Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 40.267
Filtrer
1.
BMC Oral Health ; 24(1): 756, 2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-38951854

RÉSUMÉ

OBJECTIVE: Oral lichen planus carries a risk for malignancy. The pathogenesis of the disease is mediated by various inflammatory mediators. Several mediators could be responsible for the oncogenic behavior in certain cases. Hypoxia-inducible factor-1a (HIF-1), and its possible correlation to Galactin-3 (Gal-3) and matrix metalloproteinase-9 (MMP-9) over expression represents an important indicator for malignant transformation. The investigation of these factors may present evidence-based information on malignant transformation of the disease. SUBJECTS AND METHODS: The study investigated the expression of HIF-1, Gla-3 and MMP-9 in tissue samples of OLP compared to control subjects of un-inflamed gingival overgrowth. 20 biospecimen were allocated in each group. RESULTS: Immunohistochemical findings of OLP showed immunoreactivity for Galectin 3, HIF1a and MMP-9 by most of the epithelial cells. There was a positive correlation between HIF1α and MMP-9, r = 0.9301 (P-value < 0.00001). A positive correlation was detected between Galectin 3 and MMP-9, r = 0.7292 (P-value = 0.000264) between Galectin 3 and HIF1α, r = 0.5893 (P-value = 0.006252). CONCLUSION: These findings confirm the hypothesis that the adaptive pathways to hypoxia as Gal 3 and MMP-9 expressions and their HIF-1 may play a crucial role in carcinogenesis of OLP.


Sujet(s)
Galectine -3 , Sous-unité alpha du facteur-1 induit par l'hypoxie , Lichen plan buccal , Matrix metalloproteinase 9 , Humains , Matrix metalloproteinase 9/métabolisme , Lichen plan buccal/métabolisme , Lichen plan buccal/anatomopathologie , Galectine -3/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Femelle , Mâle , Adulte d'âge moyen , Galectines/métabolisme , Adulte , Transformation cellulaire néoplasique , Cellules épithéliales/métabolisme , Études cas-témoins , Immunohistochimie , Protéines du sang
2.
J Transl Med ; 22(1): 607, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38951896

RÉSUMÉ

Clear cell renal cell carcinoma (ccRCC) is a prevalent malignancy with complex heterogeneity within epithelial cells, which plays a crucial role in tumor progression and immune regulation. Yet, the clinical importance of the malignant epithelial cell-related genes (MECRGs) in ccRCC remains insufficiently understood. This research aims to undertake a comprehensive investigation into the functions and clinical relevance of malignant epithelial cell-related genes in ccRCC, providing valuable understanding of the molecular mechanisms and offering potential targets for treatment strategies. Using data from single-cell sequencing, we successfully identified 219 MECRGs and established a prognostic model MECRGS (MECRGs' signature) by synergistically analyzing 101 machine-learning models using 10 different algorithms. Remarkably, the MECRGS demonstrated superior predictive performance compared to traditional clinical features and 92 previously published signatures across six cohorts, showcasing its independence and accuracy. Upon stratifying patients into high- and low-MECRGS subgroups using the specified cut-off threshold, we noted that patients with elevated MECRGS scores displayed characteristics of an immune suppressive tumor microenvironment (TME) and showed worse outcomes after immunotherapy. Additionally, we discovered a distinct ccRCC tumor cell subtype characterized by the high expressions of PLOD2 (procollagen-lysine,2-oxoglutarate 5-dioxygenase 2) and SAA1 (Serum Amyloid A1), which we further validated in the Renji tissue microarray (TMA) cohort. Lastly, 'Cellchat' revealed potential crosstalk patterns between these cells and other cell types, indicating their potential role in recruiting CD163 + macrophages and regulatory T cells (Tregs), thereby establishing an immunosuppressive TME. PLOD2 + SAA1 + cancer cells with intricate crosstalk patterns indeed show promise for potential therapeutic interventions.


Sujet(s)
Néphrocarcinome , Cellules épithéliales , Régulation de l'expression des gènes tumoraux , Tumeurs du rein , Microenvironnement tumoral , Humains , Néphrocarcinome/génétique , Néphrocarcinome/anatomopathologie , Microenvironnement tumoral/génétique , Tumeurs du rein/génétique , Tumeurs du rein/anatomopathologie , Pronostic , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Femelle , Mâle , Analyse de profil d'expression de gènes , Apprentissage machine
3.
Int Ophthalmol ; 44(1): 316, 2024 Jul 06.
Article de Anglais | MEDLINE | ID: mdl-38969958

RÉSUMÉ

BACKGROUND: Diabetic cataract (DC) is a common complication of diabetes and its etiology and progression are multi-factorial. In this study, the roles of specific protein 1 (SP1) and fibroblast growth factor 7 (FGF7) in DC development were explored. METHODS: DC cell model was established by treating SRA01/04 cells with high glucose (HG). MTT assay was conducted to evaluate cell viability. Transwell assay and wound-healing assay were performed to assess cell migration and invasion. Western blot assay and qRT-PCR assay were conducted to measure the expression of N-cadherin, E-cadherin, Collagen I, Fibronectin, SP1 and FGF7 expression. CHIP assay and dual-luciferase reporter assay were conducted to analyze the combination between FGF7 and SP1. RESULTS: FGF7 was upregulated in DC patients and HG-induced SRA01/04 cells. HG treatment promoted SRA01/04 cell viability, migration, invasion and epithelial-mesenchymal transition (EMT), while FGF7 knockdown abated the effects. Transcription factor SP1 activated the transcription level of FGF7 and SP1 overexpression aggravated HG-induced SRA01/04 cell injury. SP1 silencing repressed HG-induced SRA01/04 cell viability, migration, invasion and EMT, but these effects were ameliorated by upregulating FGF7. Additionally, SP1 knockdown inhibited the PI3K/AKT pathway by regulating the transcription level of FGF7. CONCLUSION: Transcription factor SP1 activated the transcription level of FGF7 and the PI3K/AKT pathway to regulate HG-induced SRA01/04 cell viability, migration, invasion and EMT.


Sujet(s)
Mouvement cellulaire , Survie cellulaire , Cellules épithéliales , Transition épithélio-mésenchymateuse , Facteur de croissance fibroblastique de type 7 , Glucose , Cristallin , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Facteur de transcription Sp1 , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Facteur de transcription Sp1/métabolisme , Facteur de transcription Sp1/génétique , Humains , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Glucose/pharmacologie , Cellules épithéliales/métabolisme , Facteur de croissance fibroblastique de type 7/métabolisme , Facteur de croissance fibroblastique de type 7/génétique , Facteur de croissance fibroblastique de type 7/pharmacologie , Cristallin/métabolisme , Cristallin/cytologie , Cataracte/métabolisme , Cellules cultivées , Régulation de l'expression des gènes
4.
Transl Vis Sci Technol ; 13(7): 2, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38949633

RÉSUMÉ

Purpose: We sought to evaluate the efficacy of growth differentiation factor (GDF)-15 treatment for suppressing epithelial-mesenchymal transition (EMT) and alleviating transforming growth factor ß2 (TGFß2)-induced lens opacity. Methods: To test whether GDF-15 is a molecule that prevents EMT, we pretreated the culture with GDF-15 in neural progenitor cells, retinal pigment epithelial cells, and lens epithelial cells and then treated with factors that promote EMT, GDF-11, and TGFß2, respectively. To further investigate the efficacy of GDF-15 on alleviating lens opacity, we used mouse lens explant culture to mimic secondary cataracts. We pretreated the lens culture with GDF-15 and then added TGFß2 to develop lens opacity (n = 3 for each group). Western blot and quantitative reverse transcription polymerase chain reaction (qRT-PCR) were used to measure EMT protein and gene expression, respectively. Results: In cell culture, GDF-15 pretreatment significantly attenuated EMT marker expression in cultured cells induced by treatment with GDF-11 or TGFß2. In the lens explant culture, GDF-15 pretreatment also reduced mouse lens opacity induced by exposure to TGFß2. Conclusions: Our results indicate that GDF-15 could alleviate TGFß2-induced EMT and is a potential therapeutic agent to slow or prevent posterior capsular opacification (PCO) progression after cataract surgery. Translational Relevance: Cataracts are the leading cause of blindness worldwide, with the only current treatment involving surgical removal of the lens and replacement with an artificial lens. However, PCO, also known as secondary cataract, is a common complication after cataract surgery. The development of an adjuvant that slows the progression of PCO will be beneficial to the field of anterior complications.


Sujet(s)
Cataracte , Transition épithélio-mésenchymateuse , Facteur-15 de croissance et de différenciation , Cristallin , Facteur de croissance transformant bêta-2 , Animaux , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Facteur de croissance transformant bêta-2/métabolisme , Facteur de croissance transformant bêta-2/pharmacologie , Facteur-15 de croissance et de différenciation/métabolisme , Facteur-15 de croissance et de différenciation/génétique , Cataracte/anatomopathologie , Cataracte/métabolisme , Cataracte/prévention et contrôle , Souris , Cristallin/métabolisme , Cristallin/anatomopathologie , Cristallin/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Cellules cultivées , Modèles animaux de maladie humaine , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Technique de Western , Épithélium pigmentaire de la rétine/effets des médicaments et des substances chimiques , Épithélium pigmentaire de la rétine/anatomopathologie , Épithélium pigmentaire de la rétine/métabolisme
5.
Nat Commun ; 15(1): 5545, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956024

RÉSUMÉ

Epithelial cells are the first point of contact for bacteria entering the respiratory tract. Streptococcus pneumoniae is an obligate human pathobiont of the nasal mucosa, carried asymptomatically but also the cause of severe pneumoniae. The role of the epithelium in maintaining homeostatic interactions or mounting an inflammatory response to invasive S. pneumoniae is currently poorly understood. However, studies have shown that chromatin modifications, at the histone level, induced by bacterial pathogens interfere with the host transcriptional program and promote infection. Here, we uncover a histone modification induced by S. pneumoniae infection maintained for at least 9 days upon clearance of bacteria with antibiotics. Di-methylation of histone H3 on lysine 4 (H3K4me2) is induced in an active manner by bacterial attachment to host cells. We show that infection establishes a unique epigenetic program affecting the transcriptional response of epithelial cells, rendering them more permissive upon secondary infection. Our results establish H3K4me2 as a unique modification induced by infection, distinct from H3K4me3 or me1, which localizes to enhancer regions genome-wide. Therefore, this study reveals evidence that bacterial infection leaves a memory in epithelial cells after bacterial clearance, in an epigenomic mark, thereby altering cellular responses to subsequent infections and promoting infection.


Sujet(s)
Cellules épithéliales , Histone , Infections à pneumocoques , Streptococcus pneumoniae , Histone/métabolisme , Streptococcus pneumoniae/métabolisme , Streptococcus pneumoniae/physiologie , Cellules épithéliales/microbiologie , Cellules épithéliales/métabolisme , Méthylation , Humains , Infections à pneumocoques/microbiologie , Infections à pneumocoques/métabolisme , Épigenèse génétique , Animaux , Souris , Lysine/métabolisme , Souris de lignée C57BL
6.
Cell Death Dis ; 15(7): 473, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956064

RÉSUMÉ

Damage to renal tubular epithelial cells (RTECs) signaled the onset and progression of sepsis-associated acute kidney injury (SA-AKI). Recent research on mitochondria has revealed that mitophagy plays a crucial physiological role in alleviating injury to RTECs and it is suppressed progressively by the inflammation response in SA-AKI. However, the mechanism by which inflammation influences mitophagy remains poorly understood. We examined how macrophage migration inhibitory factor (MIF), a pro-inflammatory protein, influences the PINK1-Parkin pathway of mitophagy by studying protein-protein interactions when MIF was inhibited or overexpressed. Surprisingly, elevated levels of MIF were found to directly bind to PINK1, disrupting its interaction with Parkin. This interference hindered the recruitment of Parkin to mitochondria and impeded the initiation of mitophagy. Furthermore, this outcome led to significant apoptosis of RTECs, which could, however, be reversed by an MIF inhibitor ISO-1 and/or a new mitophagy activator T0467. These findings highlight the detrimental impact of MIF on renal damage through its disruption of the interaction between PINK1 and Parkin, and the therapeutic potential of ISO-1 and T0467 in mitigating SA-AKI. This study offers a fresh perspective on treating SA-AKI by targeting MIF and mitophagy.


Sujet(s)
Atteinte rénale aigüe , Facteurs inhibiteurs de la migration des macrophages , Mitophagie , Protein kinases , Sepsie , Ubiquitin-protein ligases , Facteurs inhibiteurs de la migration des macrophages/métabolisme , Facteurs inhibiteurs de la migration des macrophages/génétique , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/anatomopathologie , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Protein kinases/métabolisme , Sepsie/complications , Sepsie/métabolisme , Animaux , Humains , Mitochondries/métabolisme , Tubules rénaux/métabolisme , Tubules rénaux/anatomopathologie , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Apoptose , Liaison aux protéines , Mâle , Intramolecular oxidoreductases/métabolisme
7.
Mol Med Rep ; 30(3)2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38963029

RÉSUMÉ

Viral infections in the respiratory tract are common, and, in recent years, severe acute respiratory syndrome coronavirus 2 outbreaks have highlighted the effect of viral infections on antiviral innate immune and inflammatory reactions. Specific treatments for numerous viral respiratory infections have not yet been established and they are mainly treated symptomatically. Therefore, understanding the details of the innate immune system underlying the airway epithelium is crucial for the development of new therapies. The present study aimed to investigate the function and expression of interferon (IFN)­stimulated gene (ISG)60 in non­cancerous bronchial epithelial BEAS­2B cells exposed to a Toll­like receptor 3 agonist. BEAS­2B cells were treated with a synthetic TLR3 ligand, polyinosinic­polycytidylic acid (poly IC). The mRNA and protein expression levels of ISG60 were analyzed using reverse transcription­quantitative PCR and western blotting, respectively. The levels of C­X­C motif chemokine ligand 10 (CXCL10) were examined using an enzyme­linked immunosorbent assay, and the effects of knockdown of IFN­ß, ISG60 and ISG56 were examined using specific small interfering RNAs. Notably, ISG60 expression was increased in proportion to poly IC concentration, and recombinant human IFN­ß also induced ISG60 expression. By contrast, knockdown of IFN­ß and ISG56 decreased ISG60 expression, and ISG60 knockdown reduced CXCL10 and ISG56 expression. These findings suggested that ISG60 is partly implicated in CXCL10 expression and that ISG60 may serve a role in the innate immune response of bronchial epithelial cells. The present study highlights ISG60 as a potential target for new therapeutic strategies against viral infections in the airway.


Sujet(s)
Bronches , Chimiokine CXCL10 , Cellules épithéliales , Poly I-C , Transduction du signal , Récepteur de type Toll-3 , Humains , Récepteur de type Toll-3/métabolisme , Récepteur de type Toll-3/génétique , Chimiokine CXCL10/métabolisme , Chimiokine CXCL10/génétique , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Bronches/cytologie , Bronches/métabolisme , Poly I-C/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Lignée cellulaire , Immunité innée , Interféron bêta/métabolisme , Interféron bêta/génétique , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Protéines de liaison à l'ARN , Protéines adaptatrices de la transduction du signal , Protéines régulatrices de l'apoptose
8.
Sci Rep ; 14(1): 15442, 2024 07 04.
Article de Anglais | MEDLINE | ID: mdl-38965312

RÉSUMÉ

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.


Sujet(s)
Cellules épithéliales , Muqueuse intestinale , bêta-Défensines , Humains , bêta-Défensines/métabolisme , bêta-Défensines/génétique , Muqueuse intestinale/métabolisme , Muqueuse intestinale/microbiologie , Cellules épithéliales/métabolisme , Cellules épithéliales/microbiologie , Transduction du signal , Régulation de l'expression des gènes , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme , Facteur de différenciation myéloïde-88/métabolisme , Facteur de différenciation myéloïde-88/génétique , Interférence par ARN
9.
Cell Commun Signal ; 22(1): 357, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38987851

RÉSUMÉ

BACKGROUND: Chronic kidney disease (CKD) is highly prevalent worldwide, and its global burden is substantial and growing. CKD displays a number of features of accelerated senescence. Tubular cell senescence is a common biological process that contributes to CKD progression. Tubulointerstitial inflammation is a driver of tubular cell senescence and a common characteristic of CKD. However, the mechanism by which the interstitial inflammation drives tubular cell senescence remains unclear. This paper aims to explore the role of exosomal miRNAs derived from macrophages in the development of tubular cell senescence. METHODS: Among the identified inflammation-related miRNAs, miR-155 is considered to be one of the most important miRNAs involved in the inflammatory response. Macrophages, the primary immune cells that mediate inflammatory processes, contain a high abundance of miR-155 in their released exosomes. We assessed the potential role of miR-155 in tubular cell senescence and renal fibrosis. We subjected miR-155-/- mice and wild-type controls, as well as tubular epithelial cells (TECs), to angiotensin II (AngII)-induced kidney injury. We assessed kidney function and injury using standard techniques. TECs were evaluated for cell senescence and telomere dysfunction in vivo and in vitro. Telomeres were measured by the fluorescence in situ hybridization. RESULTS: Compared with normal controls, miR-155 was up-regulated in proximal renal tubule cells in CKD patients and mouse models of CKD. Moreover, the expression of miR-155 was positively correlated with the extent of renal fibrosis, eGFR decline and p16INK4A expression. The overexpression of miR-155 exacerbated tubular senescence, evidenced by increased detection of p16INK4A/p21expression and senescence-associated ß-galactosidase activity. Notably, miR-155 knockout attenuates renal fibrosis and tubule cell senescence in vivo. Interestingly, once released, macrophages-derived exosomal miR-155 was internalized by TECs, leading to telomere shortening and dysfunction through targeting TRF1. A dual-luciferase reporter assay confirmed that TRF1 was the direct target of miR-155. Thus, our study clearly demonstrates that exosomal miR-155 may mediate communication between macrophages and TECs, subsequently inducing telomere dysfunction and senescence in TECs. CONCLUSIONS: Our work suggests a new mechanism by which macrophage exosomes are involved in the development of tubule senescence and renal fibrosis, in part by delivering miR-155 to target TRF1 to promote telomere dysfunction. Our study may provide novel strategies for the treatment of AngII-induced kidney injury.


Sujet(s)
Vieillissement de la cellule , Cellules épithéliales , Exosomes , Tubules rénaux , Macrophages , microARN , Télomère , microARN/génétique , microARN/métabolisme , Vieillissement de la cellule/génétique , Exosomes/métabolisme , Exosomes/génétique , Animaux , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Macrophages/métabolisme , Tubules rénaux/anatomopathologie , Tubules rénaux/métabolisme , Souris , Télomère/génétique , Télomère/métabolisme , Humains , Souris de lignée C57BL , Mâle , Insuffisance rénale chronique/génétique , Insuffisance rénale chronique/anatomopathologie , Fibrose/génétique , Angiotensine-II
11.
Cell Commun Signal ; 22(1): 351, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38970061

RÉSUMÉ

BACKGROUND: Accompanied by activation of the NOD-like receptor protein 3 (NLRP3) inflammasome, aberrant connexin 43 (Cx43) hemichannel-mediated ATP release is situated upstream of inflammasome assembly and inflammation and contributes to multiple secondary complications of diabetes and associated cardiometabolic comorbidities. Evidence suggests there may be a link between Cx43 hemichannel activity and inflammation in the diabetic kidney. The consequences of blocking tubular Cx43 hemichannel-mediated ATP release in priming/activation of the NLRP3 inflammasome in a model of diabetic kidney disease (DKD) was investigated. We examined downstream markers of inflammation and the proinflammatory and chemoattractant role of the tubular secretome on macrophage recruitment and activation. METHODS: Analysis of human transcriptomic data from the Nephroseq repository correlated gene expression to renal function in DKD. Primary human renal proximal tubule epithelial cells (RPTECs) and monocyte-derived macrophages (MDMs) were cultured in high glucose and inflammatory cytokines as a model of DKD to assess Cx43 hemichannel activity, NLRP3 inflammasome activation and epithelial-to-macrophage paracrine-mediated crosstalk. Tonabersat assessed a role for Cx43 hemichannels. RESULTS: Transcriptomic analysis from renal biopsies of patients with DKD showed that increased Cx43 and NLRP3 expression correlated with declining glomerular filtration rate (GFR) and increased proteinuria. In vitro, Tonabersat blocked glucose/cytokine-dependant increases in Cx43 hemichannel-mediated ATP release and reduced expression of inflammatory markers and NLRP3 inflammasome activation in RPTECs. We observed a reciprocal relationship in which NLRP3 activity exacerbated increased Cx43 expression and hemichannel-mediated ATP release, events driven by nuclear factor kappa-B (NFκB)-mediated priming and Cx43 hemichannel opening, changes blocked by Tonabersat. Conditioned media (CM) from RPTECs treated with high glucose/cytokines increased expression of inflammatory markers in MDMs, an effect reduced when macrophages were pre-treated with Tonabersat. Co-culture using conditioned media from Tonabersat-treated RPTECs dampened macrophage inflammatory marker expression and reduced macrophage migration. CONCLUSION: Using a model of DKD, we report for the first time that high glucose and inflammatory cytokines trigger aberrant Cx43 hemichannel activity, events that instigate NLRP3-induced inflammation in RPTECs and epithelial-to-macrophage crosstalk. Recapitulating observations previously reported in diabetic retinopathy, these data suggest that Cx43 hemichannel blockers (i.e., Tonabersat) may dampen multi-system damage observed in secondary complications of diabetes.


Sujet(s)
Néphropathies diabétiques , Inflammasomes , Macrophages , Protéine-3 de la famille des NLR contenant un domaine pyrine , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/anatomopathologie , Humains , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Inflammasomes/métabolisme , Connexine 43/métabolisme , Connexine 43/génétique , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Adénosine triphosphate/métabolisme , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie
12.
Int J Mol Sci ; 25(13)2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-39000465

RÉSUMÉ

The complexification of in vitro models requires the compatibility of cells with the same medium. Since immune cells are the most sensitive to growth conditions, growing intestinal epithelial cells in their usual medium seems to be necessary. This work was aimed at comparing the sensitivity of these epithelial cells to pro-inflammatory stimuli but also to dietary polyphenols in both DMEM and RPMI-1640 media. Co-cultures of Caco-2 and HT29-MTX cells were grown for 21 days in the two media before their stimulation with a cocktail of TNF-α (20 ng/mL), IL-1ß (1 ng/mL), and IFN-γ (10 ng/mL) or with LPS (10 ng/mL) from E. coli (O111:B4). The role of catechins (15 µM), a dietary polyphenol, was evaluated after its incubation with the cells before their stimulation for 6 h. The RPMI-1640 medium did not alter the intensity of the inflammatory response observed with the cytokines. By contrast, LPS failed to stimulate the co-culture in inserts regardless of the medium used. Lastly, catechins were unable to prevent the pro-inflammatory response observed with the cytokines in the two media. The preservation of the response of this model of intestinal epithelium in RPMI-1640 medium is promising when considering its complexification to evaluate the complex cellular crosstalk leading to intestinal homeostasis.


Sujet(s)
Techniques de coculture , Muqueuse intestinale , Lipopolysaccharides , Polyphénols , Humains , Techniques de coculture/méthodes , Polyphénols/pharmacologie , Cellules Caco-2 , Muqueuse intestinale/métabolisme , Muqueuse intestinale/cytologie , Muqueuse intestinale/effets des médicaments et des substances chimiques , Lipopolysaccharides/pharmacologie , Cellules HT29 , Milieux de culture/composition chimique , Milieux de culture/pharmacologie , Cytokines/métabolisme , Catéchine/pharmacologie , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie , Inflammation/métabolisme , Inflammation/anatomopathologie
13.
Int J Mol Sci ; 25(13)2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-39000481

RÉSUMÉ

Pollen, in addition to allergens, comprise low molecular weight components (LMC) smaller than 3 kDa. Emerging evidence indicates the relevance of LMC in allergic immune responses. However, the interaction of birch pollen (BP)-derived LMC and epithelial cells has not been extensively studied. We investigated epithelial barrier modifications induced by exposure to BP LMC, using the human bronchial epithelial cell line 16HBE14o-. Epithelial cell monolayers were apically exposed to the major BP allergen Bet v 1, aqueous BP extract or BP-derived LMC. Barrier integrity after the treatments was monitored by measuring transepithelial electrical resistance at regular intervals and by using the xCELLigence Real-Time Cell Analysis system. The polarized release of cytokines 24 h following treatment was measured using a multiplex immunoassay. Epithelial barrier integrity was significantly enhanced upon exposure to BP LMC. Moreover, BP LMC induced the repair of papain-mediated epithelial barrier damage. The apical release of CCL5 and TNF-α was significantly reduced after exposure to BP LMC, while the basolateral release of IL-6 significantly increased. In conclusion, the results of our study demonstrate that BP-derived LMC modify the physical and immunological properties of bronchial epithelial cells and thus regulate airway epithelial barrier responses.


Sujet(s)
Betula , Bronches , Cellules épithéliales , Masse moléculaire , Pollen , Humains , Bronches/métabolisme , Bronches/cytologie , Bronches/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Lignée cellulaire , Allergènes , Cytokines/métabolisme , Muqueuse respiratoire/métabolisme , Muqueuse respiratoire/effets des médicaments et des substances chimiques
14.
Methods Mol Biol ; 2805: 113-124, 2024.
Article de Anglais | MEDLINE | ID: mdl-39008177

RÉSUMÉ

The extracellular matrix (ECM) provides dynamic structural and molecular signals that affect the form and function of developing tissues. In order to parse how the individual features of the ECM impact cell- and tissue-level behavior during development, engineered culture models should reproduce key structural and molecular features of native ECM. Here, we describe a protocol for bioprinting epithelial cell aggregates embedded within a collagen-Matrigel ink in order to study the dynamic interplay between epithelial tissues and aligned networks of type I collagen fibers. Collagen fiber alignment and geometry can be spatially controlled by modulating the printing speed, nozzle geometry, surface chemistry, and degree of molecular crowding in the printing ink. We provide detailed procedures for generating epithelial cell aggregates, microextrusion printing collagen-Matrigel bioinks, culturing the three-dimensional (3D)-printed tissues, and imaging 3D-printed collagen-Matrigel constructs.


Sujet(s)
Bio-impression , Collagène , Cellules épithéliales , Matrice extracellulaire , Hydrogels , Impression tridimensionnelle , Ingénierie tissulaire , Bio-impression/méthodes , Hydrogels/composition chimique , Collagène/composition chimique , Collagène/métabolisme , Ingénierie tissulaire/méthodes , Cellules épithéliales/cytologie , Cellules épithéliales/métabolisme , Matrice extracellulaire/métabolisme , Matrice extracellulaire/composition chimique , Animaux , Morphogenèse , Humains , Protéoglycanes/composition chimique , Protéoglycanes/métabolisme , Structures d'échafaudage tissulaires/composition chimique , Laminine/composition chimique , Association médicamenteuse , Chiens , Épithélium/métabolisme , Épithélium/croissance et développement
15.
Sci Rep ; 14(1): 15635, 2024 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-38972889

RÉSUMÉ

This study aimed to elucidate the influence of miR-483-3p on human renal tubular epithelial cells (HK-2) under high glucose conditions and to understand its mechanism. Human proximal tubular epithelial cells (HK-2) were exposed to 50 mmol/L glucose for 48 h to establish a renal tubular epithelial cell injury model, denoted as the high glucose group (HG group). Cells were also cultured for 48 h in a medium containing 5.5 mmol/L glucose, serving as the low glucose group. Transfection was performed in various groups: HK-2 + low glucose (control group), high glucose (50 mM) (HG group), high glucose + miR-483-3p mimics (HG + mimics group), high glucose +miR-483-3p inhibitor (HG + inhibitor group), and corresponding negative controls. Real-time quantitative polymerase chain reaction (qPCR) assessed the mRNA expression of miR-483-3p, bax, bcl-2, and caspase-3. Western blot determined the corresponding protein levels. Proliferation was assessed using the CCK-8 assay, and cell apoptosis was analyzed using the fluorescence TUNEL method. Western blot and Masson's staining were conducted to observe alterations in cell fibrosis post miR-483-3p transfection. Furthermore, a dual-luciferase assay investigated the targeting relationship between miR-483-3p and IGF-1. The CCK8 assay demonstrated that the HG + mimics group inhibited HK-2 cell proliferation, while the fluorescent TUNEL method revealed induced cell apoptosis in this group. Conversely, the HG + inhibitor group promoted cell proliferation and suppressed cell apoptosis. The HG + mimics group upregulated mRNA and protein expression of pro-apoptotic markers (bax and caspase-3), while downregulating anti-apoptotic marker (bcl-2) expression. In contrast, the HG + inhibitor group showed opposite effects. Collagen I and FN protein levels were significantly elevated in the HG + mimics group compared to controls (P < 0.05). Conversely, in the HG + inhibitor group, the protein expression of Collagen I and FN was notably reduced compared to the HG group (P < 0.05). The dual luciferase reporter assay confirmed that miR-483-3p could inhibit the luciferase activity of IGF-1's 3'-UTR region (P < 0.05). miR-483-3p exerts targeted regulation on IGF-1, promoting apoptosis and fibrosis in renal tubular epithelial cells induced by high glucose conditions.


Sujet(s)
Apoptose , Prolifération cellulaire , Cellules épithéliales , Glucose , Facteur de croissance IGF-I , Tubules rénaux , microARN , Humains , microARN/génétique , microARN/métabolisme , Glucose/pharmacologie , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Facteur de croissance IGF-I/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire , Tubules rénaux/métabolisme , Tubules rénaux/cytologie , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/cytologie , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Caspase-3/métabolisme , Caspase-3/génétique
16.
Nutrients ; 16(13)2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38999871

RÉSUMÉ

IL-17A drives inflammation and oxidative stress, affecting the progression of chronic lung diseases (asthma, chronic obstructive pulmonary disease (COPD), lung cancer, and cystic fibrosis). Oleuropein (OLP) is a polyphenolic compound present in olive oil and widely included in the Mediterranean diet. It exerts antioxidant and anti-inflammatory activities, oxidative stress resistance, and anticarcinogenic effects with a conceivable positive impact on human health. We hypothesized that OLP positively affects the mechanisms of oxidative stress, apoptosis, DNA damage, cell viability during proliferation, and cell growth in alveolar epithelial cells and tested its effect in a human alveolar epithelial cell line (A549) in the presence of IL-17A. Our results show that OLP decreases the levels of oxidative stress (Reactive Oxygen Species, Mitochondrial membrane potential) and DNA damage (H2AX phosphorylation-ser139, Olive Tail Moment data) and increases cell apoptosis in A549 cells exposed to IL-17A. Furthermore, OLP decreases the number of viable cells during proliferation, the migratory potential (Scratch test), and the single cell capacity to grow within colonies as a cancer phenotype in A549 cells exposed to IL-17A. In conclusion, we suggest that OLP might be useful to protect lung epithelial cells from oxidative stress, DNA damage, cell growth, and cell apoptosis. This effect might be exerted in lung diseases by the downregulation of IL-17A activities. Our results suggest a positive effect of the components of olive oil on human lung health.


Sujet(s)
Apoptose , Prolifération cellulaire , Altération de l'ADN , Interleukine-17 , Glucosides d'iridoïdes , Iridoïdes , Stress oxydatif , Humains , Stress oxydatif/effets des médicaments et des substances chimiques , Interleukine-17/métabolisme , Glucosides d'iridoïdes/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules A549 , Altération de l'ADN/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Iridoïdes/pharmacologie , Espèces réactives de l'oxygène/métabolisme , Antioxydants/pharmacologie , Survie cellulaire/effets des médicaments et des substances chimiques , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Huile d'olive/pharmacologie , Pneumocytes/effets des médicaments et des substances chimiques , Pneumocytes/métabolisme
17.
Int J Mol Sci ; 25(13)2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38999957

RÉSUMÉ

Abnormalities in mucosal immunity are involved in the onset and progression of ulcerative colitis (UC), resulting in a high incidence of colorectal cancer (CRC). While high-mobility group box-1 (HMGB1) is overexpressed during colorectal carcinogenesis, its role in UC-related carcinogenesis remains unclear. In the present study, we investigated the role of HMGB1 in UC-related carcinogenesis and sporadic CRC. Both the azoxymethane colon carcinogenesis and dextran sulfate sodium colitis carcinogenesis models demonstrated temporal increases in mucosal HMGB1 levels. Activated CD8+ cells initially increased and then decreased, whereas exhausted CD8+ cells increased. Additionally, we observed increased regulatory CD8+ cells, decreased naïve CD8+ cells, and decreased mucosal epithelial differentiation. In the in vitro study, HMGB1 induced energy reprogramming from oxidative phosphorylation to glycolysis in CD8+ cells and intestinal epithelial cells. Furthermore, in UC dysplasia, UC-related CRC, and hyperplastic mucosa surrounding human sporadic CRC, we found increased mucosal HMGB1, decreased activated CD8+ cells, and suppressed mucosal epithelial differentiation. However, we observed increased activated CD8+ cells in active UC mucosa. These findings indicate that HMGB1 plays an important role in modulating mucosal immunity and epithelial dedifferentiation in both UC-related carcinogenesis and sporadic CRC.


Sujet(s)
Lymphocytes T CD8+ , Différenciation cellulaire , Rectocolite hémorragique , Protéine HMGB1 , Immunité muqueuse , Muqueuse intestinale , Protéine HMGB1/métabolisme , Humains , Muqueuse intestinale/métabolisme , Muqueuse intestinale/immunologie , Muqueuse intestinale/anatomopathologie , Rectocolite hémorragique/anatomopathologie , Rectocolite hémorragique/immunologie , Rectocolite hémorragique/métabolisme , Rectocolite hémorragique/induit chimiquement , Animaux , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Souris , Mâle , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Tumeurs colorectales/immunologie , Souris de lignée C57BL , Carcinogenèse/immunologie , Carcinogenèse/anatomopathologie , Carcinogenèse/métabolisme
18.
Int J Mol Sci ; 25(13)2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-39000023

RÉSUMÉ

Chronic exposure to harmful pollutants, chemicals, and pathogens from the environment can lead to pathological changes in the epithelial barrier, which increase the risk of developing an allergy. During allergic inflammation, epithelial cells send proinflammatory signals to group 2 innate lymphoid cell (ILC2s) and eosinophils, which require energy and resources to mediate their activation, cytokine/chemokine secretion, and mobilization of other cells. This review aims to provide an overview of the metabolic regulation in allergic asthma, atopic dermatitis (AD), and allergic rhinitis (AR), highlighting its underlying mechanisms and phenotypes, and the potential metabolic regulatory roles of eosinophils and ILC2s. Eosinophils and ILC2s regulate allergic inflammation through lipid mediators, particularly cysteinyl leukotrienes (CysLTs) and prostaglandins (PGs). Arachidonic acid (AA)-derived metabolites and Sphinosine-1-phosphate (S1P) are significant metabolic markers that indicate immune dysfunction and epithelial barrier dysfunction in allergy. Notably, eosinophils are promoters of allergic symptoms and exhibit greater metabolic plasticity compared to ILC2s, directly involved in promoting allergic symptoms. Our findings suggest that metabolomic analysis provides insights into the complex interactions between immune cells, epithelial cells, and environmental factors. Potential therapeutic targets have been highlighted to further understand the metabolic regulation of eosinophils and ILC2s in allergy. Future research in metabolomics can facilitate the development of novel diagnostics and therapeutics for future application.


Sujet(s)
Hypersensibilité , Humains , Hypersensibilité/métabolisme , Hypersensibilité/immunologie , Animaux , Granulocytes éosinophiles/métabolisme , Granulocytes éosinophiles/immunologie , Cellules épithéliales/métabolisme , Cellules épithéliales/immunologie , Immunité innée , Eczéma atopique/immunologie , Eczéma atopique/métabolisme , Eczéma atopique/anatomopathologie , Lymphocytes/métabolisme , Lymphocytes/immunologie , Rhinite allergique/métabolisme , Rhinite allergique/immunologie
19.
Int J Mol Sci ; 25(13)2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-39000093

RÉSUMÉ

Deoxynivalenol (DON) is a mycotoxin produced by Fusarium graminearum, and curcumin (CUR) is a natural polyphenolic compound found in turmeric. However, the combined treatment of CUR and DON to explore the mitigating effect of CUR on DON and their combined mechanism of action is not clear. Therefore, in this study, we established four treatment groups (CON, CUR, DON and CUR + DON) to investigate their mechanism in the porcine intestinal epithelial cells (IPEC-J2). In addition, the cross-talk and alleviating potential of CUR interfering with DON-induced cytotoxic factors were evaluated by in vitro experiments; the results showed that CUR could effectively inhibit DON-exposed activated TNF-α/NF-κB pathway, attenuate DON-induced apoptosis, and alleviate DON-induced endoplasmic reticulum stress and oxidative stress through PERK/CHOP pathways, which were verified at both mRNA and protein levels. In conclusion, these promising findings may contribute to the future use of CUR as a novel feed additive to protect livestock from the harmful effects of DON.


Sujet(s)
Apoptose , Curcumine , Stress du réticulum endoplasmique , Trichothécènes , Trichothécènes/pharmacologie , Trichothécènes/toxicité , Animaux , Curcumine/pharmacologie , Suidae , Apoptose/effets des médicaments et des substances chimiques , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Lignée cellulaire , Stress oxydatif/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Facteur de nécrose tumorale alpha/métabolisme
20.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-39000189

RÉSUMÉ

Impaired E-cadherin (Cdh1) functions are closely associated with cellular dedifferentiation, infiltrative tumor growth and metastasis, particularly in gastric cancer. The class-I carcinogen Helicobacter pylori (H. pylori) colonizes gastric epithelial cells and induces Cdh1 shedding, which is primarily mediated by the secreted bacterial protease high temperature requirement A (HtrA). In this study, we used human primary epithelial cell lines derived from gastroids and mucosoids from different healthy donors to investigate HtrA-mediated Cdh1 cleavage and the subsequent impact on bacterial pathogenesis in a non-neoplastic context. We found a severe impairment of Cdh1 functions by HtrA-induced ectodomain cleavage in 2D primary cells and mucosoids. Since mucosoids exhibit an intact apico-basal polarity, we investigated bacterial transmigration across the monolayer, which was partially depolarized by HtrA, as indicated by microscopy, the analyses of the transepithelial electrical resistance (TEER) and colony forming unit (cfu) assays. Finally, we investigated CagA injection and observed efficient CagA translocation and tyrosine phosphorylation in 2D primary cells and, to a lesser extent, similar effects in mucosoids. In summary, HtrA is a crucially important factor promoting the multistep pathogenesis of H. pylori in non-transformed primary gastric epithelial cells and organoid-based epithelial models.


Sujet(s)
Protéines bactériennes , Cadhérines , Cellules épithéliales , Muqueuse gastrique , Helicobacter pylori , Organoïdes , Humains , Cadhérines/métabolisme , Organoïdes/métabolisme , Cellules épithéliales/métabolisme , Cellules épithéliales/microbiologie , Muqueuse gastrique/métabolisme , Muqueuse gastrique/microbiologie , Muqueuse gastrique/anatomopathologie , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Antigènes bactériens/métabolisme , Infections à Helicobacter/métabolisme , Infections à Helicobacter/microbiologie , Infections à Helicobacter/anatomopathologie , Antigènes CD/métabolisme , Estomac/microbiologie , Estomac/anatomopathologie , Lignée cellulaire , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/microbiologie , Protéases à sérine
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...