Cells ; 13(13)2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38994991

ABSTRACT

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


Subject(s)
Bacterial Toxins , Clostridium perfringens , Epithelial Cells , Hemolysin Proteins , Intestinal Mucosa , Pyroptosis , Reactive Oxygen Species , Clostridium perfringens/pathogenicity , Bacterial Toxins/toxicity , Bacterial Toxins/metabolism , Pyroptosis/drug effects , Animals , Hemolysin Proteins/metabolism , Hemolysin Proteins/toxicity , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Reactive Oxygen Species/metabolism , Cell Line , Mice , Humans , Mitochondria/metabolism , Mitochondria/drug effects
9.
AAPS PharmSciTech ; 25(6): 163, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-38997614

ABSTRACT

Some glycoside drugs can be transported through intestinal glucose transporters (IGTs). The surfactants used in oral drug preparations can affect the function of transporter proteins. This study aimed to investigate the effect of commonly used surfactants, Poloxamer 188 and Tween 80, on the drug transport capacity of IGTs. Previous studies have shown that gastrodin is the optimal drug substrate for IGTs. Gastrodin was used as a probe drug to evaluate the effect of these two surfactants on intestinal absorption in SD rats through pharmacokinetic and in situ single-pass intestinal perfusion. Then, the effects of the two surfactants on the expression of glucose transporters and tight-junction proteins were examined using RT-PCR and western blotting. Additionally, the effect of surfactants on intestinal permeability was evaluated through hematoxylin-eosin staining. The results found that all experimental for Poloxamer 188 (0.5%, 2.0% and 8.0%) and Tween 80 (0.1% and 2.0%) were not significantly different from those of the blank group. However, the AUC(0-∞) of gastrodin increased by approximately 32% when 0.5% Tween 80 was used. The changes in IGT expression correlated with the intestinal absorption of gastrodin. A significant increase in the expression of IGTs was observed at 0.5% Tween 80. In conclusion, Poloxamer 188 had minimal effect on the drug transport capacity of IGTs within the recommended limits of use. However, the expression of IGTs increased in response to 0.5% Tween 80, which significantly enhanced the drug transport capacity of IGTs. However, 0.1% and 2.0% Tween 80 had no significant effect.


Subject(s)
Intestinal Absorption , Intestinal Mucosa , Poloxamer , Polysorbates , Rats, Sprague-Dawley , Surface-Active Agents , Animals , Poloxamer/pharmacology , Polysorbates/pharmacology , Rats , Intestinal Absorption/drug effects , Male , Surface-Active Agents/pharmacology , Biological Transport/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Glucose Transport Proteins, Facilitative/metabolism , Glucosides/pharmacology
10.
Ann Med ; 56(1): 2361825, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38973375

ABSTRACT

BACKGROUND: Intestinal barrier dysfunction and systemic inflammation are common in obstructive sleep apnoea (OSA). We aimed to investigate the role of melatonin, an anti-inflammatory mediator, in mediating the relationships between OSA, intestinal barrier dysfunction and systemic inflammation. METHODS: Two hundred and thirty-five male participants who complained with sleep problems and underwent whole night polysomnography at our sleep centre between 2017 and 2018 were enrolled. Polysomnographic data, anthropometric measurements and biochemical indicators were collected. Serum melatonin, intestinal barrier function biomarker zonula occludens-1 (ZO-1) and inflammatory biomarkers C-reactive protein (CRP) with lipopolysaccharide (LPS) were detected. Spearman's correlation analysis assessed the correlations between sleep parameters, melatonin and biomarkers (ZO-1, LPS and CRP). Mediation analysis explored the effect of OSA on intestinal barrier dysfunction and systemic inflammation in moderate-severe OSA patients. RESULTS: As OSA severity increased, serum melatonin decreased, whereas ZO-1, LPS and CRP increased. Spearman's correlation analysis showed that serum melatonin was significantly negatively correlated with ZO-1 (r = -0.19, p < .05) and LPS (r = -0.20, p < .05) in the moderate-OSA group; serum melatonin was significantly negatively correlated with ZO-1 (r = -0.46, p < .01), LPS (r = -0.35, p < .01) and CPR (r = -0.30, p < .05) in the severe-OSA group. Mediation analyses showed melatonin explain 36.12% and 35.38% of the effect of apnoea-hypopnea index (AHI) on ZO-1 and LPS in moderate to severe OSA patients. CONCLUSIONS: Our study revealed that melatonin may be involved in mediating intestinal barrier dysfunction and systemic inflammation in moderate-to-severe OSA patients.


Subject(s)
Biomarkers , C-Reactive Protein , Inflammation , Melatonin , Polysomnography , Sleep Apnea, Obstructive , Zonula Occludens-1 Protein , Humans , Melatonin/blood , Male , Sleep Apnea, Obstructive/blood , Sleep Apnea, Obstructive/physiopathology , Sleep Apnea, Obstructive/complications , Middle Aged , C-Reactive Protein/analysis , C-Reactive Protein/metabolism , Inflammation/blood , Adult , Zonula Occludens-1 Protein/metabolism , Zonula Occludens-1 Protein/blood , Biomarkers/blood , Intestinal Mucosa/metabolism , Severity of Illness Index , Lipopolysaccharides
11.
Sci Rep ; 14(1): 15706, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38977770

ABSTRACT

Maintaining the mucus layer is crucial for the innate immune system. Urolithin A (Uro A) is a gut microbiota-derived metabolite; however, its effect on mucin production as a physical barrier remains unclear. This study aimed to elucidate the protective effects of Uro A on mucin production in the colon. In vivo experiments employing wild-type mice, NF-E2-related factor 2 (Nrf2)-deficient mice, and wild-type mice treated with an aryl hydrocarbon receptor (AhR) antagonist were conducted to investigate the physiological role of Uro A. Additionally, in vitro assays using mucin-producing cells (LS174T) were conducted to assess mucus production following Uro A treatment. We found that Uro A thickened murine colonic mucus via enhanced mucin 2 expression facilitated by Nrf2 and AhR signaling without altering tight junctions. Uro A reduced mucosal permeability in fluorescein isothiocyanate-dextran experiments and alleviated dextran sulfate sodium-induced colitis. Uro A treatment increased short-chain fatty acid-producing bacteria and propionic acid concentration. LS174T cell studies confirmed that Uro A promotes mucus production through the AhR and Nrf2 pathways. In conclusion, the enhanced intestinal mucus secretion induced by Uro A is mediated through the actions of Nrf-2 and AhR, which help maintain intestinal barrier function.


Subject(s)
Colitis , Coumarins , Intestinal Mucosa , NF-E2-Related Factor 2 , Receptors, Aryl Hydrocarbon , Animals , NF-E2-Related Factor 2/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Mice , Intestinal Mucosa/metabolism , Coumarins/pharmacology , Colitis/metabolism , Colitis/chemically induced , Mucin-2/metabolism , Mucin-2/genetics , Humans , Colon/metabolism , Mice, Inbred C57BL , Signal Transduction/drug effects , Male , Gastrointestinal Microbiome , Mice, Knockout , Dextran Sulfate , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Intestinal Barrier Function
12.
Sci Rep ; 14(1): 15705, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38977802

ABSTRACT

Ulcerative colitis (UC) is a chronic inflammatory condition of the intestinal tract. Various programmed cell death pathways in the intestinal mucosa are crucial to the pathogenesis of UC. Disulfidptosis, a recently identified form of programmed cell death, has not been extensively reported in the context of UC. This study evaluated the expression of disulfidptosis-related genes (DRGs) in UC through public databases and assessed disulfide accumulation in the intestinal mucosal tissues of UC patients and dextran sulfate sodium (DSS)-induced colitis mice via targeted metabolomics. We utilized various bioinformatics techniques to identify UC-specific disulfidptosis signature genes, analyze their potential functions, and investigate their association with immune cell infiltration in UC. The mRNA and protein expression levels of these signature genes were confirmed in the intestinal mucosa of DSS-induced colitis mice and UC patients. A total of 24 DRGs showed differential expression in UC. Our findings underscore the role of disulfide stress in UC. Four UC-related disulfidptosis signature genes-SLC7A11, LRPPRC, NDUFS1, and CD2AP-were identified. Their relationships with immune infiltration in UC were analyzed using CIBERSORT, and their expression levels were validated by quantitative real-time PCR and western blotting. This study provides further insights into their potential functions and explores their links to immune infiltration in UC. In summary, disulfidptosis, as a type of programmed cell death, may significantly influence the pathogenesis of UC by modulating the homeostasis of the intestinal mucosal barrier.


Subject(s)
Colitis, Ulcerative , Intestinal Mucosa , Colitis, Ulcerative/genetics , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Animals , Humans , Mice , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Dextran Sulfate , Apoptosis/genetics , Male , Disease Models, Animal , Female , Gene Expression Profiling , Mice, Inbred C57BL , Computational Biology/methods
13.
Nat Commun ; 15(1): 5778, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987259

ABSTRACT

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


Subject(s)
Gastrointestinal Microbiome , Homeostasis , Inflammatory Bowel Diseases , Mice, Inbred C57BL , Gastrointestinal Microbiome/physiology , Animals , Humans , Mice , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/metabolism , Colitis/microbiology , Colitis/metabolism , Colitis/chemically induced , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice, Knockout , Ribonucleases/metabolism , Male , Feces/microbiology , Female , Intestines/microbiology , Antimicrobial Peptides/metabolism
14.
FASEB J ; 38(14): e23817, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39003633

ABSTRACT

Excessive apoptosis of intestinal epithelial cells leads to intestinal barrier dysfunction, which is not only one of the pathological features of inflammatory bowel disease (IBD) but also a therapeutic target. A natural plant extract, Ginkgetin (GK), has been reported to have anti-apoptotic activity, but its role in IBD is unknown. This study aimed to explore whether GK has anti-colitis effects and related mechanisms. An experimental colitis model induced by dextran sulfate sodium (DSS) was established, and GK was found to relieve colitis in DSS-induced mice as evidenced by improvements in weight loss, colon shortening, Disease Activity Index (DAI), macroscopic and tissue scores, and proinflammatory mediators. In addition, in DSS mice and TNF-α-induced colonic organoids, GK protected the intestinal barrier and inhibited intestinal epithelial cell apoptosis, by improving permeability and inhibiting the number of apoptotic cells and the expression of key apoptotic regulators (cleaved caspase 3, Bax and Bcl-2). The underlying mechanism of GK's protective effect was explored by bioinformatics, rescue experiments and molecular docking, and it was found that GK might directly target and activate EGFR, thereby interfering with PI3K/AKT signaling to inhibit apoptosis of intestinal epithelial cells in vivo and in vitro. In conclusion, GK inhibited intestinal epithelial apoptosis in mice with experimental colitis, at least in part, by activating EGFR and interfering with PI3K/AKT activation, explaining the underlying mechanism for ameliorating colitis, which may provide new options for the treatment of IBD.


Subject(s)
Apoptosis , Biflavonoids , Colitis , Dextran Sulfate , Epithelial Cells , ErbB Receptors , Intestinal Mucosa , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Animals , Apoptosis/drug effects , Mice , Proto-Oncogene Proteins c-akt/metabolism , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Colitis/pathology , ErbB Receptors/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Dextran Sulfate/toxicity , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Biflavonoids/pharmacology , Biflavonoids/therapeutic use , Male , Humans
15.
Int J Mol Sci ; 25(13)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-39000116

ABSTRACT

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


Subject(s)
Endotoxemia , Endotoxins , Gastrointestinal Microbiome , Neurodegenerative Diseases , Endotoxemia/metabolism , Endotoxemia/etiology , Humans , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/etiology , Animals , Endotoxins/metabolism , Microglia/metabolism , Microglia/pathology , Blood-Brain Barrier/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/microbiology , Inflammation/metabolism
16.
J Cell Mol Med ; 28(14): e18534, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39031467

ABSTRACT

Intestinal dysbiosis is believed to play a role in the development of necrotizing enterocolitis (NEC). The efficacy of JNK-inhibitory peptide (CPJIP) in treating NEC was assessed. Treatment with CPJIP led to a notable reduction in p-JNK expression in IEC-6 cells and NEC mice. Following LPS stimulation, the expression of RNA and protein of claudin-1, claudin-3, claudin-4 and occludin was significantly decreased, with this decrease being reversed by CPJIP administration, except for claudin-3, which remained consistent in NEC mice. Moreover, the expression levels of the inflammatory factors TNF-α, IL-1ß and IL-6 were markedly elevated, a phenomenon that was effectively mitigated by the addition of CPJIP in both IEC-6 cells and NEC mice. CPJIP administration resulted in improved survival rates, ameliorated microscopic intestinal mucosal injury, and increased the total length of the intestines and colon in NEC mice. Additionally, CPJIP treatment led to a reduction in serum concentrations of FD-4, D-lactate and DAO. Furthermore, our results revealed that CPJIP effectively inhibited intestinal cell apoptosis and promoted cell proliferation in the intestine. This study represents the first documentation of CPJIP's ability to enhance the expression of tight junction components, suppress inflammatory responses, and rescue intestinal cell fate by inhibiting JNK activation, ultimately mitigating intestinal severity. These findings suggest that CPJIP has the potential to serve as a promising candidate for the treatment of NEC.


Subject(s)
Apoptosis , Enterocolitis, Necrotizing , Inflammation , Intestinal Mucosa , Enterocolitis, Necrotizing/drug therapy , Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/pathology , Animals , Mice , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Inflammation/metabolism , Inflammation/drug therapy , Inflammation/pathology , Apoptosis/drug effects , Peptides/pharmacology , Disease Models, Animal , Cell Proliferation/drug effects , Mice, Inbred C57BL , Cell Line , Rats , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides , Intestinal Barrier Function
17.
Gut Microbes ; 16(1): 2377570, 2024.
Article in English | MEDLINE | ID: mdl-39034613

ABSTRACT

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


Subject(s)
Anti-Bacterial Agents , Bacteria , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Mucus , Humans , Gastrointestinal Microbiome/drug effects , Animals , Anti-Bacterial Agents/pharmacology , Mice , Mucus/metabolism , Mucus/microbiology , Bacteria/classification , Bacteria/genetics , Bacteria/drug effects , Bacteria/isolation & purification , Bacteria/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Male , Female , Feces/microbiology , Adult , Middle Aged , Akkermansia , Mice, Inbred C57BL , Colon/microbiology , Bacteroides fragilis/drug effects
18.
Toxicol In Vitro ; 99: 105888, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38950639

ABSTRACT

Irinotecan use is linked to the development of gastrointestinal toxicity and inflammation, or gastrointestinal mucositis. Selected phytocannabinoids have been ascribed anti-inflammatory effects in models of gastrointestinal inflammation, associated with maintaining epithelial barrier function. We characterised the mucoprotective capacity of the phytocannabinoids: cannabidiol, cannabigerol, cannabichromene and cannabidivarin in a cell-based model of intestinal epithelial stress occurring in mucositis. Transepithelial electrical resistance (TEER) was measured to determine changes in epithelial permeability in the presence of SN-38 (5 µM) or the pro-inflammatory cytokines TNFα and IL-1ß (each at 100 ng/mL), alone or with concomitant treatment with each of the phytocannabinoids (1 µM). The DCFDA assay was used to determine the ROS-scavenging ability of each phytocannabinoid following treatment with the lipid peroxidant tbhp (200 µM). Each phytocannabinoid provided significant protection against cytokine-evoked increases in epithelial permeability. Cannabidiol, cannabidivarin and cannabigerol were also able to significantly inhibit SN-38-evoked increases in permeability. None of the tested phytocannabinoids inhibited tbhp-induced ROS generation. These results highlight a novel role for cannabidiol, cannabidivarin and cannabigerol as inhibitors of SN-38-evoked increases in epithelial permeability and support the rationale for the further development of novel phytocannabinoids as supportive therapeutics in the management of irinotecan-associated mucositis.


Subject(s)
Cannabidiol , Cannabinoids , Intestinal Mucosa , Irinotecan , Permeability , Reactive Oxygen Species , Cannabinoids/pharmacology , Irinotecan/pharmacology , Permeability/drug effects , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Cannabidiol/pharmacology , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/metabolism , Interleukin-1beta/metabolism , Caco-2 Cells , Cytokines/metabolism , Intestinal Barrier Function
19.
Proc Natl Acad Sci U S A ; 121(29): e2402126121, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38980902

ABSTRACT

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.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Signal Transduction , Animals , Caenorhabditis elegans/immunology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/immunology , Signal Transduction/immunology , Intestines/immunology , Intestines/virology , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , RNA, Double-Stranded/metabolism , RNA, Double-Stranded/immunology , Immunity, Innate , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , RNA, Viral/immunology , RNA, Viral/metabolism , RNA, Viral/genetics
20.
PLoS One ; 19(7): e0307411, 2024.
Article in English | MEDLINE | ID: mdl-39024309

ABSTRACT

The microbiological environment and their corresponding secreted metabolite spectrum are an essential modulator of the enterocyte function, effecting the whole organism. Intestinal porcine jejunal epithelial cell line (IPEC-J2) is an established in vitro model for differentiation of enterocytes in different cell culture models. An improved oxygen supply seems to be the main reason for differentiation in an air-liquid-interface culture, but this has not yet been conclusively clarified. In this context, the nutrition of the cell and its influence on the metabolism is also of crucial importance. The interest in short-chain fatty acids (SCFAs) has grown steadily in recent years due to their clinical relevance in certain diseases such as multiple sclerosis and other inflammatory diseases, but not much is known of FFAR2 and FFAR3 (free fatty acid receptor 2 and 3) in pigs. We want to address the questions: 1. about the distribution of FFAR2 and FFAR3 in vivo and in vitro in sus scrofa 2. whether there is an influence of propionic acid, glucose content and cultivation on metabolism of enterocytes? The morphological analysis of FFAR2 and FFAR3 in vivo was investigated through immunostaining of frozen sections of the porcine gut segments jejunum, ileum and colon. Both receptors are expressed along the gut and were found in the smooth muscle cells of the tunica muscularis and lamina muscularis mucosae. Furthermore, a high expression of FFAR2 and a low expression of FFAR3 in the enteric nerve system was also observed in jejunum, ileum and colon of sus scrofa. In addition, FFAR2 and FFAR3 within the vessels was investigated. FFAR3 showed a strong expression on endothelial cells of veins and lymphatic vessels but was not detectable on arteries. Furthermore, we demonstrate for the first time, FFAR2 and FFAR3 in IPEC-J2 cells on RNA- and protein level, as well as with confocal microscopy. In addition, ENO1 and NDUFA4 were investigated on RNA-level in IPEC-J2 cells as 2 important genes, which play an essential role in metabolism. Here, NDUFA4 is detected in the model animal sus scrofa as well as in the porcine cell line IPEC-J2. A potential impact of propionic acid and/or glucose and/or cultivation method on the metabolism of the cells was tested with the Seahorse analyzer. Here, a significant higher ECAR was observed in the SMC than in the OCR. In summary, we were able to show that the cultivation system appears to have a greater influence than the medium composition or nutrition of the cells. However, this can be modulated by incubation time or combination of different SCFAs.


Subject(s)
Glucose , Propionates , Animals , Propionates/metabolism , Glucose/metabolism , Swine , Cell Line , Cell Culture Techniques/methods , Metabolome , Receptors, G-Protein-Coupled/metabolism , Jejunum/metabolism , Jejunum/cytology , Enterocytes/metabolism , Intestinal Mucosa/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...