Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 8.522
Filtrer
1.
J Cell Mol Med ; 28(15): e18577, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39099000

RÉSUMÉ

Lung cancer remains the leading cause of cancer-related deaths, with cigarette smoking being the most critical factor, linked to nearly 90% of lung cancer cases. NNK, a highly carcinogenic nitrosamine found in tobacco, is implicated in the lung cancer-causing effects of cigarette smoke. Although NNK is known to mutate or activate certain oncogenes, its potential interaction with p27 in modulating these carcinogenic effects is currently unexplored. Recent studies have identified specific downregulation of p27 in human squamous cell carcinoma, in contrast to adenocarcinoma. Additionally, exposure to NNK significantly suppresses p27 expression in human bronchial epithelial cells. Subsequent studies indicates that the downregulation of p27 is pivotal in NNK-induced cell transformation. Mechanistic investigations have shown that reduced p27 expression leads to increased level of ITCH, which facilitates the degradation of Jun B protein. This degradation in turn, augments miR-494 expression and its direct regulation of JAK1 mRNA stability and protein expression, ultimately activating STAT3 and driving cell transformation. In summary, our findings reveal that: (1) the downregulation of p27 increases Jun B expression by upregulating Jun B E3 ligase ITCH, which then boosts miR-494 transcription; (2) Elevated miR-494 directly binds to 3'-UTR of JAK1 mRNA, enhancing its stability and protein expression; and (3) The JAK1/STAT3 pathway is a downstream effector of p27, mediating the oncogenic effect of NNK in lung cancer. These findings provide significant insight into understanding the participation of mechanisms underlying p27 inhibition of NNK induced lung squamous cell carcinogenic effect.


Sujet(s)
Bronches , Carcinome épidermoïde , Transformation cellulaire néoplasique , Inhibiteur p27 de kinase cycline-dépendante , Cellules épithéliales , Tumeurs du poumon , Nitrosamines , Humains , Nitrosamines/toxicité , Bronches/métabolisme , Bronches/anatomopathologie , Bronches/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/anatomopathologie , Carcinome épidermoïde/induit chimiquement , Carcinome épidermoïde/métabolisme , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/génétique , Transformation cellulaire néoplasique/métabolisme , Transformation cellulaire néoplasique/induit chimiquement , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/effets des médicaments et des substances chimiques , Inhibiteur p27 de kinase cycline-dépendante/métabolisme , Inhibiteur p27 de kinase cycline-dépendante/génétique , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/induit chimiquement , Tumeurs du poumon/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , microARN/génétique , microARN/métabolisme , Régulation négative/effets des médicaments et des substances chimiques , Cancérogènes/toxicité
2.
Eur J Med Res ; 29(1): 406, 2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-39103884

RÉSUMÉ

BACKGROUND: The diagnosis of peripheral pulmonary lesions (PPL) is still challenging. We describe a novel method for sampling PPL without bronchial signs by creating invisible tunnel under electromagnetic navigation without the transbronchial access tool (TABT). METHODS: During electromagnetic navigation, we adjust the angle of the edge extended working channel catheter based on the real-time position of the lesion in relation to the locating guide rather than preset route. A biopsy brush or biopsy forceps is used to punch a hole in the bronchial wall. A locating guide is then re-inserted to real-time navigate through the lung parenchyma to the lesion. Safety and feasibility of this method was analyzed. RESULTS: A total of 32 patients who underwent electromagnetic navigation bronchoscopy were retrieved. The mean size of the lesion is 23.1 mm. The mean operative time of all patients was 12.4 min. Ten of the patients did not have a direct airway to the lesion, thus creating an invisible tunnel. For them, the length of the tunnel from the bronchial wall POE to the lesion was 11-30 mm, with a mean length of 16.9 mm and a mean operation time of 14.1 min. Adequate samples were obtained from 32 patients (100%), and the diagnostic yield was 87.5% (28/32). Diagnostic yield of with create the invisible tunnel TBAT was 90% (9/10), and one patient undergone pneumothorax after operation. CONCLUSIONS: This method is feasible and safe as a novel approach sampling pulmonary lesions without bronchial signs, and it further improves current tunnel technique.


Sujet(s)
Bronchoscopie , Phénomènes électromagnétiques , Humains , Bronchoscopie/méthodes , Femelle , Mâle , Adulte d'âge moyen , Sujet âgé , Adulte , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/diagnostic , Tumeurs du poumon/chirurgie , Bronches/anatomopathologie , Bronches/imagerie diagnostique , Sujet âgé de 80 ans ou plus
3.
Respir Res ; 25(1): 317, 2024 Aug 19.
Article de Anglais | MEDLINE | ID: mdl-39160511

RÉSUMÉ

RATIONAL: Basal cells (BCs) are bronchial progenitor/stem cells that can regenerate injured airway that, in smokers, may undergo malignant transformation. As a model for early stages of lung carcinogenesis, we set out to characterize cytologically normal BC outgrowths from never-smokers and ever-smokers without cancers (controls), as well as from the normal epithelial "field" of ever-smokers with anatomically remote cancers, including lung adenocarcinoma (LUAD) and squamous cell carcinoma (LUSC) (cases). METHODS: Primary BCs were cultured and expanded from endobronchial brushings taken remote from the site of clinical or visible lesions/tumors. Donor subgroups were tested for growth, morphology, and underlying molecular features by qRT-PCR, RNAseq, flow cytometry, immunofluorescence, and immunoblot. RESULTS: (a) the BC population includes epithelial cell adhesion molecule (EpCAM) positive and negative cell subsets; (b) smoking reduced overall BC proliferation corresponding with a 2.6-fold reduction in the EpCAMpos/ITGA6 pos/CD24pos stem cell fraction; (c) LUSC donor cells demonstrated up to 2.8-fold increase in dysmorphic BCs; and (d) cells procured from LUAD patients displayed increased proliferation and S-phase cell cycle fractions. These differences corresponded with: (i) disparate NOTCH1/NOTCH2 transcript expression and altered expression of potential downstream (ii) E-cadherin (CDH1), tumor protein-63 (TP63), secretoglobin family 1a member 1 (SCGB1A1), and Hairy/enhancer-of-split related with YRPW motif 1 (HEY1); and (iii) reduced EPCAM and increased NK2 homeobox-1 (NKX2-1) mRNA expression in LUAD donor BCs. CONCLUSIONS: These and other findings demonstrate impacts of donor age, smoking, and lung cancer case-control status on BC phenotypic and molecular traits and may suggest Notch signaling pathway deregulation during early human lung cancer pathogenesis.


Sujet(s)
Bronches , Prolifération cellulaire , Tumeurs du poumon , Transduction du signal , Fumer , Humains , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/génétique , Transduction du signal/physiologie , Mâle , Femelle , Études cas-témoins , Adulte d'âge moyen , Prolifération cellulaire/physiologie , Fumer/effets indésirables , Fumer/métabolisme , Sujet âgé , Bronches/métabolisme , Bronches/anatomopathologie , Cellules cultivées , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/métabolisme , Carcinome épidermoïde/génétique , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/métabolisme , Adénocarcinome pulmonaire/génétique
4.
Respir Res ; 25(1): 321, 2024 Aug 22.
Article de Anglais | MEDLINE | ID: mdl-39174953

RÉSUMÉ

BACKGROUND: Mitochondria is prone to oxidative damage by endogenous and exogenous sources of free radicals, including particulate matter (PM). Given the role of mitochondria in inflammatory disorders, such as asthma and chronic obstructive pulmonary disease, we hypothesized that supplementation of vitamin D may play a protective role in PM-induced mitochondrial oxidative damages of human bronchial epithelial BEAS-2B cells. METHODS: BEAS-2B cells were pretreated with 1,25(OH)2D3, an active form of vitamin D, for 1 h prior to 24-hour exposure to PM (SRM-1648a). Oxidative stress was measured by flow cytometry. Mitochondrial functions including mitochondrial membrane potential, ATP levels, and mitochondrial DNA copy number were analyzed. Additionally, mitochondrial ultrastructure was examined using transmission electron microscopy. Intracellular and mitochondrial calcium concentration changes were assessed using flow cytometry based on the expression of Fluo-4 AM and Rhod-2 AM, respectively. Pro-inflammatory cytokines, including IL-6 and MCP-1, were quantified using ELISA. The expression levels of antioxidants, including SOD1, SOD2, CAT, GSH, and NADPH, were determined. RESULTS: Our findings first showed that 24-hour exposure to PM led to the overproduction of reactive oxygen species (ROS) derived from mitochondria. PM-induced mitochondrial oxidation resulted in intracellular calcium accumulation, particularly within mitochondria, and alterations in mitochondrial morphology and functions. These changes included loss of mitochondrial membrane integrity, disarrayed cristae, mitochondrial membrane depolarization, reduced ATP production, and increased mitochondrial DNA copy number. Consequently, PM-induced mitochondrial damage triggered the release of certain inflammatory cytokines, such as IL-6 and MCP-1. Similar to the actions of mitochondrial ROS inhibitor MitoTEMPO, 1,25(OH)2D3 conferred protective effects on mtDNA alterations, mitochondrial damages, calcium dyshomeostasis, thereby decreasing the release of certain inflammatory cytokines. We found that greater cellular level of 1,25(OH)2D3 upregulated the expression of enzymatic (SOD1, SOD2, and CAT) and non-enzymatic (GSH and NADPH) antioxidants to modulate cellular redox homeostasis. CONCLUSION: Our study provides new evidence that 1,25(OH)2D3 acts as an antioxidant, enhancing BEAS-2B antioxidant responses to regulate mitochondrial ROS homeostasis and mitochondrial function, thereby enhancing epithelial defense against air pollution exposure.


Sujet(s)
Bronches , Calcium , Cellules épithéliales , Homéostasie , Mitochondries , Matière particulaire , Humains , Matière particulaire/toxicité , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Mitochondries/ultrastructure , Calcium/métabolisme , Bronches/effets des médicaments et des substances chimiques , Bronches/métabolisme , Bronches/anatomopathologie , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Cellules épithéliales/ultrastructure , Homéostasie/effets des médicaments et des substances chimiques , Homéostasie/physiologie , Lignée cellulaire , Stress oxydatif/effets des médicaments et des substances chimiques , Stress oxydatif/physiologie , Vitamine D/pharmacologie , Espèces réactives de l'oxygène/métabolisme
5.
Toxicol Appl Pharmacol ; 490: 117035, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39019094

RÉSUMÉ

Exposure to particulate matter (PM10) can induce respiratory diseases that are closely related to bronchial hyperresponsiveness. However, the involved mechanism remains to be fully elucidated. This study aimed to demonstrate the effects of PM10 on the acetylcholine muscarinic 3 receptor (CHRM3) expression and the role of the ERK1/2 pathway in rat bronchial smooth muscle. A whole-body PM10 exposure system was used to stimulate bronchial hyperresponsiveness in rats for 2 and 4 months, accompanied by MEK1/2 inhibitor U0126 injection. The whole-body plethysmography system and myography were used to detect the pulmonary and bronchoconstrictor function, respectively. The mRNA and protein levels were determined by Western blotting, qPCR, and immunofluorescence. Enzyme-linked immunosorbent assay was used to detect the inflammatory cytokines. Compared with the filtered air group, 4 months of PM10 exposure significantly increased CHRM3-mediated pulmonary function and bronchial constriction, elevated CHRM3 mRNA and protein expression levels on bronchial smooth muscle, then induced bronchial hyperreactivity. Additionally, 4 months of PM10 exposure caused an increase in ERK1/2 phosphorylation and increased the secretion of inflammatory factors in bronchoalveolar lavage fluid. Treatment with the MEK1/2 inhibitor, U0126 inhibited the PM10 exposure-induced phosphorylation of the ERK1/2 pathway, thereby reducing the PM10 exposure-induced upregulation of CHRM3 in bronchial smooth muscle and CHRM3-mediated bronchoconstriction. U0126 could rescue PM10 exposure-induced pathological changes in the bronchus. In conclusion, PM10 exposure can induce bronchial hyperresponsiveness in rats by upregulating CHRM3, and the ERK1/2 pathway may be involved in this process. These findings could reveal a potential therapeutic target for air pollution induced respiratory diseases.


Sujet(s)
Hyperréactivité bronchique , Matière particulaire , Récepteur muscarinique de type M3 , Animaux , Hyperréactivité bronchique/induit chimiquement , Hyperréactivité bronchique/physiopathologie , Hyperréactivité bronchique/métabolisme , Mâle , Matière particulaire/toxicité , Récepteur muscarinique de type M3/métabolisme , Récepteur muscarinique de type M3/génétique , Rats , Régulation positive/effets des médicaments et des substances chimiques , Bronches/effets des médicaments et des substances chimiques , Bronches/métabolisme , Bronches/anatomopathologie , Rat Sprague-Dawley , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Muscles lisses/effets des médicaments et des substances chimiques , Muscles lisses/métabolisme , Bronchoconstriction/effets des médicaments et des substances chimiques , Cytokines/métabolisme , Cytokines/génétique , Butadiènes , Nitriles
6.
Toxicol Lett ; 399: 9-18, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38971455

RÉSUMÉ

Fine particulate matter (PM2.5) is a risk factor for pulmonary diseases and lung cancer, and inhaled PM2.5 is mainly deposited in the bronchial epithelium. In this study, we investigated the effect of long-term exposure to low-dose PM2.5 on BEAS-2B cells derived from the normal bronchial epithelium. BEAS-2B cells chronically exposed to a concentration of 5 µg/ml PM2.5 for 30 passages displayed the phenotype promoting epithelial-mesenchymal transition (EMT) and cell invasion. Cellular internalization of exosomes (designated PM2.5 Exo) extracted from BEAS-2B cells chronically exposed to low-dose PM2.5 promoted cell invasion in vitro and metastatic potential in vivo. Hence, to identify the key players driving phenotypic alterations, we analyzed microRNA (miRNA) expression profiles in PM2.5 Exo. Five miRNAs with altered expression were selected: miRNA-196b-5p, miR-135a-2-5p, miR-3117-3p, miR-218-5p, and miR-497-5p. miR-196b-5p was the most upregulated in both BEAS-2B cells and isolated exosomes after PM2.5 exposure. In a functional validation study, genetically modified exosomes overexpressing a miR-196b-5p mimic induced an enhanced invasive phenotype in BEAS-2B cells. Conversely, miR-196b-5p inhibition diminished the PM2.5-enhanced EMT and cell invasion. These findings indicate that exosomal miR-196b-5p may be a candidate biomarker for predicting the malignant behavior of the bronchial epithelium and a therapeutic target for inhibiting PM2.5-triggered pathogenesis.


Sujet(s)
Bronches , Cellules épithéliales , Transition épithélio-mésenchymateuse , Exosomes , Tumeurs du poumon , microARN , Matière particulaire , microARN/génétique , microARN/métabolisme , Humains , Exosomes/métabolisme , Exosomes/génétique , Exosomes/effets des médicaments et des substances chimiques , Matière particulaire/toxicité , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/induit chimiquement , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Bronches/effets des médicaments et des substances chimiques , Bronches/métabolisme , Bronches/anatomopathologie , Bronches/cytologie , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Invasion tumorale , Animaux , Lignée cellulaire , Mouvement cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale
7.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167349, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39002703

RÉSUMÉ

Asthma is a chronic respiratory disease characterized by airway inflammation and remodeling. Epithelial-mesenchymal transition (EMT) of bronchial epithelial cells is considered to be a crucial player in asthma. Methyltransferase-like 14 (METTL14), an RNA methyltransferase, is implicated in multiple pathological processes, including EMT, cell proliferation and migration. However, the role of METTL14 in asthma remains uncertain. This research aimed to explore the biological functions of METTL14 in asthma and its underlying upstream mechanisms. METTL14 expression was down-regulated in asthmatic from three GEO datasets (GSE104468, GSE165934, and GSE74986). Consistent with this trend, METTL14 was decreased in the lung tissues of OVA-induced asthmatic mice and transforming growth factor-ß1 (TGF-ß1)-stimulated human bronchial epithelial cells (Beas-2B) in this study. Overexpression of METTL14 caused reduction in mesenchymal markers (FN1, N-cad, Col-1 and α-SMA) in TGF-ß1-treated cells, but caused increase in epithelial markers (E-cad), thus inhibiting EMT. Also, METTL14 suppressed the proliferation and migration ability of TGF-ß1-treated Beas-2B cells. Two transcription factors, ETS1 and RBPJ, could both bind to the promoter region of METTL14 and drive its expression. Elevating METTL14 expression could reversed EMT, cell proliferation and migration promoted by ETS1 or RBPJ deficiency. These results indicate that the ETS1/METTL14 and RBPJ/METTL14 transcription axes exhibit anti-EMT, anti-proliferation and anti-migration functions in TGF-ß1-induced bronchial epithelial cells, implying that METTL14 may be considered an alternative candidate target for the treatment of asthma.


Sujet(s)
Asthme , Bronches , Cellules épithéliales , Transition épithélio-mésenchymateuse , Methyltransferases , Protéine proto-oncogène c-ets-1 , Facteur de croissance transformant bêta-1 , Humains , Facteur de croissance transformant bêta-1/métabolisme , Facteur de croissance transformant bêta-1/génétique , Methyltransferases/métabolisme , Methyltransferases/génétique , Animaux , Bronches/métabolisme , Bronches/anatomopathologie , Bronches/cytologie , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Souris , Protéine proto-oncogène c-ets-1/métabolisme , Protéine proto-oncogène c-ets-1/génétique , Asthme/anatomopathologie , Asthme/métabolisme , Asthme/génétique , Lignée cellulaire , Prolifération cellulaire , Souris de lignée BALB C , Mouvement cellulaire , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques
8.
PLoS One ; 19(7): e0299447, 2024.
Article de Anglais | MEDLINE | ID: mdl-39052646

RÉSUMÉ

OBJECTIVE: To establish the diagnostic accuracy of RASSF1A (Ras association domain family 1 isoform) methylation using bronchial aspirates as an auxiliary method for diagnosing lung cancer through a systematic review and meta-analysis. METHODS: Studies published prior to October 30, 2022, were retrieved from the Embase, PubMed, Web of Science, and Wan Fang databases using the keywords "lung cancer", "RASSF1A", "methylation", and "bronchial aspirates". A fixed or random effect model was used to calculate the combined sensitivity, specificity, positive likelihood ratios (LR), negative LR, diagnostic odds ratio (DOR), along with the respective 95% confidence intervals (CIs) and the area under the curve (AUC) with Q index. The threshold effect was defined by using the Spearman correlation coefficient, and the Deeks funnel plot was generated to evaluate publication bias. RESULTS: Among the 12 trials that met the inclusion criteria, a total of 2388 participants were involved. The pooled results for the diagnosis of lung cancer were as follows, when compared to the pathological diagnosis: sensitivity of 0.47 (95% CI: 0.45-0.50), specificity of 0.96 (95% CI: 0.95-0.97), positive LR of 12.18 (95% CI: 8.96-16.55), negative LR of 0.56 (95% CI: 0.52-0.61), DOR of 24.05 (95% CI: 17.29-33.47), and AUC of 0.78 (Q index = 0.72), respectively. The sensitivity of the RASSF1A methylation assay was relatively low in a detailed subgroup analysis, fluctuating between 0.39 and 0.90, indicating a limitation in its diagnostic value for lung cancer. The RASSF1A methylation assay, on the other hand, demonstrated excellent specificity, suggesting a high exclusion value. Of note, the diagnostic sensitivity, specificity, DOR, and AUC for small cell lung cancer were 0.90 (0.84-0.94), 0.95 (0.94-0.97), 249.5 (103.94-598.8), and 0.98, respectively, showing that RASSF1A methylation was a promising biomarker for diagnosing small cell lung cancer with both high diagnostic and exclusion value. Furthermore, RASSF1A methylation using bronchial washings and bronchial aspirates showed a high AUC of 0.998 and 0.93, respectively, indicating excellent diagnostic performance. CONCLUSIONS: The methylation of RASSF1A in bronchial aspirates demonstrated a high level of diagnostic accuracy and has the potential to be a valuable supplementary diagnostic method, especially for identifying small cell lung cancer.


Sujet(s)
Méthylation de l'ADN , Tumeurs du poumon , Protéines suppresseurs de tumeurs , Humains , Tumeurs du poumon/diagnostic , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Protéines suppresseurs de tumeurs/génétique , Protéines suppresseurs de tumeurs/métabolisme , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Sensibilité et spécificité , Bronches/métabolisme , Bronches/anatomopathologie
9.
Pathol Res Pract ; 261: 155482, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39067173

RÉSUMÉ

BACKGROUND: BPD is a refractory disease affecting preterm infants with alveolar dysplasia and declined pulmonary function. However, the molecular mechanism underlying BPD is largely unknown. To explore the pathogenic mechanism of BPD and to facilitate better diagnosis and treatment of this disease. METHOD: The DEMs and DEGs in BPD vs. Control samples from the miRNA expression data in GSE108754 and mRNA expression data in the GSE108755 were screened, followed by the construction of the miRNA-mRNA regulatory network. DEGs PPI network and hub DEGs analysis were constructed by using the STRING database and Cytoscape software. Functional and pathway enrichment analyses were then performed for these DEGs and DEMs based on the ClusterProfiler package in the R and the miRWalk database. The k-mean algorithm is used to perform clustering analysis of DEGs. Cellular experiments (flow cytometry, western blot, RT-PCR, dual-luciferase reporter assay) were used to validate the results of bioinformatics. RESULTS: We obtained 20 DEMs and 262 DEGs. A 15 DEMs-11 DEGs regulatory network was constructed. miR-3202-RAG1 is a core sub-network. Hyperoxia induced a cell model of BPD. The upregulation of RAG1 and downregulation of miR-3202 were observed in BPD cells. Furthermore, siRNA targeting RAG1 was transfected into BEAS-2B cells to inhibit its expression and miR-3202 mimics was transfected into the cells to increase its expression. Inhibition of RAG1 and elevation of miR-3202 inhibit cell apoptosis and reduce ROS level caused by hyperoxia. A double-luciferase reporter assay revealed that miR-3202 directly targets RAG1. CONCLUSION: The miRNA-3202/RAG1 axis contributes into BPD-induced cell apoptosis and ROS production. The present study provides a probable target for the treatment of BPD.


Sujet(s)
Apoptose , Dysplasie bronchopulmonaire , Cellules épithéliales , microARN , Stress oxydatif , Humains , microARN/génétique , microARN/métabolisme , Apoptose/génétique , Stress oxydatif/génétique , Dysplasie bronchopulmonaire/génétique , Dysplasie bronchopulmonaire/métabolisme , Dysplasie bronchopulmonaire/anatomopathologie , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Bronches/anatomopathologie , Bronches/métabolisme , Nouveau-né
10.
Bull Exp Biol Med ; 177(1): 93-97, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38963595

RÉSUMÉ

Squamous cell lung cancer (SCLC) occurs as a result of dysregenerative changes in the bronchial epithelium: basal cell hyperplasia (BCH), squamous cell metaplasia (SM), and dysplasia. We previously suggested that combinations of precancerous changes detected in the small bronchi of patients with SCLC may reflect various "scenarios" of the precancerous process: isolated BCH→stopping at the stage of hyperplasia, BCH+SM→progression of hyperplasia into metaplasia, SM+dysplasia→progression of metaplasia into dysplasia. In this study, DNA methylome of various forms of precancerous changes in the bronchial epithelium of SCLC patients was analyzed using the genome-wide bisulfite sequencing. In BCH combined with SM, in contrast to isolated BCH, differentially methylated regions were identified in genes of the pathogenetically significant MET signaling pathway (RNMT, HPN). Differentially methylated regions affecting genes involved in inflammation regulation (IL-23, IL-23R, IL12B, IL12RB1, and FIS1) were detected in SM combined with dysplasia in comparison with SM combined with BCH. The revealed changes in DNA methylation may underlie various "scenarios" of the precancerous process in the bronchial epithelium.


Sujet(s)
Bronches , Méthylation de l'ADN , Hyperplasie , Tumeurs du poumon , Métaplasie , États précancéreux , Humains , Hyperplasie/anatomopathologie , Hyperplasie/génétique , Métaplasie/génétique , Métaplasie/anatomopathologie , Métaplasie/métabolisme , Bronches/anatomopathologie , Bronches/métabolisme , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , États précancéreux/génétique , États précancéreux/anatomopathologie , États précancéreux/métabolisme , Mâle , Femelle , Adulte d'âge moyen , Épigénome/génétique , Muqueuse respiratoire/anatomopathologie , Muqueuse respiratoire/métabolisme , Sujet âgé , Carcinome pulmonaire à petites cellules/génétique , Carcinome pulmonaire à petites cellules/anatomopathologie , Carcinome pulmonaire à petites cellules/métabolisme , Carcinome épidermoïde/génétique , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/métabolisme
11.
Pathol Res Pract ; 260: 155423, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38909404

RÉSUMÉ

Curcumin, the principal curcuminoid of turmeric (Curcuma longa extract), is very well known for its multiple biological therapeutic activities, particularly its anti-inflammatory and antioxidant potential. However, due to its low water solubility, it exhibits poor bioavailability. In order to overcome this problem, in the current study, we have employed liposomal technology to encapsulate curcumin with the aim of enhancing its therapeutic efficacy. The curcumin-loaded liposomes (PlexoZome®) were tested on a cigarette smoke extract-induced Chronic Obstructive Pulmonary Disease (COPD) in vitro model using minimally immortalized human bronchial epithelial cells (BCiNS1.1). The anti-senescence and anti-inflammatory properties of PlexoZome® were explored. 5 µM PlexoZome® curcumin demonstrated anti-senescent activity by decrease in X-gal positive cells, and reduction in the expression of p16 and p21 in immunofluorescence staining. Moreover, PlexoZome® curcumin also demonstrated a reduction in proteins related to senescence (osteopontin, FGF basic and uPAR) and inflammation (GM-CSF, EGF and ST2). Overall, the results clearly demonstrate the therapeutic potential of curcumin encapsulated liposomes in managing CSE induced COPD, providing a new direction to respiratory clinics.


Sujet(s)
Vieillissement de la cellule , Curcumine , Cellules épithéliales , Inflammation , Liposomes , Curcumine/pharmacologie , Humains , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Inflammation/traitement médicamenteux , Inflammation/anatomopathologie , Inflammation/métabolisme , Bronches/anatomopathologie , Bronches/effets des médicaments et des substances chimiques , Bronches/métabolisme , Broncho-pneumopathie chronique obstructive/traitement médicamenteux , Broncho-pneumopathie chronique obstructive/anatomopathologie , Broncho-pneumopathie chronique obstructive/métabolisme , Fumée/effets indésirables , Anti-inflammatoires/pharmacologie , Lignée cellulaire
12.
JCI Insight ; 9(15)2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38916962

RÉSUMÉ

The number of adults living with cystic fibrosis (CF) has already increased significantly because of drastic improvements in life expectancy attributable to advances in treatment, including the development of highly effective modulator therapy. Chronic airway inflammation in CF contributes to morbidity and mortality, and aging processes like inflammaging and cell senescence influence CF pathology. Our results show that single-cell RNA sequencing data, human primary bronchial epithelial cells from non-CF and CF donors, a CF bronchial epithelial cell line, and Cftr-knockout (Cftr-/-) rats all demonstrated increased cell senescence markers in the CF bronchial epithelium. This was associated with upregulation of fibroblast growth factor receptors (FGFRs) and mitogen-activated protein kinase (MAPK) p38. Inhibition of FGFRs, specifically FGFR4 and to some extent FGFR1, attenuated cell senescence and improved mucociliary clearance, which was associated with MAPK p38 signaling. Mucociliary dysfunction could also be improved using a combination of senolytics in a CF ex vivo model. In summary, FGFR/MAPK p38 signaling contributes to cell senescence in CF airways, which is associated with impaired mucociliary clearance. Therefore, attenuation of cell senescence in the CF airways might be a future therapeutic strategy improving mucociliary dysfunction and lung disease in an aging population with CF.


Sujet(s)
Vieillissement de la cellule , Mucoviscidose , Muqueuse respiratoire , Mucoviscidose/métabolisme , Mucoviscidose/anatomopathologie , Mucoviscidose/génétique , Mucoviscidose/traitement médicamenteux , Humains , Animaux , Rats , Muqueuse respiratoire/métabolisme , Muqueuse respiratoire/anatomopathologie , Récepteur facteur croissance fibroblaste/métabolisme , Récepteur facteur croissance fibroblaste/génétique , Cellules épithéliales/métabolisme , p38 Mitogen-Activated Protein Kinases/métabolisme , Protéine CFTR/métabolisme , Protéine CFTR/génétique , Mâle , Modèles animaux de maladie humaine , Lignée cellulaire , Bronches/anatomopathologie , Bronches/métabolisme , Transduction du signal , Femelle
13.
Environ Int ; 190: 108829, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38908277

RÉSUMÉ

Exposure to environmental carcinogens is a significant contributor to cancer development, with genetic and epigenetic alterations playing pivotal roles in the carcinogenic process. However, the interplay between epigenetic regulation and genetic changes in carcinogenesis has yet to receive comprehensive attention. This study investigates the impact of continuous exposure to the tobacco-specific carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) on bronchial epithelial cells, leading to malignant transformation. Our findings reveal the down-regulation of the tumor suppressor-like circular RNA circNIPBL during oncogenic processes concomitant with the accumulation of the TP53-H179R, a single nucleotide variant. Diminished circNIPBL expression enhances the proliferative, distant metastatic, and tumor-forming capabilities of NNK-induced cancerous cells and lung cancer cell lines (A549, H1299), while also promoting the accumulation of TP53-H179R during NNK-induced carcinogenesis. Mechanistic investigations demonstrate that circNIPBL interacts with HSP90α to regulate the translocation of AHR into the nucleus, which may be a potential regulatory mechanism for NNK-induced carcinogenesis and TP53-H179R accumulation. This study introduces a novel perspective on the interplay between genetic alterations and epigenetic regulation in chemical carcinogenesis, which provides novel insight into the etiology of cancer.


Sujet(s)
Bronches , Carcinogenèse , Nitrosamines , ARN circulaire , Protéine p53 suppresseur de tumeur , ARN circulaire/génétique , Nitrosamines/toxicité , Humains , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Carcinogenèse/génétique , Carcinogenèse/induit chimiquement , Bronches/anatomopathologie , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/anatomopathologie , Mutation , Cancérogènes/toxicité , Tumeurs du poumon/induit chimiquement , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Lignée cellulaire tumorale , Animaux
14.
Mol Med ; 30(1): 93, 2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38898476

RÉSUMÉ

BACKGROUND: The epithelial-mesenchymal transition (EMT) of human bronchial epithelial cells (HBECs) is essential for airway remodeling during asthma. Wnt5a has been implicated in various lung diseases, while its role in the EMT of HBECs during asthma is yet to be determined. This study sought to define whether Wnt5a initiated EMT, leading to airway remodeling through the induction of autophagy in HBECs. METHODS: Microarray analysis was used to investigate the expression change of WNT5A in asthma patients. In parallel, EMT models were induced using 16HBE cells by exposing them to house dust mites (HDM) or interleukin-4 (IL-4), and then the expression of Wnt5a was observed. Using in vitro gain- and loss-of-function approaches via Wnt5a mimic peptide FOXY5 and Wnt5a inhibitor BOX5, the alterations in the expression of the epithelial marker E-cadherin and the mesenchymal marker protein were observed. Mechanistically, the Ca2+/CaMKII signaling pathway and autophagy were evaluated. An autophagy inhibitor 3-MA was used to examine Wnt5a in the regulation of autophagy during EMT. Furthermore, we used a CaMKII inhibitor KN-93 to determine whether Wnt5a induced autophagy overactivation and EMT via the Ca2+/CaMKII signaling pathway. RESULTS: Asthma patients exhibited a significant increase in the gene expression of WNT5A compared to the healthy control. Upon HDM and IL-4 treatments, we observed that Wnt5a gene and protein expression levels were significantly increased in 16HBE cells. Interestingly, Wnt5a mimic peptide FOXY5 significantly inhibited E-cadherin and upregulated α-SMA, Collagen I, and autophagy marker proteins (Beclin1 and LC3-II). Rhodamine-phalloidin staining showed that FOXY5 resulted in a rearrangement of the cytoskeleton and an increase in the quantity of stress fibers in 16HBE cells. Importantly, blocking Wnt5a with BOX5 significantly inhibited autophagy and EMT induced by IL-4 in 16HBE cells. Mechanistically, autophagy inhibitor 3-MA and CaMKII inhibitor KN-93 reduced the EMT of 16HBE cells caused by FOXY5, as well as the increase in stress fibers, cell adhesion, and autophagy. CONCLUSION: This study illustrates a new link in the Wnt5a-Ca2+/CaMKII-autophagy axis to triggering airway remodeling. Our findings may provide novel strategies for the treatment of EMT-related diseases.


Sujet(s)
Asthme , Autophagie , Cellules épithéliales , Transition épithélio-mésenchymateuse , Protéine Wnt-5a , Humains , Protéine Wnt-5a/métabolisme , Protéine Wnt-5a/génétique , Asthme/métabolisme , Asthme/anatomopathologie , Asthme/génétique , Cellules épithéliales/métabolisme , Calcium-Calmodulin-Dependent Protein Kinase Type 2/métabolisme , Bronches/métabolisme , Bronches/anatomopathologie , Mâle , Lignée cellulaire , Femelle , Adulte d'âge moyen , Transduction du signal , Adulte
15.
Int J Mol Sci ; 25(11)2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38892373

RÉSUMÉ

SARS-CoV-2 infection has been recently shown to induce cellular senescence in vivo. A senescence-like phenotype has been reported in cystic fibrosis (CF) cellular models. Since the previously published data highlighted a low impact of SARS-CoV-2 on CFTR-defective cells, here we aimed to investigate the senescence hallmarks in SARS-CoV-2 infection in the context of a loss of CFTR expression/function. We infected WT and CFTR KO 16HBE14o-cells with SARS-CoV-2 and analyzed both the p21 and Ki67 expression using immunohistochemistry and viral and p21 gene expression using real-time PCR. Prior to SARS-CoV-2 infection, CFTR KO cells displayed a higher p21 and lower Ki67 expression than WT cells. We detected lipid accumulation in CFTR KO cells, identified as lipolysosomes and residual bodies at the subcellular/ultrastructure level. After SARS-CoV-2 infection, the situation reversed, with low p21 and high Ki67 expression, as well as reduced viral gene expression in CFTR KO cells. Thus, the activation of cellular senescence pathways in CFTR-defective cells was reversed by SARS-CoV-2 infection while they were activated in CFTR WT cells. These data uncover a different response of CF and non-CF bronchial epithelial cell models to SARS-CoV-2 infection and contribute to uncovering the molecular mechanisms behind the reduced clinical impact of COVID-19 in CF patients.


Sujet(s)
Bronches , COVID-19 , Vieillissement de la cellule , Inhibiteur p21 de kinase cycline-dépendante , Protéine CFTR , Cellules épithéliales , Antigène KI-67 , SARS-CoV-2 , Protéine CFTR/génétique , Protéine CFTR/métabolisme , Humains , Vieillissement de la cellule/génétique , SARS-CoV-2/physiologie , COVID-19/virologie , COVID-19/métabolisme , COVID-19/anatomopathologie , Cellules épithéliales/métabolisme , Cellules épithéliales/virologie , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , Inhibiteur p21 de kinase cycline-dépendante/génétique , Antigène KI-67/métabolisme , Bronches/virologie , Bronches/métabolisme , Bronches/anatomopathologie , Bronches/cytologie , Mucoviscidose/métabolisme , Mucoviscidose/génétique , Mucoviscidose/virologie , Mucoviscidose/anatomopathologie , Lignée cellulaire
16.
Khirurgiia (Mosk) ; (6): 36-44, 2024.
Article de Russe | MEDLINE | ID: mdl-38888017

RÉSUMÉ

OBJECTIVE: To evaluate the efficacy and safety of tranbronchial cryobiopsy (TBCB) with 1.9-mm and 1.1-mm cryoprobes in patients with peripheral pulmonary lesions (PPLs). MATERIAL AND METHODS: We analyzed 34 patients (mean age 60 years) with PPLs who underwent bronchoscopy with TBCB. Mean lesion size was 31.5 mm, upper lobe localization was predominant (47% of cases). CT signs of appropriate bronchus were identified in 79% (27/34) of cases. Manual branch tracking and virtual bronchoscopy (VB) were performed pre-procedurally, and radial endobronchial ultrasonography (rEBUS) was performed during bronchoscopy for accurate positioning of PPLs. TBCB was performed using 1.9-mm (n=19) or 1.1-mm (n=15) cryoprobes without fluoroscopic guidance. Incidence and severity of bleeding and pneumothorax were evaluated in all patients. RESULTS: Total efficacy of TBCB was 76.5% (26/34): 78.9% (15/19) for 1.9-mm cryoprobe and 73.3% (11/15) for 1.1-mm cryoprobe (p=0.702). Efficacy depended on the presence of CT signs of bronchus (presence - 94%, absence 14.3%, p<0.001) and PPL size (94% for PPL >30 mm and 58.8% for PPL <30 mm, p=0.016). Central probe position during rEBUS was associated with 94.7% diagnostic efficacy (18/19), adjacent probe position - 72.7% (8/11) (p=0.088). Bleeding grade 3 (Nasville) occurred in 5.8% (2/34) of cases, and no pneumothorax was observed. CONCLUSION: TBCB is an effective and safe diagnostic method for PPLs.


Sujet(s)
Bronchoscopie , Cryochirurgie , Humains , Bronchoscopie/méthodes , Bronchoscopie/effets indésirables , Adulte d'âge moyen , Mâle , Femelle , Cryochirurgie/méthodes , Diagnostic différentiel , Sujet âgé , Bronches/anatomopathologie , Bronches/imagerie diagnostique , Poumon/anatomopathologie , Poumon/imagerie diagnostique , Évaluation des résultats et des processus en soins de santé , Tomodensitométrie/méthodes , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/diagnostic
17.
Sci Rep ; 14(1): 13145, 2024 06 07.
Article de Anglais | MEDLINE | ID: mdl-38849430

RÉSUMÉ

Airway remodelling in lung diseases can be treated by inhibiting excessive smooth muscle cell proliferation. Zedoarondiol (Zed) is a natural compound isolated from the Chinese herb Curcuma longa. The caveolin-1 (CAV-1) is widely expressed in lung cells and plays a key role in platelet-derived growth factor (PDGF) signalling and cell proliferation. This study aims to investigate the effect of Zed on human bronchial smooth muscle cell (HBSMC) proliferation and explore its potential molecular mechanisms. We assessed the effect of Zed on the proliferation of PDGF-stimulated HBSMCs and performed proteomic analysis to identify potential molecular targets and pathways. CAV1 siRNA was used to validate our findings in vitro. In PDGF-stimulated HBSMCs, Zed significantly inhibited excessive proliferation of HBSMCs. Proteomic analysis of zedoarondiol-treated HBSMCs revealed significant enrichment of differentially expressed proteins in cell proliferation-related pathways and biological processes. Zed inhibition of HBSMC proliferation was associated with upregulation of CAV1, regulation of the CAV-1/PDGF pathway and inhibition of MAPK and PI3K/AKT signalling pathway activation. Treatment of HBSMCs with CAV1 siRNA partly reversed the inhibitory effect of Zed on HBSMC proliferation. Thus, this study reveals that zedoarondiol potently inhibits HBSMC proliferation by upregulating CAV-1 expression, highlighting its potential value in airway remodelling and related diseases.


Sujet(s)
Bronches , Cavéoline-1 , Prolifération cellulaire , Myocytes du muscle lisse , Facteur de croissance dérivé des plaquettes , Transduction du signal , Humains , Cavéoline-1/métabolisme , Cavéoline-1/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Bronches/métabolisme , Bronches/cytologie , Bronches/anatomopathologie , Facteur de croissance dérivé des plaquettes/métabolisme , Protéomique/méthodes , Phosphatidylinositol 3-kinases/métabolisme , Cellules cultivées
18.
Iran J Allergy Asthma Immunol ; 23(2): 211-219, 2024 Apr 07.
Article de Anglais | MEDLINE | ID: mdl-38822515

RÉSUMÉ

Asthma is a chronic respiratory disease that is characterized by airway inflammation, excessive mucus production, and airway remodeling. Prevention and treatment for asthma is an urgent issue in clinical studies. In recent years, N6-methyladenosine methylation (m6A) has emerged as a promising regulatory approach involved in multiple diseases. ALKBH5 (alkB homolog 5) is a demethylase widely studied in disease pathologies. This work aimed to explore the regulatory mechanisms underlying the ALKBH5-regulated asthma. We established an interleukin-13 (IL-13)-stimulated cell model to mimic the in vitro inflammatory environment of asthma. ALKBH5 knockdown in bronchial epithelial cells was performed using siRNAs, and the knockdown efficacy was analyzed by quantitative PCR (qPCR). Cell viability and proliferation were measured by cell counting kit 8 (CCK-8) and colony formation assay. The ferroptosis was assessed by measuring the total iron, Fe2+, lipid reactive oxygen species (ROS), malondialdehyde (MDA), and superoxide dismutase (SOD) levels. The enrichment of N6-methyladenosine methylation (m6A) modification was detected by the MeRIP assay. Knockdown of ALKBH5 significantly elevated the survival and colony formation ability of bronchial epithelial cells in the IL-13 induction model. The levels of total iron, Fe2+, lipid ROS, and MDA were remarkedly elevated, and the SOD level was reduced in IL-13-induced bronchial epithelial cells, and depletion of ALKBH5 reversed these effects. Knockdown of ALKBH5 elevated the enrichment of m6A modification and expression of glutathione peroxidase 4 (GPX4). Knockdown of GPX4 abolished the pro-proliferation and anti-ferroptosis effects of siALKBH5. Knockdown of ALKBH5 improved the proliferation of bronchial epithelial cells and alleviated cell ferroptosis.


Sujet(s)
Adénosine , AlkB Homolog 5, RNA demethylase , Asthme , AlkB Homolog 5, RNA demethylase/métabolisme , AlkB Homolog 5, RNA demethylase/génétique , Asthme/génétique , Asthme/métabolisme , Asthme/anatomopathologie , Humains , Adénosine/analogues et dérivés , Adénosine/métabolisme , Prolifération cellulaire/génétique , Méthylation , Évolution de la maladie , Lignée cellulaire , Ferroptose/génétique , Cellules épithéliales/métabolisme , Régulation négative , Bronches/anatomopathologie , Bronches/métabolisme , Techniques de knock-down de gènes , Survie cellulaire/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE