Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.905
Filtrer
1.
Front Immunol ; 15: 1346491, 2024.
Article de Anglais | MEDLINE | ID: mdl-38911863

RÉSUMÉ

Introduction: Exacerbations of chronic obstructive pulmonary disease (COPD) increase mortality risk and can lead to accelerated loss of lung function. The increased inflammatory response during exacerbations contributes to worsening of airflow limitation, but whether it also impacts epithelial repair is unclear. Therefore, we studied the effect of the soluble factor micro-environment during COPD exacerbations on epithelial repair using an exacerbation cocktail (EC), composed of four factors that are increased in COPD lungs during exacerbations (IL-1ß, IL-6, IL-8, TNF-α). Methods: Mouse organoids (primary CD31-CD45-Epcam+ cells co-cultured with CCL206 fibroblasts) were used to study epithelial progenitor behavior. Mature epithelial cell responses were evaluated using mouse precision cut lung slices (PCLS). The expression of epithelial supportive factors was assessed in CCL206 fibroblasts and primary human lung fibroblasts. Results: EC exposure increased the number and size of organoids formed, and upregulated Lamp3, Muc5ac and Muc5b expression in day 14 organoids. In PCLS, EC imparted no effect on epithelial marker expression. Pre-treatment of CCL206 fibroblasts with EC was sufficient to increase organoid formation. Additionally, the expression of Il33, Tgfa and Areg was increased in CCL206 fibroblasts from EC treated organoids, but these factors individually did not affect organoid formation or size. However, TGF-α downregulated Foxj1 expression and upregulated Aqp5 expression in day 14 organoids. Conclusions: EC exposure stimulates organoid formation and growth, but it alters epithelial differentiation. EC changes the epithelial progenitor support function of fibroblasts which contributes to observed effects on epithelial progenitors.


Sujet(s)
Cellules épithéliales , Fibroblastes , Organoïdes , Broncho-pneumopathie chronique obstructive , Animaux , Broncho-pneumopathie chronique obstructive/métabolisme , Broncho-pneumopathie chronique obstructive/anatomopathologie , Broncho-pneumopathie chronique obstructive/immunologie , Humains , Souris , Fibroblastes/métabolisme , Cellules épithéliales/métabolisme , Cytokines/métabolisme , Poumon/anatomopathologie , Poumon/immunologie , Poumon/métabolisme , Cellules cultivées , Évolution de la maladie , Muqueuse respiratoire/métabolisme , Muqueuse respiratoire/immunologie , Muqueuse respiratoire/anatomopathologie , Souris de lignée C57BL
3.
Nature ; 629(8013): 869-877, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38693267

RÉSUMÉ

Airway hillocks are stratified epithelial structures of unknown function1. Hillocks persist for months and have a unique population of basal stem cells that express genes associated with barrier function and cell adhesion. Hillock basal stem cells continually replenish overlying squamous barrier cells. They exhibit dramatically higher turnover than the abundant, largely quiescent classic pseudostratified airway epithelium. Hillocks resist a remarkably broad spectrum of injuries, including toxins, infection, acid and physical injury because hillock squamous cells shield underlying hillock basal stem cells from injury. Hillock basal stem cells are capable of massive clonal expansion that is sufficient to resurface denuded airway, and eventually regenerate normal airway epithelium with each of its six component cell types. Hillock basal stem cells preferentially stratify and keratinize in the setting of retinoic acid signalling inhibition, a known cause of squamous metaplasia2,3. Here we show that mouse hillock expansion is the cause of vitamin A deficiency-induced squamous metaplasia. Finally, we identify human hillocks whose basal stem cells generate functional squamous barrier structures in culture. The existence of hillocks reframes our understanding of airway epithelial regeneration. Furthermore, we show that hillocks are one origin of 'squamous metaplasia', which is long thought to be a precursor of lung cancer.


Sujet(s)
Plasticité cellulaire , Cellules épithéliales , Régénération , Muqueuse respiratoire , Cellules souches , Animaux , Femelle , Humains , Mâle , Souris , Cellules épithéliales/cytologie , Cellules épithéliales/anatomopathologie , Métaplasie/étiologie , Métaplasie/anatomopathologie , Muqueuse respiratoire/cytologie , Muqueuse respiratoire/traumatismes , Muqueuse respiratoire/anatomopathologie , Cellules souches/cytologie , Trétinoïne/métabolisme , Trétinoïne/pharmacologie , Rétinol/métabolisme , Rétinol/pharmacologie , Tumeurs du poumon/étiologie , Tumeurs du poumon/anatomopathologie , Souris de lignée C57BL
4.
Front Immunol ; 15: 1362404, 2024.
Article de Anglais | MEDLINE | ID: mdl-38745671

RÉSUMÉ

Introduction: The anti-inflammatory effect of green tea extract (GTE) has been confirmed in asthmatic mice, however, the pharmacological mechanism is not fully elucidated. Methods: To investigate the therapeutic efficacy of GTE in asthma and identify specific pathways, murine model of allergic asthma was established by ovalbumin (OVA) sensitization and the challenge for 4 weeks, with oral treatment using GTE and dexamethasone (DEX). Inflammatory cell counts, cytokines, OVA-specific IgE, airway hyperreactivity, and antioxidant markers in the lung were evaluated. Also, pulmonary histopathological analysis and western blotting were performed. In vitro, we established the model by stimulating the human airway epithelial cell line NCI-H292 using lipopolysaccharide, and treating with GTE and mitogen-activated protein kinases (MAPKs) inhibitors. Results: The GTE100 and GTE400 groups showed a decrease in airway hyperresponsiveness and the number of inflammatory cells in the bronchoalveolar lavage fluid (BALF) compared to the OVA group. GTE treatment also reduced interleukin (IL)-13, IL-5, and IL-4 levels in the BALF, and OVA-specific immunoglobulin E levels in the serum compared to those in the OVA group. GTE treatment decreased OVA-induced mucus secretion and airway inflammation. In addition, GTE suppressed the oxidative stress, and phosphorylation of MAPKs, which generally occurs after exposure to OVA. GTE administration also reduced matrix metalloproteinase-9 activity and protein levels. Conclusion: GTE effectively inhibited asthmatic respiratory inflammation and mucus hyperproduction induced by OVA inhalation. These results suggest that GTE has the potential to be used for the treatment of asthma.


Sujet(s)
Asthme , Cellules épithéliales , Matrix metalloproteinase 9 , Stress oxydatif , Extraits de plantes , Asthme/traitement médicamenteux , Asthme/immunologie , Asthme/métabolisme , Animaux , Stress oxydatif/effets des médicaments et des substances chimiques , Souris , Humains , Extraits de plantes/pharmacologie , Matrix metalloproteinase 9/métabolisme , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Thé/composition chimique , Femelle , Transduction du signal/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Mitogen-Activated Protein Kinases/métabolisme , Muqueuse respiratoire/métabolisme , Muqueuse respiratoire/effets des médicaments et des substances chimiques , Muqueuse respiratoire/immunologie , Muqueuse respiratoire/anatomopathologie , Cytokines/métabolisme , Ovalbumine/immunologie , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique
5.
Am J Physiol Lung Cell Mol Physiol ; 326(6): L754-L769, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38625125

RÉSUMÉ

Chronic exposure to environmental hazards causes airway epithelial dysfunction, primarily impaired physical barriers, immune dysfunction, and repair or regeneration. Impairment of airway epithelial function subsequently leads to exaggerated airway inflammation and remodeling, the main features of chronic obstructive pulmonary disease (COPD). Mitochondrial damage has been identified as one of the mechanisms of airway abnormalities in COPD, which is closely related to airway inflammation and airflow limitation. In this review, we evaluate updated evidence for airway epithelial mitochondrial damage in COPD and focus on the role of mitochondrial damage in airway epithelial dysfunction. In addition, the possible mechanism of airway epithelial dysfunction mediated by mitochondrial damage is discussed in detail, and recent strategies related to airway epithelial-targeted mitochondrial therapy are summarized. Results have shown that dysregulation of mitochondrial quality and oxidative stress may lead to airway epithelial dysfunction in COPD. This may result from mitochondrial damage as a central organelle mediating abnormalities in cellular metabolism. Mitochondrial damage mediates procellular senescence effects due to mitochondrial reactive oxygen species, which effectively exacerbate different types of programmed cell death, participate in lipid metabolism abnormalities, and ultimately promote airway epithelial dysfunction and trigger COPD airway abnormalities. These can be prevented by targeting mitochondrial damage factors and mitochondrial transfer. Thus, because mitochondrial damage is involved in COPD progression as a central factor of homeostatic imbalance in airway epithelial cells, it may be a novel target for therapeutic intervention to restore airway epithelial integrity and function in COPD.


Sujet(s)
Mitochondries , Stress oxydatif , Broncho-pneumopathie chronique obstructive , Humains , Broncho-pneumopathie chronique obstructive/anatomopathologie , Broncho-pneumopathie chronique obstructive/métabolisme , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Animaux , Muqueuse respiratoire/anatomopathologie , Muqueuse respiratoire/métabolisme , Cellules épithéliales/anatomopathologie , Cellules épithéliales/métabolisme , Espèces réactives de l'oxygène/métabolisme
7.
Am J Respir Cell Mol Biol ; 71(1): 81-94, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38531016

RÉSUMÉ

Epithelial polarity is fundamental in maintaining barrier integrity and tissue protection. In cystic fibrosis (CF), apicobasal polarity of the airway epithelium is altered, resulting in increased apical fibronectin deposition and enhanced susceptibility to bacterial infections. Here, we evaluated the effect of highly effective modulator treatment (HEMT) on fibronectin apical deposition and investigated the intracellular mechanisms triggering the defect in polarity of the CF airway epithelium. To this end, primary cultures of CF (F508del variant) human airway epithelial cells (HAECs) and a HAEC line, Calu-3, knocked down for CFTR (CF transmembrane conductance regulator) were compared with control counterparts. We show that CFTR mutation in primary HAECs and CFTR knockdown cells promote the overexpression and oversecretion of TGF-ß1 and DKK1 when cultured at an air-liquid interface. These dynamic changes result in hyperactivation of the TGF-ß pathway and inhibition of the Wnt pathway through degradation of ß-catenin leading to imbalanced proliferation and polarization. The abnormal interplay between TGF-ß and Wnt signaling pathways is reinforced by aberrant Akt signaling. Pharmacological manipulation of TGF-ß, Wnt, and Akt pathways restored polarization of the F508del CF epithelium, a correction that was not achieved by HEMT. Our data shed new insights into the signaling pathways that fine-tune apicobasal polarization in primary airway epithelial cells and may provide an explanation to the mitigated efficacy of HEMT on lung infection in people with CF.


Sujet(s)
Polarité de la cellule , Protéine CFTR , Mucoviscidose , Cellules épithéliales , Protéines et peptides de signalisation intercellulaire , Protéines proto-oncogènes c-akt , Muqueuse respiratoire , Voie de signalisation Wnt , Humains , Mucoviscidose/métabolisme , Mucoviscidose/anatomopathologie , Mucoviscidose/génétique , Protéine CFTR/métabolisme , Protéine CFTR/génétique , Polarité de la cellule/effets des médicaments et des substances chimiques , Protéines et peptides de signalisation intercellulaire/métabolisme , Protéines et peptides de signalisation intercellulaire/génétique , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Protéines proto-oncogènes c-akt/métabolisme , Muqueuse respiratoire/métabolisme , Muqueuse respiratoire/anatomopathologie , bêta-Caténine/métabolisme , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta-1/métabolisme , Cellules cultivées , Lignée cellulaire , Fibronectines/métabolisme
8.
Am J Pathol ; 194(7): 1185-1196, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38548270

RÉSUMÉ

Acute lung injury (ALI) is a devastating clinical syndrome caused by different factors, with high morbidity and mortality. Lung injury and inflammation caused by lipopolysaccharide (LPS) can be modulated by NLRP3 inflammasome activation, yet its exact function within the airway epithelium is still unknown. Meanwhile, glucose transporter protein 1 (GLUT1) contributes to a number of inflammatory illnesses, including ALI. The present study aimed to assess GLUT1's function in NLRP3 inflammasome activation of airway epithelium in LPS-induced acute lung injury. BALB/c mice and BEAS-2B cells were exposed to LPS (5 mg/kg and 200 µg/mL, respectively), with or without GLUT1 antagonists (WZB117 or BAY876). LPS up-regulated pulmonary expression of NLRP3 and GLUT1 in mice, which could be blocked by WZB117 or BAY876. Pharmacological inhibition of GLUT1 in vivo significantly attenuated lung tissue damage, neutrophil accumulation, and proinflammatory factors release (TNF-α, IL-6, and IL-1ß) in LPS-exposed mice. Meanwhile, the activation markers of NLRP3 inflammasome (ASC, caspase-1, IL-1ß, and IL-18) induced by LPS were also suppressed. In cultured BEAS-2B cells, LPS induced an increase in GLUT1 expression and triggered activation of the NLRP3 inflammasome, both of which were inhibited by GLUT1 antagonists. These results illustrate that GLUT1 participates in LPS-induced ALI and promotes the activation of the NLRP3 inflammasome in airway epithelial cells.


Sujet(s)
Lésion pulmonaire aigüe , Transporteur de glucose de type 1 , Inflammasomes , Lipopolysaccharides , Souris de lignée BALB C , Protéine-3 de la famille des NLR contenant un domaine pyrine , Animaux , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Lésion pulmonaire aigüe/métabolisme , Lésion pulmonaire aigüe/anatomopathologie , Lésion pulmonaire aigüe/induit chimiquement , Inflammasomes/métabolisme , Souris , Transporteur de glucose de type 1/métabolisme , Humains , Mâle , Muqueuse respiratoire/métabolisme , Muqueuse respiratoire/anatomopathologie
9.
Ophthalmic Plast Reconstr Surg ; 40(4): e133-e138, 2024.
Article de Anglais | MEDLINE | ID: mdl-38427820

RÉSUMÉ

A primary orbital respiratory cyst is a congenital choristoma that presents in the orbit and with different signs and symptoms depending on the location, which might also change the surgical approach. The aim of this report is to describe 2 new cases of primary respiratory epithelial cysts and to review the literature on presentation, management, and risk factors with different surgical approaches and complications. Two cases presenting with gradually increased proptosis had a confirmed diagnosis of a respiratory epithelial cyst. CT and MRI imaging revealed a thin-walled intraconal cystic lesion. Complete surgical removal was not possible and an incisional biopsy of the wall was performed. Rarely, orbital respiratory epithelium cysts are primary lesions. The high risk of breaking the capsule during surgery and the presence of residual epithelial cells within the orbit may cause cyst's recurrence. Indeed, we recommend careful attention during dissection maneuvers and a long-term follow-up.


Sujet(s)
Kystes , Imagerie par résonance magnétique , Maladies de l'orbite , Tomodensitométrie , Humains , Kystes/diagnostic , Kystes/chirurgie , Maladies de l'orbite/diagnostic , Maladies de l'orbite/chirurgie , Mâle , Femelle , Muqueuse respiratoire/anatomopathologie , Choristome/diagnostic , Choristome/chirurgie
10.
Am J Physiol Lung Cell Mol Physiol ; 326(5): L618-L626, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38469627

RÉSUMÉ

Thymic stromal lymphopoietin (TSLP) is an epithelial-derived pleiotropic cytokine that regulates T-helper 2 (Th2) immune responses in the lung and plays a major role in severe uncontrolled asthma. Emerging evidence suggests a role for endoplasmic reticulum (ER) stress in the pathogenesis of asthma. In this study, we determined if ER stress and the unfolded protein response (UPR) signaling are involved in TSLP induction in the airway epithelium. For this, we treated human bronchial epithelial basal cells and differentiated primary bronchial epithelial cells with ER stress inducers and the TSLP mRNA and protein expression was determined. A series of siRNA gene knockdown experiments were conducted to determine the ER stress-induced TSLP signaling pathways. cDNA collected from asthmatic bronchial biopsies was used to determine the gene correlation between ER stress and TSLP. Our results show that ER stress signaling induces TSLP mRNA expression via the PERK-C/EBP homologous protein (CHOP) signaling pathway. AP-1 transcription factor is important in regulating this ER stress-induced TSLP mRNA induction, though ER stress alone cannot induce TSLP protein production. However, ER stress significantly enhances TLR3-induced TSLP protein secretion in the airway epithelium. TSLP and ER stress (PERK) mRNA expression positively correlates in bronchial biopsies from participants with asthma, particularly in neutrophilic asthma. In conclusion, these results suggest that ER stress primes TSLP that is then enhanced further upon TLR3 activation, which may induce severe asthma exacerbations. Targeting ER stress using pharmacological interventions may provide novel therapeutics for severe uncontrolled asthma.NEW & NOTEWORTHY TSLP is an epithelial-derived cytokine and a key regulator in the pathogenesis of severe uncontrolled asthma. We demonstrate a novel mechanism by which endoplasmic reticulum stress signaling upregulates airway epithelial TSLP mRNA expression via the PERK-CHOP signaling pathway and enhances TLR3-mediated TSLP protein secretion.


Sujet(s)
Asthme , Cytokines , Stress du réticulum endoplasmique , Cellules épithéliales , Lymphopoïétine stromale thymique , Récepteur de type Toll-3 , Réponse aux protéines mal repliées , Humains , Cytokines/métabolisme , Récepteur de type Toll-3/métabolisme , Récepteur de type Toll-3/génétique , Asthme/métabolisme , Asthme/anatomopathologie , Asthme/génétique , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Facteur de transcription CHOP/métabolisme , Facteur de transcription CHOP/génétique , Transduction du signal , Muqueuse respiratoire/métabolisme , Muqueuse respiratoire/anatomopathologie , Bronches/métabolisme , Bronches/anatomopathologie , eIF-2 Kinase/métabolisme , eIF-2 Kinase/génétique , Cellules cultivées , Femelle , ARN messager/génétique , ARN messager/métabolisme
11.
Biomaterials ; 308: 122546, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38552367

RÉSUMÉ

Patients with cystic fibrosis (CF) experience severe lung disease, including persistent infections, inflammation, and irreversible fibrotic remodeling of the airways. Although therapy with transmembrane conductance regulator (CFTR) protein modulators reached optimal results in terms of CFTR rescue, lung transplant remains the best line of care for patients in an advanced stage of CF. Indeed, chronic inflammation and tissue remodeling still represent stumbling blocks during treatment, and underlying mechanisms are still unclear. Nowadays, animal models are not able to fully replicate clinical features of the human disease and the conventional in vitro models lack a stromal compartment undergoing fibrotic remodeling. To address this gap, we show the development of a 3D full-thickness model of CF with a human bronchial epithelium differentiated on a connective airway tissue. We demonstrated that the epithelial cells not only underwent mucociliary differentiation but also migrated in the connective tissue and formed gland-like structures. The presence of the connective tissue stimulated the pro-inflammatory behaviour of the epithelium, which activated the fibroblasts embedded into their own extracellular matrix (ECM). By varying the composition of the model with CF epithelial cells and a CF or healthy connective tissue, it was possible to replicate different moments of CF disease, as demonstrated by the differences in the transcriptome of the CF epithelium in the different conditions. The possibility to faithfully represent the crosstalk between epithelial and connective in CF through the full thickness model, along with inflammation and stromal activation, makes the model suitable to better understand mechanisms of disease genesis, progression, and response to therapy.


Sujet(s)
Tissu conjonctif , Mucoviscidose , Cellules épithéliales , Humains , Mucoviscidose/anatomopathologie , Mucoviscidose/métabolisme , Tissu conjonctif/anatomopathologie , Tissu conjonctif/métabolisme , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Muqueuse respiratoire/métabolisme , Muqueuse respiratoire/anatomopathologie , Matrice extracellulaire/métabolisme , Différenciation cellulaire , Modèles biologiques , Fibroblastes/métabolisme
12.
Am J Respir Cell Mol Biol ; 70(5): 379-391, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38301257

RÉSUMÉ

GDF15 (growth differentiation factor 15) is a stress cytokine with several proposed roles, including support of stress erythropoiesis. Higher circulating GDF15 levels are prognostic of mortality during acute respiratory distress syndrome, but the cellular sources and downstream effects of GDF15 during pathogen-mediated lung injury are unclear. We quantified GDF15 in lower respiratory tract biospecimens and plasma from patients with acute respiratory failure. Publicly available data from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection were reanalyzed. We used mouse models of hemorrhagic acute lung injury mediated by Pseudomonas aeruginosa exoproducts in wild-type mice and mice genetically deficient for Gdf15 or its putative receptor, Gfral. In critically ill humans, plasma levels of GDF15 correlated with lower respiratory tract levels and were higher in nonsurvivors. SARS-CoV-2 infection induced GDF15 expression in human lung epithelium, and lower respiratory tract GDF15 levels were higher in coronavirus disease (COVID-19) nonsurvivors. In mice, intratracheal P. aeruginosa type II secretion system exoproducts were sufficient to induce airspace and plasma release of GDF15, which was attenuated with epithelial-specific deletion of Gdf15. Mice with global Gdf15 deficiency had decreased airspace hemorrhage, an attenuated cytokine profile, and an altered lung transcriptional profile during injury induced by P. aeruginosa type II secretion system exoproducts, which was not recapitulated in mice deficient for Gfral. Airspace GDF15 reconstitution did not significantly modulate key lung cytokine levels but increased circulating erythrocyte counts. Lung epithelium releases GDF15 during pathogen injury, which is associated with plasma levels in humans and mice and can increase erythrocyte counts in mice, suggesting a novel lung-blood communication pathway.


Sujet(s)
COVID-19 , Facteur-15 de croissance et de différenciation , Poumon , Pseudomonas aeruginosa , SARS-CoV-2 , Facteur-15 de croissance et de différenciation/génétique , Facteur-15 de croissance et de différenciation/métabolisme , Animaux , COVID-19/métabolisme , COVID-19/virologie , Humains , Souris , Poumon/métabolisme , Poumon/anatomopathologie , Poumon/virologie , Mâle , Infections à Pseudomonas/métabolisme , Lésion pulmonaire aigüe/anatomopathologie , Lésion pulmonaire aigüe/métabolisme , Femelle , Souris de lignée C57BL , Souris knockout , Muqueuse respiratoire/métabolisme , Muqueuse respiratoire/anatomopathologie , Modèles animaux de maladie humaine
13.
Am J Respir Crit Care Med ; 209(12): 1453-1462, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38324627

RÉSUMÉ

Rationale: Pseudomonas aeruginosa is the major bacterial pathogen colonizing the airways of adult patients with cystic fibrosis (CF) and causes chronic infections that persist despite antibiotic therapy. Intracellular bacteria may represent an unrecognized reservoir of bacteria that evade the immune system and antibiotic therapy. Although the ability of P. aeruginosa to invade and survive within epithelial cells has been described in vitro in different epithelial cell models, evidence of this intracellular lifestyle in human lung tissues is currently lacking. Objectives: To detect and characterize intracellular P. aeruginosa in CF airway epithelium from human lung explant tissues. Methods: We sampled lung explant tissues from patients with CF undergoing lung transplantation and non-CF lung donor control tissue. We analyzed lung tissue sections for the presence of intracellular P. aeruginosa using quantitative culture and microscopy, in parallel to histopathology and airway morphometry. Measurements and Main Results: P. aeruginosa was isolated from the lungs of seven patients with CF undergoing lung transplantation. Microscopic assessment revealed the presence of intracellular P. aeruginosa within airway epithelial cells in three of the seven patients analyzed at a varying but low frequency. We observed those events occurring in lung regions with high bacterial burden. Conclusions: This is the first study describing the presence of intracellular P. aeruginosa in CF lung tissues. Although intracellular P. aeruginosa in airway epithelial cells is likely relatively rare, our findings highlight the plausible occurrence of this intracellular bacterial reservoir in chronic CF infections.


Sujet(s)
Mucoviscidose , Transplantation pulmonaire , Poumon , Infections à Pseudomonas , Pseudomonas aeruginosa , Muqueuse respiratoire , Humains , Mucoviscidose/microbiologie , Mucoviscidose/complications , Femelle , Mâle , Adulte , Muqueuse respiratoire/microbiologie , Muqueuse respiratoire/anatomopathologie , Infections à Pseudomonas/microbiologie , Poumon/microbiologie , Poumon/anatomopathologie , Jeune adulte , Cellules épithéliales/microbiologie
14.
Thorax ; 79(6): 524-537, 2024 May 20.
Article de Anglais | MEDLINE | ID: mdl-38286613

RÉSUMÉ

INTRODUCTION: Environmental pollutants injure the mucociliary elevator, thereby provoking disease progression in chronic obstructive pulmonary disease (COPD). Epithelial resilience mechanisms to environmental nanoparticles in health and disease are poorly characterised. METHODS: We delineated the impact of prevalent pollutants such as carbon and zinc oxide nanoparticles, on cellular function and progeny in primary human bronchial epithelial cells (pHBECs) from end-stage COPD (COPD-IV, n=4), early disease (COPD-II, n=3) and pulmonary healthy individuals (n=4). After nanoparticle exposure of pHBECs at air-liquid interface, cell cultures were characterised by functional assays, transcriptome and protein analysis, complemented by single-cell analysis in serial samples of pHBEC cultures focusing on basal cell differentiation. RESULTS: COPD-IV was characterised by a prosecretory phenotype (twofold increase in MUC5AC+) at the expense of the multiciliated epithelium (threefold reduction in Ac-Tub+), resulting in an increased resilience towards particle-induced cell damage (fivefold reduction in transepithelial electrical resistance), as exemplified by environmentally abundant doses of zinc oxide nanoparticles. Exposure of COPD-II cultures to cigarette smoke extract provoked the COPD-IV characteristic, prosecretory phenotype. Time-resolved single-cell transcriptomics revealed an underlying COPD-IV unique basal cell state characterised by a twofold increase in KRT5+ (P=0.018) and LAMB3+ (P=0.050) expression, as well as a significant activation of Wnt-specific (P=0.014) and Notch-specific (P=0.021) genes, especially in precursors of suprabasal and secretory cells. CONCLUSION: We identified COPD stage-specific gene alterations in basal cells that affect the cellular composition of the bronchial elevator and may control disease-specific epithelial resilience mechanisms in response to environmental nanoparticles. The identified phenomena likely inform treatment and prevention strategies.


Sujet(s)
Cellules épithéliales , Broncho-pneumopathie chronique obstructive , Humains , Broncho-pneumopathie chronique obstructive/étiologie , Cellules épithéliales/métabolisme , Mâle , Adulte d'âge moyen , Cellules cultivées , Bronches/anatomopathologie , Femelle , Sujet âgé , Oxyde de zinc , Muqueuse respiratoire/métabolisme , Muqueuse respiratoire/anatomopathologie , Cils vibratiles , Nanoparticules , Différenciation cellulaire
15.
PLoS Comput Biol ; 19(8): e1011356, 2023 08.
Article de Anglais | MEDLINE | ID: mdl-37566610

RÉSUMÉ

Human airway epithelium (HAE) represents the primary site of viral infection for SARS-CoV-2. Comprising different cell populations, a lot of research has been aimed at deciphering the major cell types and infection dynamics that determine disease progression and severity. However, the cell type-specific replication kinetics, as well as the contribution of cellular composition of the respiratory epithelium to infection and pathology are still not fully understood. Although experimental advances, including Air-liquid interface (ALI) cultures of reconstituted pseudostratified HAE, as well as lung organoid systems, allow the observation of infection dynamics under physiological conditions in unprecedented level of detail, disentangling and quantifying the contribution of individual processes and cells to these dynamics remains challenging. Here, we present how a combination of experimental data and mathematical modelling can be used to infer and address the influence of cell type specific infectivity and tissue composition on SARS-CoV-2 infection dynamics. Using a stepwise approach that integrates various experimental data on HAE culture systems with regard to tissue differentiation and infection dynamics, we develop an individual cell-based model that enables investigation of infection and regeneration dynamics within pseudostratified HAE. In addition, we present a novel method to quantify tissue integrity based on image data related to the standard measures of transepithelial electrical resistance measurements. Our analysis provides a first aim of quantitatively assessing cell type specific infection kinetics and shows how tissue composition and changes in regeneration capacity, as e.g. in smokers, can influence disease progression and pathology. Furthermore, we identified key measurements that still need to be assessed in order to improve inference of cell type specific infection kinetics and disease progression. Our approach provides a method that, in combination with additional experimental data, can be used to disentangle the complex dynamics of viral infection and immunity within human airway epithelial culture systems.


Sujet(s)
COVID-19 , Humains , COVID-19/métabolisme , Cellules épithéliales/métabolisme , SARS-CoV-2 , Cellules cultivées , Épithélium , Muqueuse respiratoire/métabolisme , Muqueuse respiratoire/anatomopathologie
16.
Cells ; 12(8)2023 04 07.
Article de Anglais | MEDLINE | ID: mdl-37190013

RÉSUMÉ

The airway surface liquid (ASL) is a thin sheet of fluid that covers the luminal aspect of the airway epithelium. The ASL is a site of several first-line host defenses, and its composition is a key factor that determines respiratory fitness. Specifically, the acid-base balance of ASL has a major influence on the vital respiratory defense processes of mucociliary clearance and antimicrobial peptide activity against inhaled pathogens. In the inherited disorder cystic fibrosis (CF), loss of cystic fibrosis transmembrane conductance regulator (CFTR) anion channel function reduces HCO3- secretion, lowers the pH of ASL (pHASL), and impairs host defenses. These abnormalities initiate a pathologic process whose hallmarks are chronic infection, inflammation, mucus obstruction, and bronchiectasis. Inflammation is particularly relevant as it develops early in CF and persists despite highly effective CFTR modulator therapy. Recent studies show that inflammation may alter HCO3- and H+ secretion across the airway epithelia and thus regulate pHASL. Moreover, inflammation may enhance the restoration of CFTR channel function in CF epithelia exposed to clinically approved modulators. This review focuses on the complex relationships between acid-base secretion, airway inflammation, pHASL regulation, and therapeutic responses to CFTR modulators. These factors have important implications for defining optimal ways of tackling CF airway inflammation in the post-modulator era.


Sujet(s)
Mucoviscidose , Humains , Mucoviscidose/anatomopathologie , Protéine CFTR , Muqueuse respiratoire/anatomopathologie , Inflammation/anatomopathologie , Concentration en ions d'hydrogène
17.
Int J Surg Pathol ; 31(7): 1414-1419, 2023 Oct.
Article de Anglais | MEDLINE | ID: mdl-36802923

RÉSUMÉ

Biphenotypic sinonasal sarcoma is a newly established tumor entity that is associated with distinct clinicopathological findings. Biphenotypic sinonasal sarcoma is a rare, low-grade spindle cell sarcoma that arises in middle-aged females, exclusively in the sinonasal tract. A fusion gene involving PAX3 is detected in most biphenotypic sinonasal sarcomas, which aids in its diagnosis. Here, we report a case of biphenotypic sinonasal sarcoma with its cytological findings. The patient was a 73-year-old woman who presented with purulent nasal discharge and dull pain in the left cheek area. Computed tomography showed a mass extending from the left nasal cavity to the left ethmoid sinus, the left frontal sinus, and the frontal skull base. She underwent a combined transcranial and endoscopic approach for en bloc resection with a safety margin. Histologically, spindle-shaped tumor cells have been thought to proliferate mainly in the subepithelial stroma. Here, nasal mucosal epithelial hyperplasia was noted, and the tumor had invaded the bone tissue accompanying the epithelial cells. Fluorescence in situ hybridization (FISH) analysis showed a PAX3 rearrangement, and next-generation sequencing identified a PAX3::MAML3 fusion. Based on FISH, split signals were observed not in respiratory cells but in stromal cells. This indicated that respiratory cells were non-neoplastic. In the diagnosis of biphenotypic sinonasal sarcoma, the inverted growth of the respiratory epithelium can be a diagnostic pitfall. FISH analysis using a PAX3 break-apart probe is helpful not only for an accurate diagnosis but also for detecting the true neoplastic cells.


Sujet(s)
Tumeurs des sinus de la face , Sarcomes , Tumeurs des tissus mous , Adulte d'âge moyen , Femelle , Humains , Sujet âgé , Hybridation fluorescente in situ , Tumeurs des sinus de la face/diagnostic , Tumeurs des sinus de la face/génétique , Tumeurs des sinus de la face/anatomopathologie , Sarcomes/anatomopathologie , Muqueuse respiratoire/anatomopathologie , Os et tissu osseux/anatomopathologie
18.
J Oral Pathol Med ; 52(6): 548-553, 2023 Jul.
Article de Anglais | MEDLINE | ID: mdl-36504219

RÉSUMÉ

BACKGROUND: Respiratory epithelial adenomatoid hamartoma (REAH) is a sinonasal glandular overgrowth arising from the surface respiratory epithelium and invaginating into the stroma. Clinically, it appears as a polypoid mass that may cause nasal obstruction, anosmia, and epistaxis. The presence of cartilaginous and/or osseous areas move the lesion to a chondro-osseous respiratory epithelial (CORE) hamartoma subtype. Scattered small seromucinous glands may be observed between typical REAH glands and when it is the only feature, it represents seromucinous hamartoma (SH). The molecular pathogenesis of REAH has been poorly explored and remains unclear. Given that KRAS, BRAF, and EGFR mutations have been detected in a variety of sinonasal tumors, we aimed to assess these mutations in REAH and SH. METHODS: Ten REAH (including one CORE subtype), in addition to two SH cases, were Sanger sequenced by standard techniques. The targeted regions included KRAS exons 2-4 (encompassing hotspots codons 12, 13, 61, and 146), BRAF exons 11 and 15 (spanning the V600 codon), and EGFR exons 19 and 20. RESULTS: All REAH and SH samples showed wild-type sequences for KRAS, BRAF, and EGFR genes. CONCLUSION: Our results demonstrate a lack of KRAS, BRAF, or EGFR pathogenic variants with further evaluation of REAH and SH needed to elucidate driver genetic events.


Sujet(s)
Adénomes , Hamartomes , Humains , Protéines proto-oncogènes B-raf/génétique , Protéines proto-oncogènes p21(ras)/génétique , Muqueuse respiratoire/anatomopathologie , Adénomes/anatomopathologie , Hamartomes/génétique , Hamartomes/diagnostic , Hamartomes/anatomopathologie , Récepteurs ErbB/génétique , Diagnostic différentiel
19.
Int Forum Allergy Rhinol ; 13(9): 1808-1811, 2023 09.
Article de Anglais | MEDLINE | ID: mdl-36533304

RÉSUMÉ

KEY POINTS: Respiratory epithelial adenomatoid hamartoma (REAH) is easily confused with nasal polyps (NP). The typical manifestation of REAH on CT is the enlargement of bilateral olfactory clefts (OCs). The widening of the OCs in the CT scan is a biomarker for diagnosing REAH associated with NP.


Sujet(s)
Adénomes , Hamartomes , Polypes du nez , Humains , Polypes du nez/imagerie diagnostique , Polypes du nez/anatomopathologie , Hamartomes/imagerie diagnostique , Hamartomes/anatomopathologie , Tomodensitométrie , Muqueuse respiratoire/imagerie diagnostique , Muqueuse respiratoire/anatomopathologie , Diagnostic différentiel
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...