Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.899
Filtrer
1.
AAPS J ; 26(5): 90, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39107477

RÉSUMÉ

Changes to blood-brain barrier structure and function may affect the delivery of drugs into the brain. It is worthwhile to exploring more study on how the blood-brain barrier changes in structure and function and how that affects drug transport in high-altitude hypoxic environment. The DIA high-throughput sequencing technique indicate that the rats blood-brain barrier has been identified to have 7252 proteins overall and 8 tight junction proteins, among which Claudin-7 was a plateau-specific tight junction protein under high-altitude hypoxia, and based on the interaction network study, 2421 proteins are found to interact with one another, with ZO-1 being the primary target. The results of the projected gene function analysis demonstrated that changes in tight junction proteins are related to the control of TRP channels by inflammatory mediators, the wnt signaling pathway, the ABC transporter system, and drug metabolism-CYP450 enzyme regulation. Additionally, the electron microscopy, the Evans blue combination with confocal laser scanning microscopy, and the Western Blot and RT-qPCR revealed that high-altitude hypoxic environment induces blood-brain barrier tight junctions to open, blood-brain barrier permeability increases, ZO-1, Occludin, Claudin-5 protein and mRNA expression decreased. Our research implies that structural and functional alterations in the blood-brain barrier induced by high altitude hypoxia may impact drug transport inside the central nervous system, and that drug transporters and drug-metabolizing enzymes may be key players in this process.


Sujet(s)
Barrière hémato-encéphalique , Protéines de la jonction serrée , Animaux , Barrière hémato-encéphalique/métabolisme , Protéines de la jonction serrée/métabolisme , Protéines de la jonction serrée/génétique , Rats , Hypoxie/métabolisme , Mâle , Altitude , Rat Sprague-Dawley , Transport biologique , Perméabilité , Jonctions serrées/métabolisme
2.
Cells ; 13(15)2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-39120333

RÉSUMÉ

The yak (Bos grunniens) is a valuable livestock animal endemic to the Qinghai-Tibet Plateau in China with low reproductive rates. Cryptorchidism is one of the primary causes of infertility in male yaks. Compared with normal testes, the tight junctions (TJs) of Sertoli cells (SCs) and the integrity of the blood-testis barrier (BTB) in cryptorchidism are both disrupted. MicroRNAs are hairpin-derived RNAs of about 19-25 nucleotides in length and are involved in a variety of biological processes. Numerous studies have shown the involvement of microRNAs in the reproductive physiology of yak. In this study, we executed RNA sequencing (RNA-seq) to describe the expression profiles of mRNAs and microRNAs in yaks with normal testes and cryptorchidism to identify differentially expressed genes. GO and KEGG analyses were used to identify the biological processes and signaling pathways which the target genes of the differentially expressed microRNAs primarily engaged. It was found that novel-m0230-3p is an important miRNA that significantly differentiates between cryptorchidism and normal testes, and it is down-regulated in cryptorchidism with p < 0.05. Novel-m0230-3p and its target gene CSF1 both significantly contribute to the regulation of cell adhesion and tight junctions. The binding sites of novel-m0230-3p with CSF1 were validated by a dual luciferase reporter system. Then, mimics and inhibitors of novel-m0230-3p were transfected in vitro into SCs, respectively. A further analysis using qRT-PCR, immunofluorescence (IF), and Western blotting confirmed that the expression of cell adhesion and tight-junction-related proteins Occludin and ZO-1 both showed changes. Specifically, both the mRNA and protein expression levels of Occludin and ZO-1 in SCs decreased after transfection with the novel-m0230-3p mimics, while they increased after transfection with the inhibitors, with p < 0.05. These were achieved via the CSF1/CSF1R/Ras signaling pathway. In summary, our findings indicate a negative miRNA-mRNA regulatory network involving the CSF1/CSF1R/Ras signaling pathway in yak SCs. These results provide new insights into the molecular mechanisms of CSF1 and suggest that novel-m0230-3p and its target protein CSF1 could be used as potential therapeutic targets for yak cryptorchidism.


Sujet(s)
Barrière hématotesticulaire , microARN , Transduction du signal , Jonctions serrées , Animaux , Mâle , Barrière hématotesticulaire/métabolisme , Jonctions serrées/métabolisme , microARN/génétique , microARN/métabolisme , Bovins , Cellules de Sertoli/métabolisme , Testicule/métabolisme , Régulation de l'expression des gènes
3.
Mar Drugs ; 22(8)2024 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-39195457

RÉSUMÉ

Tight junctional complexes (TJCs) between cerebral microvascular endothelial cells (CMECs) are essential parts of the blood-brain barrier (BBB), whose regulation closely correlates to the BBB's integrity and function. hCMEC/D3 is the typical cell line used to imitate and investigate the barrier function of the BBB via the construction of an in vitro model. This study aims to investigate the protective effect of the deep-sea-derived fibrinolytic compound FGFC1 against H2O2-induced dysfunction of TJCs and to elucidate the underlying mechanism. The barrier function was shown to decline following exposure to 1 mM H2O2 in an in vitro model of hCMEC/D3 cells, with a decreasing temperature-corrected transendothelial electrical resistance (tcTEER) value. The decrease in the tcTEER value was significantly inhibited by 80 or 100 µM FGFC1, which suggested it efficiently protected the barrier integrity, allowing it to maintain its function against the H2O2-induced dysfunction. According to immunofluorescence microscopy (IFM) and quantitative real-time polymerase chain reaction (qRT-PCR), compared to the H2O2-treated group, 80~100 µM FGFC1 enhanced the expression of claudin-5 (CLDN-5) and VE-cadherin (VE-cad). And this enhancement was indicated to be mainly achieved by both up-regulation of CLDN-5 and inhibition of the down-regulation by H2O2 of VE-cad at the transcriptional level. Supported by FGFC1's molecular docking to these proteins with reasonable binding energy, FGFC1 was proved to exert a positive effect on TJCs' barrier function in hCMEC/D3 cells via targeting CLDN-5 and VE-cad. This is the first report on the protection against H2O2-induced barrier dysfunction by FGFC1 in addition to its thrombolytic effect. With CLDN-5 and VE-cad as the potential target proteins of FGFC1, this study provides evidence at the cellular and molecular levels for FGFC1's reducing the risk of bleeding transformation following its application in thrombolytic therapy for cerebral thrombosis.


Sujet(s)
Cadhérines , Cellules endothéliales , Peroxyde d'hydrogène , Jonctions serrées , Humains , Jonctions serrées/effets des médicaments et des substances chimiques , Jonctions serrées/métabolisme , Lignée cellulaire , Peroxyde d'hydrogène/toxicité , Peroxyde d'hydrogène/pharmacologie , Cadhérines/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Barrière hémato-encéphalique/métabolisme , Fibrinolytiques/pharmacologie , Claudine-5/métabolisme , Antigènes CD/métabolisme , Simulation de docking moléculaire , Facteurs de croissance fibroblastique/pharmacologie
4.
JCI Insight ; 9(14)2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-39133648

RÉSUMÉ

Neutrophils (polymorphonuclear leukocytes, PMNs) comprise a major component of the immune cell infiltrate during acute mucosal inflammation and have an important role in molding the inflammatory tissue environment. While PMNs are essential to clearance of invading microbes, the major PMN antimicrobial enzyme myeloperoxidase (MPO) can also promote bystander tissue damage. We hypothesized that blocking MPO would attenuate acute colitis and prevent the development of chronic colitis by limiting bystander tissue damage. Using the acute and chronic dextran sodium sulfate model of murine colitis, we demonstrated that MPO-deficient mice experienced less inflammation and more rapidly resolved colitis relative to wild-type controls. Mechanistic studies demonstrated that activated MPO disrupted intestinal epithelial barrier function through the dysregulation of the epithelial tight junction proteins. Our findings revealed that activated MPO chlorinates tyrosine within several tight junction proteins, thereby promoting tight junction mislocalization and dysfunction. These observations in cell models and in murine colitis were validated in human intestinal biopsies from individuals with ulcerative colitis and revealed a strong correlation between disease severity (Mayo score) and tissue chlorinated tyrosine levels. In summary, these findings implicate MPO as a viable therapeutic target to limit bystander tissue damage and preserve mucosal barrier function during inflammation.


Sujet(s)
Modèles animaux de maladie humaine , Muqueuse intestinale , Granulocytes neutrophiles , Myeloperoxidase , Protéines de la jonction serrée , Myeloperoxidase/métabolisme , Animaux , Souris , Humains , Muqueuse intestinale/anatomopathologie , Muqueuse intestinale/métabolisme , Granulocytes neutrophiles/métabolisme , Granulocytes neutrophiles/immunologie , Protéines de la jonction serrée/métabolisme , Colite/anatomopathologie , Colite/métabolisme , Colite/induit chimiquement , Halogénation , Inflammation/métabolisme , Inflammation/anatomopathologie , Mâle , Souris knockout , Sulfate dextran/toxicité , Jonctions serrées/métabolisme , Femelle , Souris de lignée C57BL , Rectocolite hémorragique/anatomopathologie , Rectocolite hémorragique/métabolisme
5.
ACS Nano ; 18(32): 21038-21051, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39096293

RÉSUMÉ

A general platform for the safe and effective oral delivery of biologics would revolutionize the administration of protein-based drugs, improving access for patients and lowering the financial burden on the health-care industry. Because of their dimensions and physiochemical properties, nanomaterials stand as promising vehicles for navigating the complex and challenging environment in the gastrointestinal (GI) tract. Recent developments have led to materials that protect protein drugs from degradation and enable controlled release in the small intestine, the site of absorption for most proteins. Yet, once present in the small intestine, the protein must transit through the secreted mucus and epithelial cells of the intestinal mucosa into systemic circulation, a process that remains a bottleneck for nanomaterial-based delivery. One attractive pathway through the intestinal mucosa is the paracellular route, which avoids cell trafficking and other degradative processes in the interior of cells. Direct flux between cells is regulated by epithelial tight junctions (TJs) that seal the paracellular space and prevent protein flux. Here, we describe a smart nanoparticle system that directly and transiently disrupts TJs for improved protein delivery, an unrealized goal to-date. We take inspiration from enteropathogenic bacteria that adhere to intestinal epithelia and secrete inhibitors that block TJ interactions in the local environment. To mimic these natural mechanisms, we engineer nanoparticles (EnteroPatho NPs) that attach to the epithelial glycocalyx and release TJ modulators in response to the intestinal pH. We show that EnteroPatho NPs lead to TJ disruption and paracellular protein delivery, giving rise to a general platform for oral delivery.


Sujet(s)
Nanoparticules , Jonctions serrées , Humains , Nanoparticules/composition chimique , Jonctions serrées/métabolisme , Jonctions serrées/effets des médicaments et des substances chimiques , Systèmes de délivrance de médicaments , Protéines/composition chimique , Protéines/métabolisme , Muqueuse intestinale/métabolisme , Vecteurs de médicaments/composition chimique , Cellules Caco-2 , Animaux
6.
Nature ; 632(8025): 647-655, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39112699

RÉSUMÉ

Biomolecular condensates enable cell compartmentalization by acting as membraneless organelles1. How cells control the interactions of condensates with other cellular structures such as membranes to drive morphological transitions remains poorly understood. We discovered that formation of a tight-junction belt, which is essential for sealing epithelial tissues, is driven by a wetting phenomenon that promotes the growth of a condensed ZO-1 layer2 around the apical membrane interface. Using temporal proximity proteomics in combination with imaging and thermodynamic theory, we found that the polarity protein PATJ mediates a transition of ZO-1 into a condensed surface layer that elongates around the apical interface. In line with the experimental observations, our theory of condensate growth shows that the speed of elongation depends on the binding affinity of ZO-1 to the apical interface and is constant. Here, using PATJ mutations, we show that ZO-1 interface binding is necessary and sufficient for tight-junction belt formation. Our results demonstrate how cells exploit the collective biophysical properties of protein condensates at membrane interfaces to shape mesoscale structures.


Sujet(s)
Condensats biomoléculaires , Membrane cellulaire , Jonctions serrées , Mouillabilité , Animaux , Chiens , Humains , Condensats biomoléculaires/métabolisme , Condensats biomoléculaires/composition chimique , Compartimentation cellulaire , Membrane cellulaire/métabolisme , Membrane cellulaire/composition chimique , Épithélium , Cellules HEK293 , Cellules rénales canines Madin-Darby , Mutation , Liaison aux protéines , Thermodynamique , Protéines de la jonction serrée/métabolisme , Jonctions serrées/métabolisme , Jonctions serrées/composition chimique , Protéine-1 de la zonula occludens/génétique , Protéine-1 de la zonula occludens/métabolisme , Protéomique
7.
Photobiomodul Photomed Laser Surg ; 42(8): 514-523, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39150379

RÉSUMÉ

Background: Although low-level laser therapy (LLLT) is a widely used noninvasive treatment because of photobiomodulation effects, its application for xerostomia remained uncertain. Tight junctions (TJs), mainly composed of claudins, occludin, and ZO family members, are crucial structures that determine material transport through paracellular pathway in salivary gland epithelial cells. This work aimed to investigate whether LLLT affected salivary secretion through epithelial TJs. Methods: Transepithelial electrical resistance (TER) measurement and paracellular permeability assay were applied to evaluate paracellular permeability in submandibular gland (SMG)-C6 cells after irradiation with 540 nm green light. Immunofluorescence and western blot were used to detect the expression of TJ proteins. Quantitative phosphoproteomics were performed to explore possible intracellular signals. Results: We found that irradiation with 540 nm green light significantly decreased TER values while increased paracellular transport in SMG-C6 cells. 540 nm green light-induced redistribution of claudin-1, -3, and -4, but not occludin or ZO-1. Moreover, above phenomena were abolished by preincubation with capsazepine, an antagonist of transient receptor potential vanilloid subtype 1. Notably, irradiation with 540 nm green light on the skin covering the whole submandibular gland regions promoted salivary secretion and attenuated lymphocytic infiltration in 21-week-old non-obese diabetic mice (n = 5 per group), a xerostomia animal model for Sjögren's syndrome. Through in-depth bioinformatics analysis and expression verification, ERK1/2 and EphA2 served as potential canonical and noncanonical signals underlying 540 nm green light. Conclusions: Our findings uncovered the novel therapeutic effects of 540 nm green light on xerostomia through regulation on the expression and distribution of TJs.


Sujet(s)
Photothérapie de faible intensité , Animaux , Souris , Glande submandibulaire/effets des radiations , Glande submandibulaire/métabolisme , Salive/métabolisme , Xérostomie/étiologie , Jonctions serrées/effets des radiations , Jonctions serrées/métabolisme , Rats ,
8.
Phytomedicine ; 133: 155953, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39154527

RÉSUMÉ

BACKGROUND: Mycoplasma gallisepticum (MG) has long been a pathogenic microorganism threatening the global poultry industry. Previous studies have demonstrated that the mechanism by which quercetin (QUE) inhibits the colonization of MG in chicks differs from that of antibiotics. However, the molecular mechanism by which QUE facilitates the clearance of MG remains unclear. PURPOSE: The aim of this study was to investigate the molecular mechanism of MG clearance by QUE, with the expectation of providing new options for the treatment of MG. METHODS: A model of MG infection in chicks and MG-induced M1 polarization in HD-11 cells were established. The mechanism of QUE clearance of MG was investigated by evaluating the relationship between tracheal mucosal barrier integrity, antibody levels, Th1/Th2 immune balance and macrophage metabolism and M1/M2 polarization balance. Furthermore, network pharmacology and molecular docking techniques were employed to explore the potential molecular pathways connecting QUE, M2 polarization, and fatty acid oxidation (FAO). RESULTS: The findings indicate that QUE remodels tracheal mucosal barrier function by regulating tight junctions and secretory immunoglobulin A (sIgA) expression levels. This process entails the regulatory function of QUE on the Th1/Th2 immune imbalance that is induced by MG infection in the tracheal mucosa. Moreover, QUE intervention impeded the M1 polarization of HD-11 cells induced by MG infection, while simultaneously promoting M2 polarization through the induction of FAO. Conversely, inhibitors of the FAO pathway impede this effect. The results of computer network analysis suggest that QUE may induce FAO via the PI3K/AKT pathway to promote M2 polarization. Notably, inhibition of the PI3K/AKT pathway was found to effectively inhibit M2 polarization in HD-11 cells, while having a limited effect on FAO. CONCLUSIONS: QUE promotes M2 polarization of HD-11 cells to enhance Th2 immune response through FAO and PI3K/AKT pathways, thereby restoring tracheal mucosal barrier function and ultimately inhibiting MG colonization.


Sujet(s)
Poulets , Infections à Mycoplasma , Mycoplasma gallisepticum , Maladies de la volaille , Quercétine , Lymphocytes auxiliaires Th2 , Animaux , Quercétine/pharmacologie , Mycoplasma gallisepticum/effets des médicaments et des substances chimiques , Infections à Mycoplasma/traitement médicamenteux , Infections à Mycoplasma/immunologie , Maladies de la volaille/traitement médicamenteux , Maladies de la volaille/immunologie , Lymphocytes auxiliaires Th2/effets des médicaments et des substances chimiques , Lymphocytes auxiliaires Th2/immunologie , Muqueuse respiratoire/effets des médicaments et des substances chimiques , Muqueuse respiratoire/immunologie , Trachée/effets des médicaments et des substances chimiques , Simulation de docking moléculaire , Jonctions serrées/effets des médicaments et des substances chimiques , Immunoglobuline A sécrétoire/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Lymphocytes auxiliaires Th1/effets des médicaments et des substances chimiques , Lymphocytes auxiliaires Th1/immunologie , Macrophages/effets des médicaments et des substances chimiques , Acides gras
10.
Biomolecules ; 14(8)2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39199277

RÉSUMÉ

We extended our model of the S1 tubular segment to address the mechanisms by which SGLT1 interacts with lateral Na/K pumps and tight junctional complexes to generate isosmotic fluid reabsorption via tubular segment S3. The strategy applied allowed for simulation of laboratory experiments. Reproducing known experimental results constrained the range of acceptable model outputs and contributed to minimizing the free parameter space. (1) In experimental conditions, published Na and K concentrations of proximal kidney cells were found to deviate substantially from their normal physiological levels. Analysis of the mechanisms involved suggested insufficient oxygen supply as the cause and, indirectly, that a main function of the Na/H exchanger (NHE3) is to extrude protons stemming from mitochondrial energy metabolism. (2) The water path from the lumen to the peritubular space passed through aquaporins on the cell membrane and claudin-2 at paracellular tight junctions, with an additional contribution to water transport by the coupling of 1 glucose:2 Na:400 H2O in SGLT1. (3) A Na-uptake component passed through paracellular junctions via solvent drag in Na- and water-permeable claudin-2, thus bypassing the Na/K pump, in agreement with the findings of early studies. (4) Electrical crosstalk between apical rheogenic SGLT1 and lateral rheogenic Na/K pumps resulted in tight coupling of luminal glucose uptake and transepithelial water flow. (5) Isosmotic transport was achieved by Na-mediated ion recirculation at the peritubular membrane.


Sujet(s)
Transporteur-1 sodium-glucose , Transporteur-1 sodium-glucose/métabolisme , Transporteur-1 sodium-glucose/génétique , Sodium/métabolisme , Humains , Transport biologique , Modèles biologiques , Eau/métabolisme , Rein/métabolisme , Jonctions serrées/métabolisme , Membrane cellulaire/métabolisme , Animaux , Sodium-Potassium-Exchanging ATPase/métabolisme , Glucose/métabolisme , Potassium/métabolisme
11.
Int J Mol Sci ; 25(16)2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39201334

RÉSUMÉ

Recent studies point to intestinal permeability as an important factor in the establishment and development of rheumatoid arthritis (RA). Tight junctions (TJs) play a major role in intestinal homeostasis. The alteration of this homeostasis is related to RA. Furthermore, RA patients present dysbiosis and a lower microbiota diversity compared to healthy individuals. A cross-sectional study including RA patients and sex- and age-matched healthy controls was performed. The quantification of TJ proteins was carried out by ELISA. Gut microbiota was evaluated by NGS platform Ion Torrent S. The inflammatory variables included were DAS28, CRP, inflammatory cytokines (IL-6, IL-1, TNF-α) and oxidised LDL. Claudin-1 levels showed significant differences between groups. Results evidenced a correlation between claudin-1 values and age (r: -0.293; p < 0.05), IL6 (r: -0.290; p < 0.05) and CRP (r: -0.327; p < 0.05), and between zonulin values and both age (r: 0.267; p < 0.05) and TNFα (r: 0.266; p < 0.05). Moreover, claudin-1 and CRP levels are related in RA patients (ß: -0.619; p: 0.045), and in patients with high inflammatory activity, the abundance of the genus Veillonella is positively associated with claudin-1 levels (ß: 39.000; p: 0.004).


Sujet(s)
Polyarthrite rhumatoïde , Dysbiose , Microbiome gastro-intestinal , Inflammation , Humains , Polyarthrite rhumatoïde/microbiologie , Polyarthrite rhumatoïde/métabolisme , Dysbiose/microbiologie , Femelle , Mâle , Adulte d'âge moyen , Études transversales , Inflammation/métabolisme , Protéines de la jonction serrée/métabolisme , Adulte , Sujet âgé , Claudine-1/métabolisme , Protéine C-réactive/métabolisme , Protéine C-réactive/analyse , Cytokines/métabolisme , Muqueuse intestinale/métabolisme , Muqueuse intestinale/microbiologie , Jonctions serrées/métabolisme , Haptoglobines , Précurseurs de protéines
12.
Diab Vasc Dis Res ; 21(4): 14791641241278506, 2024.
Article de Anglais | MEDLINE | ID: mdl-39187253

RÉSUMÉ

Human microglia (HMC) are stress-induced inflammatory cells of the retina. It is unknown whether severe hypoglycaemia causes inflammation in microglia, affects the permeability of human retinal microvascular endothelial cells (HRMECs), and causes retinal damage. This study aimed to explore the effects of severe hypoglycaemia on retinal microglial inflammation and endothelial cell permeability and evaluate the damage caused by hypoglycaemia to the retina. The CCK-8 assay was used to measure cell viability. Western blotting was used to detect IL-1ß, IL-6, TNF- α, claudin-1, and occludin expression. ELISA was used to detect IL-1ß, IL-6, and TNF- α. Transmission electron microscopy (TEM) and haematoxylin and eosin staining were used to observe the retinal structure. Immunohistochemistry and immunofluorescence staining assays were also used to detect IL-1ß, IL-6, TNF- α, claudin-1, and occludin expression. Severe hypoglycaemia promoted inflammation in HMC3 cells. Inflammation caused by hypoglycaemia leads to the decreased expression of tight junction proteins. In vivo, severe hypoglycaemia induced structural damage to the retina, increased the expression of inflammatory factors, and decreased the expression of tight junction proteins. Our results suggest that severe hypoglycaemia leads to acute retinal inflammation, affecting the permeability of HRMECs and causing retinal damage.


Sujet(s)
Perméabilité capillaire , Cellules endothéliales , Hypoglycémie , Médiateurs de l'inflammation , Microglie , Vaisseaux rétiniens , Humains , Cellules endothéliales/anatomopathologie , Cellules endothéliales/métabolisme , Cellules endothéliales/ultrastructure , Microglie/anatomopathologie , Microglie/métabolisme , Animaux , Vaisseaux rétiniens/anatomopathologie , Vaisseaux rétiniens/métabolisme , Médiateurs de l'inflammation/métabolisme , Lignée cellulaire , Hypoglycémie/métabolisme , Hypoglycémie/anatomopathologie , Modèles animaux de maladie humaine , Occludine/métabolisme , Microvaisseaux/anatomopathologie , Microvaisseaux/métabolisme , Jonctions serrées/métabolisme , Jonctions serrées/anatomopathologie , Jonctions serrées/ultrastructure , Cytokines/métabolisme , Claudine-1/métabolisme , Claudine-1/génétique , Mâle , Glycémie/métabolisme , Souris de lignée C57BL , Barrière hématorétinienne/anatomopathologie , Barrière hématorétinienne/métabolisme , Transduction du signal
13.
J Ocul Pharmacol Ther ; 40(6): 379-388, 2024.
Article de Anglais | MEDLINE | ID: mdl-39172123

RÉSUMÉ

Purpose: To explore the effects of the tight junction protein zonula occludens 1 (ZO-1) on experimental corneal neovascularization (CNV). Methods: CNV models were established in the left eyes of BALB/c mice using NaOH. Anti-ZO-1 neutralizing antibody was topically applied to the burnt corneas after modeling thrice a day for 1 week. CD31 expression was analyzed to calculate the ratio of CNV number to area using a corneal whole-mount fluorescent immunohistochemical assay. Messenger ribonucleic acid (mRNA) and protein expression levels of ZO-1, vascular endothelial growth factor (VEGF), interleukin (IL)-1ß, IL-6, IL-8, IL-18, monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor alpha (TNF-α), phosphorylated protein kinase C (pPKC), and clusterin in burned corneas were detected by reverse transcriptase polymerase chain reaction (PCR) and western blot analyses. Infiltration of neutrophils, macrophages, and progenitor cells was examined by flow cytometry. Results: CNV was obviously greater in 45 s than in 15 s alkali injury group. In another experiment, CNV was obviously greater in the ZO-1 antibody group than in the vehicle-treated group. Corneal mRNA and protein expression levels of VEGF, IL-1ß, IL-6, IL-8, IL-18, and MCP-1 were significantly higher in the ZO-1 antibody group than in the control group. Infiltration of neutrophils, macrophages, and progenitor cells was significantly greater in the ZO-1 antibody group than in the control group. TNF-α expression was much higher in 45 s than in 15 s alkali injury group. However, protein expression of pPKC and clusterin was much lower in 45 s than in 15 s alkali injury group. Conclusions: Anti-ZO-1 neutralizing antibody-treated mice exhibited enhanced alkali-induced CNV through enhanced intracorneal infiltration of progenitor and inflammatory cells.


Sujet(s)
Néovascularisation cornéenne , Modèles animaux de maladie humaine , Souris de lignée BALB C , Protéine-1 de la zonula occludens , Animaux , Néovascularisation cornéenne/métabolisme , Néovascularisation cornéenne/anatomopathologie , Protéine-1 de la zonula occludens/métabolisme , Souris , ARN messager/métabolisme , Hydroxyde de sodium , Anticorps neutralisants/pharmacologie , Mâle , Jonctions serrées/métabolisme , Cytokines/métabolisme
14.
J Med Virol ; 96(8): e29864, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39158051

RÉSUMÉ

Inflammation and autoimmune responses contribute to the pathophysiology of Long COVID, and its affective and chronic fatigue syndrome symptoms, labeled "the physio-affective phenome." To investigate whether Long COVID and its physio-affective phenome are linked to autoimmunity to the tight junction proteins, zonulin and occludin (ZOOC), and immune reactivity to lipopolysaccharides (LPS), and whether the latter are associated with signs of human herpes virus-6 (HHV-6) reactivation, autoimmunity directed against oligodendrocyte and neuronal proteins, including myelin basic protein. IgA/IgM/IgG responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), HHV-6, ZOOC, and neuronal proteins, C-reactive protein (CRP), and advanced oxidation protein products (AOPPs), were measured in 90 Long COVID patients and 90 healthy controls. The physio-affective phenome was conceptualized as a factor extracted from physical and affective symptom domains. Neural network identified IgA directed to LPS (IgA-LPS), IgG-ZOOC, IgG-LPS, and IgA-ZOOC as important variables associated with Long COVID diagnosis with an area under the ROC curve of 0.755. Partial Least Squares analysis showed that 40.9% of the variance in the physio-affective phenome was explained by CRP, IgA-myelin basic protein (MBP), and IgG-MBP. A large part of the variances in both autoimmune responses to MBP (36.3%-39.7%) was explained by autoimmunity (IgA and IgG) directed to ZOOC. The latter was strongly associated with indicants of HHV-6 reactivation, which in turn was associated with increased IgM-SARS-CoV-2. Autoimmunity against components of the tight junctions and increased bacterial translocation may be involved in the pathophysiology of Long COVID's physio-affective phenome.


Sujet(s)
Auto-immunité , COVID-19 , Syndrome de fatigue chronique , Herpèsvirus humain de type 6 , Inflammation , Jonctions serrées , Humains , Syndrome de fatigue chronique/immunologie , Syndrome de fatigue chronique/virologie , Herpèsvirus humain de type 6/immunologie , Femelle , Mâle , Adulte d'âge moyen , Jonctions serrées/immunologie , COVID-19/immunologie , Inflammation/immunologie , Adulte , Occludine , Dépression/immunologie , SARS-CoV-2/immunologie , Sujet âgé , Immunoglobuline G/sang , Syndrome de post-COVID-19 , Immunoglobuline A/sang , Lipopolysaccharides/immunologie , Autoanticorps/sang , Autoanticorps/immunologie , Anticorps antiviraux/sang , Infections à roséolovirus/immunologie , Infections à roséolovirus/complications , Infections à roséolovirus/virologie , Haptoglobines , Précurseurs de protéines
16.
Gut Microbes ; 16(1): 2389320, 2024.
Article de Anglais | MEDLINE | ID: mdl-39150987

RÉSUMÉ

The intestinal barrier, an indispensable guardian of gastrointestinal health, mediates the intricate exchange between internal and external environments. Anchored by evolutionarily conserved junctional complexes, this barrier meticulously regulates paracellular permeability in essentially all living organisms. Disruptions in intestinal junctional complexes, prevalent in inflammatory bowel diseases and irritable bowel syndrome, compromise barrier integrity and often lead to the notorious "leaky gut" syndrome. Critical to the maintenance of the intestinal barrier is a finely orchestrated network of intrinsic and extrinsic factors that modulate the expression, composition, and functionality of junctional complexes. This review navigates through the composition of key junctional complex components and the common methods used to assess intestinal permeability. It also explores the critical intracellular signaling pathways that modulate these junctional components. Lastly, we delve into the complex dynamics between the junctional complexes, microbial communities, and environmental chemicals in shaping the intestinal barrier function. Comprehending this intricate interplay holds paramount importance in unraveling the pathophysiology of gastrointestinal disorders. Furthermore, it lays the foundation for the development of precise therapeutic interventions targeting barrier dysfunction.


Sujet(s)
Microbiome gastro-intestinal , Muqueuse intestinale , Perméabilité , Humains , Muqueuse intestinale/métabolisme , Muqueuse intestinale/microbiologie , Animaux , Jonctions serrées/métabolisme , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/physiopathologie , Transduction du signal , Syndrome du côlon irritable/métabolisme , Syndrome du côlon irritable/physiopathologie
17.
Mol Nutr Food Res ; 68(16): e2400230, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39086054

RÉSUMÉ

Tight junction disruption can lead to pathogenesis of various diseases without therapeutic strategy to recover intestinal barrier integrity. The main objective of this study is to demonstrate the effect of Solanum melongena L. extract (SMLE) on intestinal tight junction recovery and its underlying mechanism. Intestinal barrier function is attenuated by Ca2+ depletion. SMLE treatment increased TER value across T84 cell monolayers. Permeability assay reveals that Ca2+ depletion promotes 4-kDa FITC-dextran permeability, but not 70-kDa FITC-dextran. SMLE suppresses the rate of 4-kDa FITC-dextran permeability, indicating that SMLE inhibits paracellular leak pathway permeability. SMLE-mediated TER increase and leak pathway suppression are abolished by neither calcium/calmodulin-dependent protein kinase kinase ß (CaMKKß) inhibitor nor AMP-activated protein kinase (AMPK) inhibitor. Furthermore, mammalian target of rapamycin (mTOR) and extracellular signal-regulated kinase (ERK) inhibitors have no effects on SMLE-mediated TER increase and leak pathway suppression. Interestingly, SMLE is unable to enhance TER value and diminish leak pathway permeability in T84 cell monolayers pre-treated with sirtuin-1 (SIRT-1) inhibitor. Immunofluorescence staining reveals that SMLE enhances re-assembly of tight junction proteins, including occludin and ZO-1 to intercellular space but this effect is abolished by SIRT-1 inhibitor. These data suggest that SMLE promotes intestinal tight junction re-assembly via SIRT-1-dependent manner.


Sujet(s)
Extraits de plantes , Sirtuine-1 , Jonctions serrées , Sirtuine-1/métabolisme , Jonctions serrées/effets des médicaments et des substances chimiques , Jonctions serrées/métabolisme , Extraits de plantes/pharmacologie , Humains , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/métabolisme , Occludine/métabolisme , Perméabilité/effets des médicaments et des substances chimiques , Calcium/métabolisme , Lignée cellulaire , Protéine-1 de la zonula occludens/métabolisme , Dextrane , Sérine-thréonine kinases TOR/métabolisme , Fluorescéine-5-isothiocyanate/analogues et dérivés
18.
J Nanobiotechnology ; 22(1): 456, 2024 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-39085856

RÉSUMÉ

Spinal cord injury (SCI) compromises the blood-spinal cord barrier (BSCB) and induces neuroinflammation, potentially exacerbating neuronal damage. This underscores the importance of maintaining BSCB integrity and mitigating neuroinflammation in SCI treatment. Our study explores an innovative approach to treating SCI by utilizing platelet-rich plasma-derived exosomes (PRP-Exos) to stabilize BSCB function and alleviate neuroinflammation. We successfully isolated exosomes from platelet-rich plasma and conducted both in vivo and in vitro experiments to assess the therapeutic effects of PRP-Exos and explore their potential mechanisms in stabilizing the BSCB, reducing neuroinflammation, and promoting neural functional recovery.In vitro results demonstrate that PRP-Exos significantly reduce the permeability of bEnd.3 cells under hypoxic-hypoglycemic conditions, thereby restoring the integrity of tight junctions. Additionally, our study elucidates the critical role of the NF-κB signaling pathway in the amelioration of neuroinflammation by PRP-Exos. In the SCI model, local injection of hydrogel-encapsulated PRP-Exos reduced Evans blue dye leakage, enhanced the expression of tight junction proteins, alleviated the inflammatory environment in the damaged area, and improved neural functional recovery. In conclusion, PRP-Exos presents a promising and effective treatment option for SCI.


Sujet(s)
Exosomes , Maladies neuro-inflammatoires , Plasma riche en plaquettes , Traumatismes de la moelle épinière , Moelle spinale , Traumatismes de la moelle épinière/thérapie , Exosomes/métabolisme , Plasma riche en plaquettes/métabolisme , Plasma riche en plaquettes/composition chimique , Animaux , Souris , Moelle spinale/métabolisme , Lignée cellulaire , Mâle , Souris de lignée C57BL , Barrière hémato-encéphalique/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Jonctions serrées/métabolisme , Inflammation , Transduction du signal , Femelle
19.
Front Immunol ; 15: 1348010, 2024.
Article de Anglais | MEDLINE | ID: mdl-39081324

RÉSUMÉ

Background: Defective intestinal epithelial tight junction (TJ), characterized by an increase in intestinal TJ permeability, has been shown to play a critical role in the pathogenesis of inflammatory bowel disease (IBD). Tumor necrosis factor-α (TNF-α) is a key pro-inflammatory cytokine involved in the immunopathology of IBD and has been shown to cause an increase in intestinal epithelial TJ permeability. Although TNF-α antibodies and other biologics have been advanced for use in IBD treatment, these therapies are associated with severe side effects and have limited efficacy, and there is an urgent need for therapies with benign profiles and high therapeutic efficacy. Probiotic bacteria have beneficial effects and are generally safe and represent an important class of potential therapeutic agents in IBD. Lactobacillus acidophilus (LA) is one of the most used probiotics for wide-ranging health benefits, including in gastrointestinal, metabolic, and inflammatory disorders. A specific strain of LA, LA1, was recently demonstrated to have protective and therapeutic effects on the intestinal epithelial TJ barrier. However, the mechanisms of actions of LA1 remain largely unknown. Methods: The primary aim of this study was to investigate microbial-epithelial interactions and novel signaling pathways that regulate the effect of LA1 on TNF-α-induced increase in intestinal epithelial TJ permeability, using cell culture and animal model systems. Results and Conclusion: Pre-treatment of filter-grown Caco-2 monolayers with LA1 prevented the TNF-α-induced increase in intestinal epithelial TJ permeability by inhibiting TNF-α-induced activation of NF-κB p50/p65 and myosin light chain kinase (MLCK) gene and kinase activity in a TLR-2-dependent manner. LA1 produced a TLR-2- and MyD88-dependent activation of NF-κB p50/p65 in immune cells; however, LA1, in intestinal cells, inhibited the NF-κB p50/p65 activation in a TLR-2-dependent but MyD88-independent manner. In addition, LA1 inhibition of NF-κB p50/p65 and MLCK gene was mediated by TLR-2 pathway activation of phosphatidylinositol 3-kinase (PI3K) and IKK-α phosphorylation. Our results demonstrated novel intracellular signaling pathways by which LA1/TLR-2 suppresses the TNF-α pathway activation of NF-κB p50/p65 in intestinal epithelial cells and protects against the TNF-α-induced increase in intestinal epithelial TJ permeability.


Sujet(s)
Muqueuse intestinale , Lactobacillus acidophilus , Facteur de transcription NF-kappa B , Phosphatidylinositol 3-kinases , Probiotiques , Jonctions serrées , Récepteur de type Toll-2 , Facteur de nécrose tumorale alpha , Lactobacillus acidophilus/physiologie , Facteur de nécrose tumorale alpha/métabolisme , Jonctions serrées/métabolisme , Humains , Muqueuse intestinale/métabolisme , Muqueuse intestinale/immunologie , Muqueuse intestinale/microbiologie , Animaux , Probiotiques/pharmacologie , Récepteur de type Toll-2/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Souris , Perméabilité , Transduction du signal/effets des médicaments et des substances chimiques , Cellules Caco-2 , Maladies inflammatoires intestinales/immunologie , Maladies inflammatoires intestinales/métabolisme
20.
J Cell Mol Med ; 28(14): e18545, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39031471

RÉSUMÉ

Hypoxia plays an important role in the pathological process of bladder outlet obstruction. Previous research has mostly focused on the dysfunction of bladder smooth muscle cells, which are directly related to bladder contraction. This study delves into the barrier function changes of the urothelial cells under exposure to hypoxia. Results indicated that after a 5-day culture, SV-HUC-1 formed a monolayer and/or bilayer of cell sheets, with tight junction formation, but no asymmetrical unit membrane was observed. qPCR and western blotting revealed the expression of TJ-associated proteins (occludin, claudin1 and ZO-1) was significantly decreased in the hypoxia group in a time-dependent manner. No expression changes were observed in uroplakins. When compared to normoxic groups, immunofluorescent staining revealed a reduction in the expression of TJ-associated proteins in the hypoxia group. Transepithelial electrical resistance (TEER) revealed a statistically significant decrease in resistance in the hypoxia group. Fluorescein isothiocyanate-conjugated dextran assay was inversely proportional to the results of TEER. Taken together, hypoxia down-regulates the expression of TJ-associated proteins and breaks tight junctions, thus impairing the barrier function in human urothelial cells.


Sujet(s)
Hypoxie cellulaire , Protéines de la jonction serrée , Jonctions serrées , Urothélium , Humains , Urothélium/métabolisme , Urothélium/anatomopathologie , Jonctions serrées/métabolisme , Protéines de la jonction serrée/métabolisme , Protéines de la jonction serrée/génétique , Lignée cellulaire , Protéine-1 de la zonula occludens/métabolisme , Protéine-1 de la zonula occludens/génétique , Occludine/métabolisme , Occludine/génétique , Claudine-1/métabolisme , Claudine-1/génétique , Impédance électrique , Régulation de l'expression des gènes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE