Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 8.111
Filtrer
1.
Mediators Inflamm ; 2024: 3282679, 2024.
Article de Anglais | MEDLINE | ID: mdl-38962170

RÉSUMÉ

Ulcerative colitis (UC) is a chronic intestinal inflammatory disease with complex etiology. Interleukin-35 (IL-35), as a cytokine with immunomodulatory function, has been shown to have therapeutic effects on UC, but its mechanism is not yet clear. Therefore, we constructed Pichia pastoris stably expressing IL-35 which enables the cytokines to reach the diseased mucosa, and explored whether upregulation of T-cell protein tyrosine phosphatase (TCPTP) in macrophages is involved in the mechanisms of IL-35-mediated attenuation of UC. After the successful construction of engineered bacteria expressing IL-35, a colitis model was successfully induced by giving BALB/c mice a solution containing 3% dextran sulfate sodium (DSS). Mice were treated with Pichia/IL-35, empty plasmid-transformed Pichia (Pichia/0), or PBS by gavage, respectively. The expression of TCPTP in macrophages (RAW264.7, BMDMs) and intestinal tissues after IL-35 treatment was detected. After administration of Pichia/IL-35, the mice showed significant improvement in weight loss, bloody stools, and shortened colon. Colon pathology also showed that the inflammatory condition of mice in the Pichia/IL-35 treatment group was alleviated. Notably, Pichia/IL-35 treatment not only increases local M2 macrophages but also decreases the expression of inflammatory cytokine IL-6 in the colon. With Pichia/IL-35 treatment, the proportion of M1 macrophages, Th17, and Th1 cells in mouse MLNs were markedly decreased, while Tregs were significantly increased. In vitro experiments, IL-35 significantly promoted the expression of TCPTP in macrophages stimulated with LPS. Similarly, the mice in the Pichia/IL-35 group also expressed more TCPTP than that of the untreated group and the Pichia/0 group.


Sujet(s)
Interleukines , Macrophages , Souris de lignée BALB C , Animaux , Souris , Interleukines/métabolisme , Macrophages/métabolisme , Cellules RAW 264.7 , Colite/induit chimiquement , Colite/métabolisme , Sulfate dextran , Modèles animaux de maladie humaine , Rectocolite hémorragique/métabolisme , Rectocolite hémorragique/induit chimiquement , Mâle , Régulation positive , Saccharomycetales
2.
Gut Microbes ; 16(1): 2361493, 2024.
Article de Anglais | MEDLINE | ID: mdl-38958039

RÉSUMÉ

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


Sujet(s)
Bactéries , Colite , Microbiome gastro-intestinal , Muqueuse intestinale , Oxygène , Colite/microbiologie , Colite/induit chimiquement , Colite/métabolisme , Animaux , Humains , Oxygène/métabolisme , Bactéries/métabolisme , Bactéries/classification , Bactéries/isolement et purification , Bactéries/génétique , Souris , Muqueuse intestinale/métabolisme , Muqueuse intestinale/microbiologie , Fèces/microbiologie , Souris de lignée C57BL , Sulfate dextran , Côlon/microbiologie , Côlon/métabolisme , Mâle
3.
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
4.
FASEB J ; 38(13): e23791, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38963340

RÉSUMÉ

Inflammatory bowel disease (IBD) is a kind of recurrent inflammatory disorder of the intestinal tract. The purpose of this study was to investigate the effects of Weissella paramesenteroides NRIC1542 on colitis in mice. A colitis model was induced by adding 1.5% DSS to sterile distilled water for seven consecutive days. During this process, mice were administered different concentrations of W. paramesenteroides NRIC1542. Colitis was assessed by DAI, colon length and hematoxylin-eosin staining of colon sections. The expressions of NF-κB signaling proteins and the tight junction proteins ZO-1 and occludin were detected by western blotting, and the gut microbiota was analyzed by 16S rDNA. The results showed that W. paramesenteroides NRIC1542 significantly reduced the degree of pathological tissue damage and the levels of TNF-α and IL-1ß in colonic tissue, inhibiting the NF-κB signaling pathway and increasing the expression of SIRT1, ZO-1 and occludin. In addition, W. paramesenteroides NRIC1542 can modulate the structure of the gut microbiota, characterized by increased relative abundance of Muribaculaceae_unclassified, Paraprevotella, Prevotellaceae_UCG_001 and Roseburia, and decrease the relative abundance of Akkermansia and Alloprevotella induced by DSS. The above results suggested that W. paramesenteroides NRIC1542 can protect against DSS-induced colitis in mice through anti-inflammatory, intestinal barrier maintenance and flora modulation.


Sujet(s)
Colite , Sulfate dextran , Microbiome gastro-intestinal , Facteur de transcription NF-kappa B , Transduction du signal , Sirtuine-1 , Weissella , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Sirtuine-1/métabolisme , Souris , Colite/induit chimiquement , Colite/métabolisme , Colite/microbiologie , Sulfate dextran/toxicité , Transduction du signal/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/métabolisme , Weissella/métabolisme , Mâle , Probiotiques/pharmacologie
5.
Pharmacol Res Perspect ; 12(4): e1234, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38961539

RÉSUMÉ

The association of hormonal contraception with increased risk of inflammatory bowel disease (IBD) observed in females suggests involvement of ovarian hormones, such as estradiol, and the estrogen receptors in the progression of intestinal inflammation. Here, we investigated the effects of prophylactic SERM2 and estradiol supplementation in dextran sulfate sodium-induced colitis using mice with intact ovaries and ovariectomized (OVX) female mice. We found that graded colitis score was threefold reduced in the OVX mice, compared to mice with intact ovaries. Estradiol supplementation, however, aggravated the colitis in OVX mice, increasing the colitis score to a similar level than what was observed in the intact mice. Further, we observed that immune infiltration and gene expression of inflammatory interleukins Il1b, Il6, and Il17a were up to 200-fold increased in estradiol supplemented OVX colitis mice, while a mild but consistent decrease was observed by SERM2 treatment in intact animals. Additionally, cyclo-oxygenase 2 induction was increased in the colon of colitis mice, in correlation with increased serum estradiol levels. Measured antagonist properties of SERM2, together with the other results presented here, indicates an exaggerating role of ERα signaling in colitis. Our results contribute to the knowledge of ovarian hormone effects in colitis and encourage further research on the potential use of ER antagonists in the colon, in order to alleviate inflammation.


Sujet(s)
Colite , Sulfate dextran , Oestradiol , Récepteur alpha des oestrogènes , Ovariectomie , Animaux , Femelle , Récepteur alpha des oestrogènes/métabolisme , Colite/induit chimiquement , Colite/métabolisme , Colite/traitement médicamenteux , Souris , Oestradiol/pharmacologie , Oestradiol/sang , Souris de lignée C57BL , Oestrogènes/pharmacologie , Cyclooxygenase 2/métabolisme , Modèles animaux de maladie humaine , Interleukine-17/métabolisme , Côlon/anatomopathologie , Côlon/effets des médicaments et des substances chimiques , Côlon/métabolisme , Interleukine-6/métabolisme , Interleukine-1 bêta/métabolisme
6.
Curr Pharm Des ; 30(17): 1377, 2024 May 17.
Article de Anglais | MEDLINE | ID: mdl-38975683

RÉSUMÉ

A typographical error appeared in the title of the article "Mechanism of HSP90 Inhibitor in the Treatment of DSS-induced Colitis in Mice by Inhibiting MAPK Pathway and Synergistic Effect of Compound Sophora Decoction", published in Current Pharmaceutical Design, 2022; 28(42): 3456-3468 [1]. Details of the error and a correction are provided below. Original: Mechanism of HSP90 Inhibitor in the Treatment of DSS-induced Colitis in Mice by Inhibiting MAPK Pathway and Synergistic Effect of Compound Sophora Decoction Corrected: Mechanism of HSP90 Inhibitor in the Treatment of DSS-induced Colitis in Mice by Inhibiting MAPK Pathway and Synergistic Effect of Compound Sophorae Decoction We regret the error and apologize to readers. The original article can be found online at: https://www.eurekaselect.com/article/127740.


Sujet(s)
Colite , Sulfate dextran , Protéines du choc thermique HSP90 , Animaux , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Protéines du choc thermique HSP90/métabolisme , Souris , Colite/traitement médicamenteux , Colite/induit chimiquement , Sophora/composition chimique , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques
7.
Sci Rep ; 14(1): 15706, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38977770

RÉSUMÉ

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.


Sujet(s)
Colite , Coumarines , Muqueuse intestinale , Facteur-2 apparenté à NF-E2 , Récepteurs à hydrocarbure aromatique , Animaux , Facteur-2 apparenté à NF-E2/métabolisme , Récepteurs à hydrocarbure aromatique/métabolisme , Souris , Muqueuse intestinale/métabolisme , Coumarines/pharmacologie , Colite/métabolisme , Colite/induit chimiquement , Mucine-2/métabolisme , Mucine-2/génétique , Humains , Côlon/métabolisme , Souris de lignée C57BL , Transduction du signal/effets des médicaments et des substances chimiques , Mâle , Microbiome gastro-intestinal , Souris knockout , Sulfate dextran , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique ,
8.
Sci Rep ; 14(1): 15705, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38977802

RÉSUMÉ

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.


Sujet(s)
Rectocolite hémorragique , Muqueuse intestinale , Rectocolite hémorragique/génétique , Rectocolite hémorragique/induit chimiquement , Rectocolite hémorragique/métabolisme , Rectocolite hémorragique/anatomopathologie , Animaux , Humains , Souris , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Sulfate dextran , Apoptose/génétique , Mâle , Modèles animaux de maladie humaine , Femelle , Analyse de profil d'expression de gènes , Souris de lignée C57BL , Biologie informatique/méthodes
9.
Food Funct ; 15(14): 7641-7657, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38953279

RÉSUMÉ

Edible plant-derived nanovesicles (ePDNs) have shown potential as a non-pharmacological option for inflammatory bowel disease (IBD) by maintaining gut health and showing anti-inflammatory effects. However, the effects of Allium tuberosum-derived nanovesicles (ADNs) on colitis have not been studied to date. Here, we extracted exosome-like nanovesicles from Allium tuberosum and investigated whether they have an anti-inflammatory effect in RAW 264.7 cells and colitis mice. The results showed that ADNs reduced the elevated levels of inflammatory factors such as IL-1ß, IL-6, TNF-α, and NF-κB pathway-related proteins as a consequence of lipopolysaccharide (LPS) stimulation in RAW 264.7 cells. Furthermore, our mouse experiments demonstrated that ADNs could ameliorate dextran sulfate sodium (DSS)-induced colitis symptoms (e.g., increased disease activity index score, intestinal permeability, and histological appearance). Additionally, ADNs counteracted DSS-induced colitis by downregulating the expression of serum amyloid A (SAA), IL-1ß, IL-6, and TNF-α and increasing the expression of tight junction proteins (ZO-1 and occludin) and the anti-inflammatory cytokine IL-10. 16S rRNA gene sequencing showed that ADN intervention restored the gut microbial composition, which was similar to that of the DSS non-treated group, by decreasing the ratio of Firmicutes to Bacteroidetes and the relative abundance of Proteobacteria. Furthermore, ADNs induced acetic acid production along with an increase in the abundance of Lactobacillus. Overall, our findings suggest that ADN supplementation has a crucial role in maintaining gut health and is a novel preventive therapy for IBD.


Sujet(s)
Anti-inflammatoires , Colite , Sulfate dextran , Microbiome gastro-intestinal , Animaux , Souris , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Colite/induit chimiquement , Sulfate dextran/effets indésirables , Anti-inflammatoires/pharmacologie , Cellules RAW 264.7 , Souris de lignée C57BL , Mâle , Modèles animaux de maladie humaine , Facteur de transcription NF-kappa B/métabolisme , Extraits de plantes/pharmacologie , Extraits de plantes/administration et posologie
10.
Int J Mol Sci ; 25(13)2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-39000289

RÉSUMÉ

Inflammatory bowel disease (IBD) is an immunologically complex disorder involving genetic, microbial, and environmental risk factors. Its global burden has continued to rise since industrialization, with epidemiological studies suggesting that ambient particulate matter (PM) in air pollution could be a contributing factor. Prior animal studies have shown that oral PM10 exposure promotes intestinal inflammation in a genetic IBD model and that PM2.5 inhalation exposure can increase intestinal levels of pro-inflammatory cytokines. PM10 and PM2.5 include ultrafine particles (UFP), which have an aerodynamic diameter of <0.10 µm and biophysical and biochemical properties that promote toxicity. UFP inhalation, however, has not been previously studied in the context of murine models of IBD. Here, we demonstrated that ambient PM is toxic to cultured Caco-2 intestinal epithelial cells and examined whether UFP inhalation affected acute colitis induced by dextran sodium sulfate and 2,4,6-trinitrobenzenesulfonic acid. C57BL/6J mice were exposed to filtered air (FA) or various types of ambient PM reaerosolized in the ultrafine size range at ~300 µg/m3, 6 h/day, 3-5 days/week, starting 7-10 days before disease induction. No differences in weight change, clinical disease activity, or histology were observed between the PM and FA-exposed groups. In conclusion, UFP inhalation exposure did not exacerbate intestinal inflammation in acute, chemically-induced colitis models.


Sujet(s)
Colite , Sulfate dextran , Souris de lignée C57BL , Matière particulaire , Acide 2,4,6-trinitro-benzènesulfonique , Matière particulaire/toxicité , Animaux , Colite/induit chimiquement , Colite/anatomopathologie , Souris , Humains , Sulfate dextran/toxicité , Cellules Caco-2 , Acide 2,4,6-trinitro-benzènesulfonique/toxicité , Acide 2,4,6-trinitro-benzènesulfonique/effets indésirables , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/anatomopathologie , Muqueuse intestinale/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/anatomopathologie , Cellules épithéliales/métabolisme , Modèles animaux de maladie humaine , Mâle , Taille de particule
11.
Drug Des Devel Ther ; 18: 2847-2868, 2024.
Article de Anglais | MEDLINE | ID: mdl-39006190

RÉSUMÉ

Purpose: This study aimed to delineate the molecular processes underlying the therapeutic effects of berberine on UC by employing network pharmacology tactics, molecular docking, and dynamic simulations supported by empirical validations both in vivo and in vitro. Patients and Methods: We systematically screened potential targets and relevant pathways affected by berberine for UC treatment from comprehensive databases, including GeneCards, DisGeNET, and GEO. Molecular docking and simulation protocols were used to assess the interaction stability between berberine and its principal targets. The predictions were validated using both a DSS-induced UC mouse model and a lipopolysaccharide (LPS)-stimulated NCM460 cellular inflammation model. Results: Network pharmacology analysis revealed the regulatory effect of the TLR4/NF-κB/HIF-1α pathway in the ameliorative action of berberine in UC. Docking and simulation studies predicted the high-affinity interactions of berberine with pivotal targets: TLR4, NF-κB, HIF-1α, and the HIF inhibitor KC7F2. Moreover, in vivo analyses demonstrated that berberine attenuates clinical severity, as reflected by decreased disease activity index (DAI) scores, reduced weight loss, and mitigated intestinal inflammation in DSS-challenged mice. These outcomes include suppression of the proinflammatory cytokines IL-6 and TNF-α and downregulation of TLR4/NF-κB/HIF-1α mRNA and protein levels. Correspondingly, in vitro findings indicate that berberine decreases cellular inflammatory injury and suppresses TLR4/NF-κB/HIF-1α signaling, with notable effectiveness similar to that of the HIF-1α inhibitor KC7F2. Conclusion: Through network pharmacology analysis and experimental substantiation, this study confirmed that berberine enhances UC treatment outcomes by inhibiting the TLR4/NF-κB/HIF-1α axis, thereby mitigating inflammatory reactions and improving colonic pathology.


Sujet(s)
Berbérine , Rectocolite hémorragique , Biologie informatique , Sous-unité alpha du facteur-1 induit par l'hypoxie , Facteur de transcription NF-kappa B , Récepteur de type Toll-4 , Berbérine/pharmacologie , Berbérine/composition chimique , Récepteur de type Toll-4/métabolisme , Récepteur de type Toll-4/antagonistes et inhibiteurs , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/antagonistes et inhibiteurs , Facteur de transcription NF-kappa B/métabolisme , Facteur de transcription NF-kappa B/antagonistes et inhibiteurs , Animaux , Souris , Humains , Rectocolite hémorragique/traitement médicamenteux , Rectocolite hémorragique/métabolisme , Rectocolite hémorragique/induit chimiquement , Rectocolite hémorragique/anatomopathologie , Simulation de docking moléculaire , Lipopolysaccharides/pharmacologie , Lipopolysaccharides/antagonistes et inhibiteurs , Mâle , Sulfate dextran , Souris de lignée C57BL , Modèles animaux de maladie humaine , Pharmacologie des réseaux
12.
Bull Exp Biol Med ; 177(1): 162-168, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38960963

RÉSUMÉ

In adult male C57BL/6 mice with high (HR) and low (LR) resistance to hypoxia, morphological features of colon tumors and blood parameters were evaluated 70 days after intraperitoneal injection of azoxymethane and subsequent consumption of 3 cycles of dextran sulfate sodium. On macroscopic analysis, tumors were found in the distal colon in 35% (7 of 20 animals) of HR and 31% (4 of 13 animals) of LR animals. Microscopic analysis of the distal colon revealed tumors in 75% (15 of 20 animals) of HR and 69% (9 of 13 animals) of LR mice. The tumors were presented by areas of glandular intraepithelial neoplasia and adenocarcinomas; the incidence and the area of the tumors did not differ in groups of HR and LR mice. The number of neuroendocrine and goblet cells in the distal colon mucosa in the areas of tumors was similar in the compared groups. However, in both HR and LR mice of the experimental groups, the content of goblet cells in tumors was lower and the content of endocrine cells was higher than in the corresponding control groups. In the peripheral blood, the erythrocyte count and hemoglobin content decreased in HR and LR mice of the experimental groups; the relative number of monocytes increased only in HR mice and the absolute number of lymphocytes and monocytes decreased in LR mice. Thus, 70 days after azoxymethane administration and dextran sulfate sodium consumption, the tumors in mice were presented by glandular intraepithelial neoplasia and adenocarcinomas, and their incidence and area did not differ between animals with different tolerance to hypoxia.


Sujet(s)
Adénocarcinome , Oxyde de diméthyl-diazène , Tumeurs du côlon , Sulfate dextran , Souris de lignée C57BL , Animaux , Souris , Tumeurs du côlon/anatomopathologie , Tumeurs du côlon/induit chimiquement , Tumeurs du côlon/métabolisme , Mâle , Sulfate dextran/toxicité , Oxyde de diméthyl-diazène/toxicité , Adénocarcinome/anatomopathologie , Adénocarcinome/induit chimiquement , Adénocarcinome/métabolisme , Hypoxie/anatomopathologie , Côlon/anatomopathologie , Cellules caliciformes/anatomopathologie , Cellules caliciformes/métabolisme , Muqueuse intestinale/anatomopathologie , Hémoglobines/métabolisme , Monocytes/anatomopathologie , Monocytes/métabolisme , Numération des érythrocytes
13.
BMC Gastroenterol ; 24(1): 223, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38992591

RÉSUMÉ

Ulcerative colitis (UC) is a persistent inflammatory condition that specifically targets the colon and rectum. Existing therapies fail to adequately address the clinical requirements of people suffering from this ailment. Despite the acknowledged potential of nanomedicines in the field of anti-inflammatory treatment, their widespread use in clinical settings is impeded by their expensive nature and the uncertainty surrounding their safety profiles. This study illustrates that two naturally occurring phytochemicals, Costunolide (COS) and Glycyrrhizic acid (GA), form carrier-free, multifunctional spherical nanoparticles (NPs) through noncovalent interactions, such as π-π stacking and hydrogen bonding. The COS-GA NPs displayed a synergistic anti-inflammatory effect, providing much more evidently improved therapeutic benefits for dextran sodium sulfate (DSS)-induced UC mice due to more effective reduction in inflammation and oxidative stress than did equal dosages of COS or GA used alone. In addition, COS-GA NPs have biocompatibility and biosafety properties unique to them. This study will serve as affirmation of the potential of COS-GA NPs as innovative natural anti-inflammatory and antioxidant activities and also such agents as drug discovery in UC, leading possibly to better outcomes in people living with this disabling condition.


Sujet(s)
Anti-inflammatoires , Rectocolite hémorragique , Sulfate dextran , Acide glycyrrhizique , Nanoparticules , Rectocolite hémorragique/traitement médicamenteux , Animaux , Acide glycyrrhizique/usage thérapeutique , Acide glycyrrhizique/pharmacologie , Anti-inflammatoires/usage thérapeutique , Souris , Stress oxydatif/effets des médicaments et des substances chimiques , Antioxydants/pharmacologie , Modèles animaux de maladie humaine , Mâle , Synergie des médicaments , Sesquiterpènes
14.
FASEB J ; 38(14): e23817, 2024 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-39003633

RÉSUMÉ

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.


Sujet(s)
Apoptose , Biflavonoïdes , Colite , Sulfate dextran , Cellules épithéliales , Récepteurs ErbB , Muqueuse intestinale , Souris de lignée C57BL , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Animaux , Apoptose/effets des médicaments et des substances chimiques , Souris , Protéines proto-oncogènes c-akt/métabolisme , Colite/induit chimiquement , Colite/traitement médicamenteux , Colite/métabolisme , Colite/anatomopathologie , Récepteurs ErbB/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Muqueuse intestinale/métabolisme , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/anatomopathologie , Sulfate dextran/toxicité , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Biflavonoïdes/pharmacologie , Biflavonoïdes/usage thérapeutique , Mâle , Humains
15.
FASEB J ; 38(11): e23721, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38822662

RÉSUMÉ

Schistosome infection and schistosome-derived products have been implicated in the prevention and alleviation of inflammatory bowel disease by manipulating the host immune response, whereas the role of gut microbiota in this protective effect remains poorly understood. In this study, we found that the intraperitoneal immunization with Schistosoma japonicum eggs prior to dextran sulfate sodium (DSS) application significantly ameliorated the symptoms of DSS-induced acute colitis, which was characterized by higher body weight, lower disease activity index score and macroscopic inflammatory scores. We demonstrated that the immunomodulatory effects of S. japonicum eggs were accompanied by an influence on gut microbiota composition, abundance, and diversity, which increased the abundance of genus Turicibacter, family Erysipelotrichaceae, phylum Firmicutes, and decreased the abundance of genus Odoribacter, family Marinifilaceae, order Bacteroidales, class Bacteroidia, phylum Bacteroidota. In addition, Lactobacillus was identified as a biomarker that distinguishes healthy control mice from DSS-induced colitis mice. The present study revealed the importance of the gut microbiota in S. japonicum eggs exerting protective effects in an experimental ulcerative colitis (UC) model, providing an alternative strategy for the discovery of UC prevention and treatment drugs.


Sujet(s)
Rectocolite hémorragique , Sulfate dextran , Modèles animaux de maladie humaine , Microbiome gastro-intestinal , Schistosoma japonicum , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Rectocolite hémorragique/microbiologie , Rectocolite hémorragique/immunologie , Souris , Schistosoma japonicum/immunologie , Sulfate dextran/toxicité , Femelle , Immunisation/méthodes , Ovule , Souris de lignée C57BL
16.
Int J Mol Sci ; 25(12)2024 Jun 16.
Article de Anglais | MEDLINE | ID: mdl-38928319

RÉSUMÉ

Matrine (MT) possesses anti-inflammatory, anti-allergic and antioxidative properties. However, the impact and underlying mechanisms of matrine on colitis are unclear. The purpose of this research was to examine the protective impact and regulatory mechanism of matrine on dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) in mice. MT alleviated DSS-induced UC by inhibiting weight loss, relieving colon shortening and reducing the disease activity index (DAI). Moreover, DSS-induced intestinal injury and the number of goblet cells were reversed by MT, as were alterations in the expression of zonula occludens-1 (ZO-1) and occludin in colon. Simultaneously, matrine not only effectively restored DSS-induced oxidative stress in colonic tissues but also reduced the production of inflammatory cytokines. Furthermore, MT could treat colitis mice by regulating the regulatory T cell (Treg)/T helper 17 (Th17) cell imbalance. We observed further evidence that MT alleviated the decrease in intestinal flora diversity, reduced the proportion of Firmicutes and Bacteroidetes, decreased the proportion of Proteobacteria and increased the relative abundance of Lactobacillus and Akkermansia in colitis mice. In conclusion, these results suggest that MT may mitigate DSS-induced colitis by enhancing the colon barrier integrity, reducing the Treg/Th17 cell imbalance, inhibiting intestinal inflammation, modulating oxidative stress and regulating the gut microbiota. These findings provide strong evidence for the development and application of MT as a dietary treatment for UC.


Sujet(s)
Alcaloïdes , Sulfate dextran , Microbiome gastro-intestinal , , Stress oxydatif , Quinolizines , Lymphocytes T régulateurs , Animaux , Alcaloïdes/pharmacologie , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Stress oxydatif/effets des médicaments et des substances chimiques , Quinolizines/pharmacologie , Quinolizines/usage thérapeutique , Souris , Lymphocytes T régulateurs/métabolisme , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Lymphocytes T régulateurs/immunologie , Mâle , Colite/induit chimiquement , Colite/traitement médicamenteux , Colite/métabolisme , Colite/microbiologie , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Inflammation/anatomopathologie , Protéine-1 de la zonula occludens/métabolisme , Côlon/anatomopathologie , Côlon/métabolisme , Côlon/effets des médicaments et des substances chimiques , Côlon/microbiologie , Cellules Th17/effets des médicaments et des substances chimiques , Cellules Th17/métabolisme , Cellules Th17/immunologie , Modèles animaux de maladie humaine , Cytokines/métabolisme , Souris de lignée C57BL , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Rectocolite hémorragique/traitement médicamenteux , Rectocolite hémorragique/induit chimiquement , Rectocolite hémorragique/microbiologie , Rectocolite hémorragique/métabolisme , Rectocolite hémorragique/anatomopathologie , Occludine/métabolisme
17.
Commun Biol ; 7(1): 731, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38879692

RÉSUMÉ

Ulcerative colitis (UC) is a significant inflammatory bowel disease caused by an abnormal immune response to gut microbes. However, there are still gaps in our understanding of how immune and metabolic changes specifically contribute to this disease. Our research aims to address this gap by examining mouse colons after inducing ulcerative colitis-like symptoms. Employing single-cell RNA-seq and 16 s rRNA amplicon sequencing to analyze distinct cell clusters and microbiomes in the mouse colon at different time points after induction with dextran sodium sulfate. We observe a significant reduction in epithelial populations during acute colitis, indicating tissue damage, with a partial recovery observed in chronic inflammation. Analyses of cell-cell interactions demonstrate shifts in networking patterns among different cell types during disease progression. Notably, macrophage phenotypes exhibit diversity, with a pronounced polarization towards the pro-inflammatory M1 phenotype in chronic conditions, suggesting the role of macrophage heterogeneity in disease severity. Increased expression of Nampt and NOX2 complex subunits in chronic UC macrophages contributes to the inflammatory processes. The chronic UC microbiome exhibits reduced taxonomic diversity compared to healthy conditions and acute UC. The study also highlights the role of T cell differentiation in the context of dysbiosis and its implications in colitis progression, emphasizing the need for targeted interventions to modulate the inflammatory response and immune balance in colitis.


Sujet(s)
Rectocolite hémorragique , Sulfate dextran , Microbiome gastro-intestinal , Macrophages , Analyse sur cellule unique , Animaux , Rectocolite hémorragique/microbiologie , Rectocolite hémorragique/immunologie , Rectocolite hémorragique/génétique , Rectocolite hémorragique/induit chimiquement , Rectocolite hémorragique/anatomopathologie , Macrophages/immunologie , Macrophages/microbiologie , Macrophages/métabolisme , Sulfate dextran/toxicité , Sulfate dextran/effets indésirables , Souris , RNA-Seq , Souris de lignée C57BL , Modèles animaux de maladie humaine , Codage à barres de l'ADN pour la taxonomie , ARN ribosomique 16S/génétique , Mâle , Analyse de l'expression du gène de la cellule unique
18.
J Mater Sci Mater Med ; 35(1): 29, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38884680

RÉSUMÉ

Mesoporous silica nanoparticles (MSNPs) coated by chitosan (CS) were shown to be a proper candidate as a carrier for drug delivery purposes. However, choosing the suitable drug-containing complexes to be applied on MSNPs-CS is of much greater importance to evaluate the possible candidate for an efficient combination of cell viability, drug release kinetics, and atherosclerosis prevention. In this regard, this study concentrates on the synthesis and assessment of coated MSNPs-CS designed for drug delivery purposes. The MSNPs are coated with polyelectrolyte complexes (PEC) composed of CS and dextran sulfate (MSNPs-CS-DX), serving as a versatile drug carrier with favorable biological characteristics. CS-DX is applied to MSNPs without requiring complex or multi-step synthesis procedures. Rosuvastatin, a cholesterol-lowering medication, is chosen for its therapeutic relevance. Additionally, CS-DX is found to relatively impede the uptake of low-density lipoproteins (LDLs) by macrophages, enhancing their potential therapeutic utility. FTIR pattern, FESEM, and TEM images prove MSNPs-CS-DX formation. DLS measurement demonstrates the average particle size of 110 nm for MSNPs, with the combined thickness of CS and DX layers ranging from 10 to 15 nm. BET test is carried out to evaluate the pore size and porosity of structure, showing outstanding results that cause an entrapment efficiency of 57% for MSNPs-CS-DX. Furthermore, the findings demonstrate the pH sensitivity of MSNPs-CS-DX on drug release kinetics. Notably, the CS-DX layer exhibits a significant enhancement in cell viability of human umbilical vein endothelial cells (HUVEC) by approximately 24% within a 24 h timeframe compared to MSNPs lacking CS-DX.


Sujet(s)
Chitosane , Sulfate dextran , Systèmes de délivrance de médicaments , Silice , Chitosane/composition chimique , Silice/composition chimique , Concentration en ions d'hydrogène , Sulfate dextran/composition chimique , Porosité , Humains , Vecteurs de médicaments/composition chimique , Nanoparticules/composition chimique , Taille de particule , Libération de médicament , Survie cellulaire/effets des médicaments et des substances chimiques , Animaux , Rosuvastatine de calcium/pharmacocinétique , Rosuvastatine de calcium/composition chimique , Rosuvastatine de calcium/administration et posologie , Rosuvastatine de calcium/pharmacologie , Souris , Matériaux revêtus, biocompatibles/composition chimique , Lipoprotéines LDL , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Cellules RAW 264.7
19.
J Vis Exp ; (207)2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38884498

RÉSUMÉ

Ulcerative colitis (UC) is a chronic immune-mediated disease that affects the entire colon and rectum with a relapsing and remitting course, causing lifelong morbidity. When medical treatment is ineffective, especially in cases of massive gastrointestinal bleeding, perforation, toxic megacolon, or carcinogenesis, surgery becomes the last line of defense to cure UC. Total colorectal resection and ileal pouch-anal anastomosis (IPAA) offer the best chance for long-term treatment. Pouchitis is the most common and troublesome postoperative complication. In this investigation, microsurgery is employed to create an ileal pouch model in experimental rats via IPAA surgery. Subsequently, a sustained rat model of pouchitis is established by inducing inflammation of the ileal pouch with dextran sulfate sodium (DSS). The successful establishment of rat pouchitis is validated through analysis of postoperative general status, weight, food and water intake, fecal data, as well as pouch tissue pathology, immunohistochemistry, and inflammatory factor analysis. This experimental animal model of pouchitis provides a foundation for studying the pathogenesis and treatment of the condition.


Sujet(s)
Poches coliques , Sulfate dextran , Modèles animaux de maladie humaine , Pochite , Proctocolectomie restauratrice , Animaux , Pochite/étiologie , Rats , Proctocolectomie restauratrice/méthodes , Proctocolectomie restauratrice/effets indésirables , Poches coliques/effets indésirables , Complications postopératoires/étiologie , Mâle , Anastomose chirurgicale/méthodes , Anastomose chirurgicale/effets indésirables
20.
Nutrients ; 16(11)2024 May 22.
Article de Anglais | MEDLINE | ID: mdl-38892496

RÉSUMÉ

The imbalance of gut microbiota is an important factor leading to inflammatory bowel disease (IBD). Diffusible signal factor (DSF) is a novel quorum-sensing signal that regulates bacterial growth, metabolism, pathogenicity, and host immune response. This study aimed to explore the therapeutic effect and underlying mechanisms of DSF in a zebrafish colitis model induced by sodium dextran sulfate (DSS). The results showed that intake of DSF can significantly improve intestinal symptoms in the zebrafish colitis model, including ameliorating the shortening of the intestine, reducing the increase in the goblet cell number, and restoring intestinal pathological damage. DSF inhibited the upregulation of inflammation-related genes and promoted the expression of claudin1 and occludin1 to protect the tightness of intestinal tissue. The gut microbiome analysis demonstrated that DSF treatment helped the gut microbiota of the zebrafish colitis model recover to normal at the phylum and genus levels, especially in terms of pathogenic bacteria; DSF treatment downregulated the relative abundance of Aeromonas hydrophila and Staphylococcus aureus, and it was confirmed in microbiological experiments that DSF could effectively inhibit the colonization and infection of these two pathogens in the intestine. This study suggests that DSF can alleviate colitis by inhibiting the proliferation of intestinal pathogens and inflammatory responses in the intestine. Therefore, DSF has the potential to become a dietary supplement that assists in the antibiotic and nutritional treatment of IBD.


Sujet(s)
Colite , Sulfate dextran , Modèles animaux de maladie humaine , Microbiome gastro-intestinal , Détection du quorum , Danio zébré , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Colite/induit chimiquement , Colite/microbiologie , Colite/traitement médicamenteux , Détection du quorum/effets des médicaments et des substances chimiques , Intestins/microbiologie , Aeromonas hydrophila , Inflammation , Staphylococcus aureus/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...