Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 107.140
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.
Am J Reprod Immunol ; 92(1): e13893, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38958245

RÉSUMÉ

PROBLEM: Vulvovaginal candidiasis (VVC) is a common mucosal fungal infection, and Candida albicans is the main causative agent. The NLRP3 inflammasome plays an important role in VVC, but the underlying mechanism is unknown. METHOD OF STUDY: Vaginal epithelial cells were divided into three groups: control, C. albicans strain SC5314 (wild-type, WT), and WT+ Matt Cooper Compound 950 (MCC950, a specific NLRP3 inhibitor). After human vaginal epithelial cells were pretreated with 1 µmol/L MCC950 for 2 h, C. albicans (MOI = 1) was cocultured with the human vaginal epithelial cells for 12 h. The cell supernatants were collected, LDH was detected, and the IL-1ß and IL-18 levels were determined by ELISA. The expression of the pyroptosis-related proteins NLRP3, Caspase-1 p20 and GSDMD was measured by Western blotting analysis. The protein expression of the pyroptosis-related N-terminus of GSDMD (GSDMD-N) was detected by immunofluorescence. RESULTS: In this study, we showed that the WT C. albicans strain induced pyroptosis in vaginal epithelial cells, as indicated by the LDH and proinflammatory cytokine levels and the upregulated levels of the pyroptosis-related proteins NLRP3, Caspase-1 p20, and GSDMD-N. MCC950 reversed the changes in the expression of these proteins and proinflammatory cytokines in vaginal epithelial cells. CONCLUSION: C. albicans activated the NLRP3 inflammasome to induce vaginal epithelial cell pyroptosis. MCC950 inhibited the NLRP3 inflammasome, reduced vaginal epithelial cell pyroptosis, and decreased the release of inflammatory cytokines.


Sujet(s)
Candida albicans , Candidose vulvovaginale , Cellules épithéliales , Inflammasomes , Protéine-3 de la famille des NLR contenant un domaine pyrine , Pyroptose , Vagin , Femelle , Humains , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Candidose vulvovaginale/immunologie , Candidose vulvovaginale/microbiologie , Candidose vulvovaginale/métabolisme , Cellules épithéliales/immunologie , Cellules épithéliales/métabolisme , Inflammasomes/métabolisme , Inflammasomes/immunologie , Candida albicans/immunologie , Vagin/microbiologie , Vagin/immunologie , Vagin/anatomopathologie , Interleukine-18/métabolisme , Interleukine-1 bêta/métabolisme , Indènes , Furanes/pharmacologie , Caspase-1/métabolisme , Composés hétérocycliques avec 4 noyaux ou plus/pharmacologie , Protéines de liaison aux phosphates/métabolisme , Cellules cultivées , Sulfonamides
4.
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
5.
Biomed Microdevices ; 26(3): 32, 2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38963644

RÉSUMÉ

Fetal membrane (amniochorion), the innermost lining of the intrauterine cavity, surround the fetus and enclose amniotic fluid. Unlike unidirectional blood flow, amniotic fluid subtly rocks back and forth, and thus, the innermost amnion epithelial cells are continuously exposed to low levels of shear stress from fluid undulation. Here, we tested the impact of fluid motion on amnion epithelial cells (AECs) as a bearer of force impact and their potential vulnerability to cytopathologic changes that can destabilize fetal membrane functions. A previously developed amnion membrane (AM) organ-on-chip (OOC) was utilized but with dynamic flow to culture human fetal amnion membrane cells. The applied flow was modulated to perfuse culture media back and forth for 48 h to mimic fluid motion. A static culture condition was used as a negative control, and oxidative stress (OS) condition was used as a positive control representing pathophysiological changes. The impacts of fluidic motion were evaluated by measuring cell viability, cellular transition, and inflammation. Additionally, scanning electron microscopy (SEM) imaging was performed to observe microvilli formation. The results show that regardless of the applied flow rate, AECs and AMCs maintained their viability, morphology, innate meta-state, and low production of pro-inflammatory cytokines. E-cadherin expression and microvilli formation in the AECs were upregulated in a flow rate-dependent fashion; however, this did not impact cellular morphology or cellular transition or inflammation. OS treatment induced a mesenchymal morphology, significantly higher vimentin to cytokeratin 18 (CK-18) ratio, and pro-inflammatory cytokine production in AECs, whereas AMCs did not respond in any significant manner. Fluid motion and shear stress, if any, did not impact AEC cell function and did not cause inflammation. Thus, when using an amnion membrane OOC model, the inclusion of a dynamic flow environment is not necessary to mimic in utero physiologic cellular conditions of an amnion membrane.


Sujet(s)
Liquide amniotique , Membranes extraembryonnaires , Laboratoires sur puces , Humains , Liquide amniotique/cytologie , Membranes extraembryonnaires/cytologie , Membranes extraembryonnaires/métabolisme , Amnios/cytologie , Amnios/métabolisme , Survie cellulaire , Cellules épithéliales/cytologie , Cellules épithéliales/métabolisme , Déplacement , Stress oxydatif , Modèles biologiques , Systèmes microphysiologiques
7.
J Biomed Opt ; 29(7): 076002, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38966847

RÉSUMÉ

Significance: Optical coherence tomography has great utility for capturing dynamic processes, but such applications are particularly data-intensive. Samples such as biological tissues exhibit temporal features at varying time scales, which makes data reduction challenging. Aim: We propose a method for capturing short- and long-term correlations of a sample in a compressed way using non-uniform temporal sampling to reduce scan time and memory overhead. Approach: The proposed method separates the relative contributions of white noise, fluctuating features, and stationary features. The method is demonstrated on mammary epithelial cell spheroids in three-dimensional culture for capturing intracellular motility without loss of signal integrity. Results: Results show that the spatial patterns of motility are preserved and that hypothesis tests of spheroids treated with blebbistatin, a motor protein inhibitor, are unchanged with up to eightfold compression. Conclusions: The ability to measure short- and long-term correlations compressively will enable new applications in (3+1)D imaging and high-throughput screening.


Sujet(s)
Tomographie par cohérence optique , Tomographie par cohérence optique/méthodes , Humains , Sphéroïdes de cellules/effets des médicaments et des substances chimiques , Mouvement cellulaire/physiologie , Mouvement cellulaire/effets des médicaments et des substances chimiques , Traitement d'image par ordinateur/méthodes , Cellules épithéliales/effets des médicaments et des substances chimiques , Algorithmes , Composés hétérocycliques avec 4 noyaux ou plus
8.
Sci Adv ; 10(27): eadk8958, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38959315

RÉSUMÉ

The luminal-to-basal transition in mammary epithelial cells (MECs) is accompanied by changes in epithelial cell lineage plasticity; however, the underlying mechanism remains elusive. Here, we report that deficiency of Frmd3 inhibits mammary gland lineage development and induces stemness of MECs, subsequently leading to the occurrence of triple-negative breast cancer. Loss of Frmd3 in PyMT mice results in a luminal-to-basal transition phenotype. Single-cell RNA sequencing of MECs indicated that knockout of Frmd3 inhibits the Notch signaling pathway. Mechanistically, FERM domain-containing protein 3 (FRMD3) promotes the degradation of Disheveled-2 by disrupting its interaction with deubiquitinase USP9x. FRMD3 also interrupts the interaction of Disheveled-2 with CK1, FOXK1/2, and NICD and decreases Disheveled-2 phosphorylation and nuclear localization, thereby impairing Notch-dependent luminal epithelial lineage plasticity in MECs. A low level of FRMD3 predicts poor outcomes for breast cancer patients. Together, we demonstrated that FRMD3 is a tumor suppressor that functions as an endogenous activator of the Notch signaling pathway, facilitating the basal-to-luminal transformation in MECs.


Sujet(s)
Cellules épithéliales , Récepteurs Notch , Transduction du signal , Animaux , Cellules épithéliales/métabolisme , Femelle , Récepteurs Notch/métabolisme , Humains , Souris , Lignage cellulaire , Glandes mammaires animales/métabolisme , Glandes mammaires animales/cytologie , Protéines suppresseurs de tumeurs/métabolisme , Protéines suppresseurs de tumeurs/génétique , Différenciation cellulaire , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/génétique
9.
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
10.
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
11.
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
12.
Nat Commun ; 15(1): 5645, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38969629

RÉSUMÉ

Many critical biological processes, like wound healing, require densely packed cell monolayers/tissues to transition from a jammed solid-like to a fluid-like state. Although numerical studies anticipate changes in the cell shape alone can lead to unjamming, experimental support for this prediction is not definitive because, in living systems, fluidization due to density changes cannot be ruled out. Additionally, a cell's ability to modulate its motility only compounds difficulties since even in assemblies of rigid active particles, changing the nature of self-propulsion has non-trivial effects on the dynamics. Here, we design and assemble a monolayer of synthetic cell-mimics and examine their collective behaviour. By systematically increasing the persistence time of self-propulsion, we discovered a cell shape-driven, density-independent, re-entrant jamming transition. Notably, we observed cell shape and shape variability were mutually constrained in the confluent limit and followed the same universal scaling as that observed in confluent epithelia. Dynamical heterogeneities, however, did not conform to this scaling, with the fast cells showing suppressed shape variability, which our simulations revealed is due to a transient confinement effect of these cells by their slower neighbors. Our experiments unequivocally establish a morphodynamic link, demonstrating that geometric constraints alone can dictate epithelial jamming/unjamming.


Sujet(s)
Forme de la cellule , Cellules artificielles , Mouvement cellulaire , Modèles biologiques , Animaux , Cellules épithéliales , Humains
13.
Immun Inflamm Dis ; 12(7): e1301, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38967361

RÉSUMÉ

OBJECTIVE: Acute pancreatitis (AP) stands as a frequent cause for clinical emergency hospital admissions. The X-box binding protein 1 (XBP1) was found to be implicated in pancreatic acinar cell apoptosis. The objective is to unveil the potential mechanisms governed by XBP1 and SIRT6 in the context of AP. METHODS: Caerulein-treated human pancreatic duct epithelial (HPDE) cells to establish an in vitro research model. The levels and regulatory role of SIRT6 in the treated cells were evaluated, including its effects on inflammatory responses, oxidative stress, apoptosis, and endoplasmic reticulum stress. The relationship between XBP1 and SIRT6 was explored by luciferase and ChIP experiments. Furthermore, the effect of XBP1 overexpression on the regulatory function of SIRT6 on cells was evaluated. RESULTS: Caerulein promoted the decrease of SIRT6 and the increase of XBP1 in HPDE cells. Overexpression of SIRT6 slowed down the secretion of inflammatory factors, oxidative stress, apoptosis level, and endoplasmic reticulum stress in HPDE cells. However, XBP1 negatively regulated SIRT6, and XBP1 overexpression partially reversed the regulation of SIRT6 on the above aspects. CONCLUSION: Our study illuminates the role of XBP1 in downregulating SIRT6 in HPDE cells, thereby promoting cellular injury. Inhibiting XBP1 or augmenting SIRT6 levels holds promise in preserving cell function and represents a potential therapeutic avenue in the management of AP.


Sujet(s)
Apoptose , Régulation négative , Cellules épithéliales , Conduits pancréatiques , Pancréatite , Sirtuines , Protéine-1 liant la boite X , Humains , Sirtuines/métabolisme , Sirtuines/génétique , Cellules épithéliales/métabolisme , Protéine-1 liant la boite X/métabolisme , Protéine-1 liant la boite X/génétique , Pancréatite/métabolisme , Pancréatite/anatomopathologie , Conduits pancréatiques/métabolisme , Conduits pancréatiques/anatomopathologie , Stress du réticulum endoplasmique , Stress oxydatif , Lignée cellulaire , Céruléine/toxicité
14.
Sci Rep ; 14(1): 15195, 2024 07 02.
Article de Anglais | MEDLINE | ID: mdl-38956443

RÉSUMÉ

The intestinal epithelium dynamically controls cell cycle, yet no experimental platform exists for directly analyzing cell cycle phases in non-immortalized human intestinal epithelial cells (IECs). Here, we present two reporters and a complete platform for analyzing cell cycle phases in live primary human IECs. We interrogate the transcriptional identity of IECs grown on soft collagen, develop two fluorescent cell cycle reporter IEC lines, design and 3D print a collagen press to make chamber slides for optimal imaging while supporting primary human IEC growth, live image cell cycle dynamics, then assemble a computational pipeline building upon free-to-use programs for semi-automated analysis of cell cycle phases. The PIP-FUCCI construct allows for assigning cell cycle phase from a single image of living cells, and our PIP-H2A construct allows for semi-automated direct quantification of cell cycle phase lengths using our publicly available computational pipeline. Treating PIP-FUCCI IECs with oligomycin demonstrates that inhibiting mitochondrial respiration lengthens G1 phase, and PIP-H2A cells allow us to measure that oligomycin differentially lengthens S and G2/M phases across heterogeneous IECs. These platforms provide opportunities for future studies on pharmaceutical effects on the intestinal epithelium, cell cycle regulation, and more.


Sujet(s)
Cycle cellulaire , Cellules épithéliales , Muqueuse intestinale , Humains , Cellules épithéliales/cytologie , Cellules épithéliales/métabolisme , Muqueuse intestinale/cytologie , Muqueuse intestinale/métabolisme , Oligomycines/pharmacologie , Cellules cultivées
15.
J Vis Exp ; (208)2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38975789

RÉSUMÉ

Lens epithelial cells (LECs) play multiple important roles in maintaining the homeostasis and normal function of the lens. LECs determine lens growth, development, size, and transparency. Conversely, dysfunctional LECs can lead to cataract formation and posterior capsule opacification (PCO). Consequently, establishing a robust primary LEC culture system is important to researchers engaged in lens development, biochemistry, cataract therapeutics, and PCO prevention. However, cultivating primary LECs has long presented challenges due to their limited availability, slow proliferation rate, and delicate nature. This study addresses these hurdles by presenting a comprehensive protocol for primary LEC culture. The protocol encompasses essential steps such as the formulation of an optimized culture medium, precise isolation of lens capsules, trypsinization techniques, subculture procedures, harvest protocols, and guidelines for storage and shipment. Throughout the culture process, cell morphology was monitored using phase-contrast microscopy. To confirm the authenticity of the cultured LECs, immunofluorescence assays were conducted to detect the presence and subcellular distribution of critical lens proteins, namely αA- and γ-crystallins. This detailed protocol equips researchers with a valuable resource for cultivating and characterizing primary LECs, enabling advancements in our comprehension of lens biology and the development of therapeutic strategies for lens-related disorders.


Sujet(s)
Cellules épithéliales , Cristallin , Trypsine , Cellules épithéliales/cytologie , Cristallin/cytologie , Animaux , Souris , Trypsine/composition chimique , Trypsine/métabolisme , Techniques de culture cellulaire/méthodes , Culture de cellules primaires/méthodes
16.
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
17.
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
18.
Adv Exp Med Biol ; 1445: 11-36, 2024.
Article de Anglais | MEDLINE | ID: mdl-38967747

RÉSUMÉ

Although V(D)J recombination and immunoglobulin (Ig) production are traditionally recognised to occur only in B lymphocytes and plasma cells, the expression of Igs in non-lymphoid cells, which we call non B cell-derived Igs (non B Igs), has been documented by growing studies. It has been demonstrated that non B-Igs can be widely expressed in most cell types, including, but not limited to, epithelial cells, cardiomyocytes, hematopoietic stem/progenitor cells, myeloid cells, and cells from immune-privileged sites, such as neurons and spermatogenic cells. In particular, malignant tumour cells express high level of IgG. Moreover, different from B-Igs that mainly localised on the B cell membrane and in the serum and perform immune defence function mainly, non B-Igs have been found to distribute more widely and play critical roles in immune defence, maintaining cell proliferation and survival, and promoting progression. The findings of non B-Igs may provide a wealthier breakthrough point for more therapeutic strategies for a wide range of immune-related diseases.


Sujet(s)
Immunoglobulines , Humains , Animaux , Immunoglobulines/génétique , Immunoglobulines/métabolisme , Immunoglobulines/immunologie , Cellules souches hématopoïétiques/métabolisme , Cellules souches hématopoïétiques/immunologie , Cellules souches hématopoïétiques/cytologie , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Cellules épithéliales/métabolisme , Cellules épithéliales/immunologie , Myocytes cardiaques/métabolisme , Myocytes cardiaques/immunologie , Cellules myéloïdes/immunologie , Cellules myéloïdes/métabolisme
19.
Adv Exp Med Biol ; 1445: 157-168, 2024.
Article de Anglais | MEDLINE | ID: mdl-38967758

RÉSUMÉ

As the locus for air exchange, lung tissue is perpetually exposed to a significant quantity of foreign pathogens. Consequently, lung has developed a refined and intricate immune system. Beyond their physical and chemical barrier roles, lung epithelial cells can contribute to immune defence through the expression of Toll-like receptors (TLRs) and other pattern recognition receptors, along with the secretion of cytokines. Emerging evidence demonstrates that lung epithelial cells can generate and secrete immunoglobulins (Igs), including IgM, IgA, or IgG, thus performing antibody function. Moreover, malignantly transformed lung epithelial cells have been discovered to produce high levels of Ig, predominantly IgG, which do not fulfill the role of antibodies, but instead carries out tumour-promoting activity. Structural analysis has indicated that the biological activity of IgG produced by lung cancer cells differs from that of Igs produced by normal lung epithelial cells due to the unique glycosylation modification. Specifically, the sialylated IgG (SIA-IgG), characterised by a non-traditional N-glycosylation modification at the Asn162 site of Igγ CH1, is highly expressed in tumour stem cells. It has been demonstrated that SIA-IgG relies on this unique sialylation modification to promote tumorigenesis, metastasis, and immune evasion. Current results have proven that the Ig produced by lung epithelial cells has multifaceted biological activities, including immune defence functions under physiological conditions, while acquiring tumour-promoting activity during malignant transformation. These insights possess potential for the diagnosis and treatment of lung cancer as novel biomarkers and targets.


Sujet(s)
Tumeurs du poumon , Humains , Tumeurs du poumon/immunologie , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Animaux , Cellules épithéliales/métabolisme , Cellules épithéliales/immunologie , Cellules épithéliales/anatomopathologie , Glycosylation , Poumon/immunologie , Poumon/anatomopathologie , Poumon/métabolisme , Immunoglobulines/métabolisme , Immunoglobulines/immunologie , Immunoglobuline G/immunologie , Immunoglobuline G/métabolisme
20.
Adv Exp Med Biol ; 1445: 169-177, 2024.
Article de Anglais | MEDLINE | ID: mdl-38967759

RÉSUMÉ

Over the past 20 years, increasing evidence has demonstrated that immunoglobulins (Igs) can be widely generated from non B cells, including normal and malignant mammary epithelial cells. In normal breast tissue, the expression of IgG and IgA has been identified in epithelial cells of mammary glands during pregnancy and lactation, which can be secreted into milk, and might participate in neonatal immunity. On the other hand, non B-IgG is highly expressed in breast cancer cells, correlating with the poor prognosis of patients with breast cancer. Importantly, a specific group of IgG, bearing a unique N-linked glycan on the Asn162 site and aberrant sialylation modification at the end of the novel glycan (referred to as sialylated IgG (SIA-IgG)), has been found in breast cancer stem/progenitor-like cells. SIA-IgG can significantly promote the capacity of migration, invasiveness, and metastasis, as well as enhance self-renewal and tumorigenicity in vitro and in vivo. These findings suggest that breast epithelial cells can produce Igs with different biological activities under physiological and pathological conditions. During lactation, these Igs could be the main source of milk Igs to protect newborns from pathogenic infections, while under pathological conditions, they display oncogenic activity and promote the occurrence and progression of breast cancer.


Sujet(s)
Tumeurs du sein , Cellules épithéliales , Glandes mammaires humaines , Humains , Femelle , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Tumeurs du sein/immunologie , Cellules épithéliales/métabolisme , Animaux , Glandes mammaires humaines/métabolisme , Glandes mammaires humaines/anatomopathologie , Lactation/métabolisme , Grossesse , Immunoglobuline G/métabolisme , Immunoglobuline G/immunologie , Immunoglobulines/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...