Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.555
Filter
1.
Int J Med Sci ; 21(10): 1929-1944, 2024.
Article in English | MEDLINE | ID: mdl-39113893

ABSTRACT

Fine particulate matter (PM2.5) can damage airway epithelial barriers. The anion transport system plays a crucial role in airway epithelial barriers. However, the detrimental effect and mechanism of PM2.5 on the anion transport system are still unclear. In this study, airway epithelial cells and ovalbumin (OVA)-induced asthmatic mice were used. In transwell model, the adenosine triphosphate (ATP)-induced transepithelial anion short-circuit current (Isc) and airway surface liquid (ASL) significantly decreased after PM2.5 exposure. In addition, PM2.5 exposure decreased the expression levels of P2Y2R, CFTR and cytoplasmic free-calcium, but ATP can increase the expressions of these proteins. PM2.5 exposure increased the levels of Th2-related cytokines of bronchoalveolar lavage fluid, lung inflammation, collagen deposition and hyperplasisa of goblet cells. Interestingly, the administration of ATP showed an inhibitory effect on lung inflammation induced by PM2.5. Together, our study reveals that PM2.5 impairs the ATP-induced transepithelial anion Isc through downregulating P2Y2R/CFTR pathway, and this process may participate in aggravating airway hyperresponsiveness and airway inflammation. These findings may provide important insights on PM2.5-mediated airway epithelial injury.


Subject(s)
Asthma , Cystic Fibrosis Transmembrane Conductance Regulator , Particulate Matter , Receptors, Purinergic P2Y2 , Animals , Mice , Receptors, Purinergic P2Y2/metabolism , Receptors, Purinergic P2Y2/genetics , Asthma/metabolism , Asthma/pathology , Asthma/drug therapy , Asthma/chemically induced , Asthma/immunology , Particulate Matter/adverse effects , Particulate Matter/toxicity , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Humans , Adenosine Triphosphate/metabolism , Ovalbumin/immunology , Signal Transduction/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Down-Regulation/drug effects , Respiratory Mucosa/metabolism , Respiratory Mucosa/drug effects , Respiratory Mucosa/pathology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology
2.
Pharmacol Res Perspect ; 12(5): e1242, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39210688

ABSTRACT

Cyclosporin A (CsA), an immunosuppressive drug used in transplant recipients, inhibits graft rejection by binding to cyclophilins and competitively inhibiting calcineurin. While concerns about respiratory infections in immunosuppressed patients exist, contradictory data emerged during the COVID-19 pandemic, prompting investigations into CsA's impact on viral infections. This study explores CsA's antiviral effects on SARS-CoV-2 Omicron BA.1, Delta variants, and human parainfluenza virus 3 (HPIV3) using an ex vivo model of human airway epithelium (HAE). CsA exhibited a dose-dependent antiviral effect against the SARS-CoV-2 Delta variant, reducing viral load over 10 days. However, no significant impact was observed against SARS-CoV-2 Omicron or HPIV3, indicating a virus-specific effect. At high concentrations, CsA was associated with an increase of IL-8 and a decrease of IFNλ expression in infected and noninfected HAE. This study highlights the complexity of CsA's antiviral mechanisms, more likely involving intricate inflammatory pathways and interactions with specific viral proteins. The research provides novel insights into CsA's effects on respiratory viruses, emphasizing the need for understanding drug-virus interactions in optimizing therapeutic approaches for transplant recipients and advancing knowledge on immunosuppressive treatments' implications on respiratory viral infections. Limitations include the model's inability to assess T lymphocyte activation, suggesting the necessity for further comprehensive studies to decipher the intricate dynamics of immunosuppressive treatments on respiratory viral infections.


Subject(s)
Antiviral Agents , Cyclosporine , Immunosuppressive Agents , SARS-CoV-2 , Virus Replication , Humans , Cyclosporine/pharmacology , Virus Replication/drug effects , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Antiviral Agents/pharmacology , Immunosuppressive Agents/pharmacology , Parainfluenza Virus 3, Human/drug effects , Parainfluenza Virus 3, Human/physiology , Respiratory Mucosa/virology , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Viral Load/drug effects , Interferons/pharmacology , Interferons/metabolism , Interleukin-8/metabolism , COVID-19/virology , Dose-Response Relationship, Drug
3.
In Vivo ; 38(5): 2294-2299, 2024.
Article in English | MEDLINE | ID: mdl-39187341

ABSTRACT

BACKGROUND/AIM: Cigarette smoke has been shown to induce a phenotype in humans known as "acquired cystic fibrosis". This occurs because the cystic fibrosis transmembrane conductance regulator (CFTR) functions are impaired systemically due to the deleterious effects of smoke components. Elucidation of cigarette smoke effects on the tracheal epithelium is important. The aim of this study was to develop an ex vivo sheep tracheal model to investigate tracheal ion function. In this model, the epithelial sodium channel (ENaC) is inhibited after exposure to cigarette smoke extract (CSE) as a proof of principle. MATERIALS AND METHODS: Tracheas were isolated from healthy sheep and the tracheal epithelium was surgically excised. Tissues were mounted in Ussing chambers and the short circuit current (Isc) was measured after incubation with 5% CSE in PBS or PBS alone for 30 min. The function of ENaC was investigated by the addition of amiloride (10-5M) apically. Western blot analysis was performed to assess differences in ENaC quantity after CSE exposure. Some specimens were stained with H&E for detection of histological alterations. RESULTS: The amiloride effect on normal epithelium led to a significant decrease in Isc [ΔI=33±5.92 µA/cm2; p<0.001 versus control experiments (ΔI=1.44±0.71 µA/cm2)]. After incubation with CSE, ENaC Isc was significantly reduced (ΔI=14.80±1.96 µA/cm2; p<0.001). No differences in αENaC expression were observed between CSE-exposed and normal tracheal epithelium. Histological images post CSE incubation revealed decreases in the height of the epithelium, with basal cell hyperplasia and loss of ciliated cells. CONCLUSION: Reduced ENaC inhibition by amiloride after CSE incubation could be due to alterations in the tracheal epithelium.


Subject(s)
Epithelial Sodium Channels , Trachea , Animals , Epithelial Sodium Channels/metabolism , Sheep , Trachea/metabolism , Trachea/drug effects , Trachea/pathology , Pilot Projects , Smoke/adverse effects , Amiloride/pharmacology , Respiratory Mucosa/metabolism , Respiratory Mucosa/drug effects , Respiratory Mucosa/pathology , Epithelium/drug effects , Epithelium/metabolism , Epithelium/pathology
4.
FASEB J ; 38(15): e23846, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39093041

ABSTRACT

Epithelial cells play a crucial role in asthma, contributing to chronic inflammation and airway hyperresponsiveness. m6A modification, which involves key proteins such as the demethylase fat mass and obesity-associated protein (FTO), is crucial in the regulation of various diseases, including asthma. However, the role of FTO in epithelial cells and the development of asthma remains unclear. In this study, we investigated the demethylase activity of FTO using a small-molecule inhibitor FB23 in epithelial cells and allergic inflammation in vivo and in vitro. We examined the FTO-regulated transcriptome-wide m6A profiling by methylated RNA immunoprecipitation sequencing (MeRIP-seq) and RNA-seq under FB23 treatment and allergic inflammation conditions. Immunofluorescence staining was performed to assess the tissue-specific expression of FTO in asthmatic bronchial mucosa. We demonstrated that FB23 alleviated allergic inflammation in IL-4/IL-13-treated epithelial cells and house dust mite (HDM)-induced allergic airway inflammation mouse model. The demethylase activity of FTO contributed to the regulation of TNF-α signaling via NF-κB and epithelial-mesenchymal transition-related pathways under allergic inflammation conditions in epithelial cells. FTO was expressed in epithelial, submucosal gland, and smooth muscle cells in human bronchial mucosa. In conclusion, FB23-induced inhibition of FTO alleviates allergic inflammation in epithelial cells and HDM-induced mice, potentially through diverse cellular processes and epithelial-mesenchymal transition signaling pathways, suggesting that FTO is a potential therapeutic target in asthma management.


Subject(s)
Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Asthma , Inflammation , Animals , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Mice , Asthma/metabolism , Asthma/genetics , Inflammation/metabolism , Humans , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Epithelial Cells/metabolism , Mice, Inbred BALB C , Female , Hypersensitivity/metabolism , Hypersensitivity/drug therapy , Epithelial-Mesenchymal Transition/drug effects , Mice, Inbred C57BL
5.
PLoS One ; 19(8): e0306259, 2024.
Article in English | MEDLINE | ID: mdl-39141636

ABSTRACT

The antibacterial oxidative response, which relies on the production of hydrogen peroxide (H2O2) and hypothiocyanite (OSCN-), is a major line of defense protecting the human airway epithelium (HAE) from lesions when infected. The in vitro studies of the oxidative responses are performed mainly by one-shot H2O2 exposure that does not recapitulate the complex H2O2/LPO/SCN- system releasing the reactive oxygen species in airway secretions. A cell-free in vitro assay mimicking this system has been described but was not fully characterized. Here, we comprehensively characterized the hourly H2O2/OSCN- concentrations produced within this in vitro assay and assessed the resistance of Pseudomonas aeruginosa and Staphylococcus aureus clinical strains to the HAE oxidative response. We found that H2O2/OSCN- were steadily produced from 7h and up to 25h, but OSCN- was detoxified in 15 minutes by bacteria upon exposure. Preliminary tests on PA14 showed survival rates at 1-hour post-exposure (hpe) to H2O2 of roughly 50% for 105 and 107 colony-forming unit (CFU)/mL inocula, while 102 and 104 CFU/mL inocula were cleared after one hpe. Thirteen clinical strains were then exposed, highlighting that conversely to P. aeruginosa, S. aureus showed resistance to oxidative stress independently of its antibiotic resistance phenotype. Our results demonstrated how this in vitro assay can be helpful in assessing whether pathogens can resist the antibacterial oxidative HAE response. We anticipate these findings as a starting point for more sophisticated in vitro models that could serve as high-throughput screening for molecules targeting the bacterial antioxidant response.


Subject(s)
Hydrogen Peroxide , Oxidative Stress , Pseudomonas aeruginosa , Staphylococcus aureus , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/drug effects , Staphylococcus aureus/metabolism , Humans , Hydrogen Peroxide/metabolism , Respiratory Mucosa/microbiology , Respiratory Mucosa/metabolism , Oxidation-Reduction , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Pseudomonas Infections/microbiology , Pseudomonas Infections/metabolism , Thiocyanates
6.
Bull Exp Biol Med ; 177(1): 93-97, 2024 May.
Article in English | MEDLINE | ID: mdl-38963595

ABSTRACT

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.


Subject(s)
Bronchi , DNA Methylation , Hyperplasia , Lung Neoplasms , Metaplasia , Precancerous Conditions , Humans , Hyperplasia/pathology , Hyperplasia/genetics , Metaplasia/genetics , Metaplasia/pathology , Metaplasia/metabolism , Bronchi/pathology , Bronchi/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Precancerous Conditions/metabolism , Male , Female , Middle Aged , Epigenome/genetics , Respiratory Mucosa/pathology , Respiratory Mucosa/metabolism , Aged , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Small Cell Lung Carcinoma/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism
8.
Nat Commun ; 15(1): 5817, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987270

ABSTRACT

Respiratory infections caused by the human fungal pathogen Aspergillus fumigatus are a major cause of mortality for immunocompromised patients. Exposure to these pathogens occurs through inhalation, although the role of the respiratory epithelium in disease pathogenesis has not been fully defined. Employing a primary human airway epithelial model, we demonstrate that fungal melanins potently block the post-translational secretion of the chemokines CXCL1 and CXCL8 independent of transcription or the requirement of melanin to be phagocytosed, leading to a significant reduction in neutrophil recruitment to the apical airway both in vitro and in vivo. Aspergillus-derived melanin, a major constituent of the fungal cell wall, dampened airway epithelial chemokine secretion in response to fungi, bacteria, and exogenous cytokines. Furthermore, melanin muted pathogen-mediated calcium fluxing and hindered actin filamentation. Taken together, our results reveal a critical role for melanin interaction with airway epithelium in shaping the host response to fungal and bacterial pathogens.


Subject(s)
Aspergillus fumigatus , Calcium , Chemokine CXCL1 , Interleukin-8 , Melanins , Melanins/metabolism , Humans , Interleukin-8/metabolism , Calcium/metabolism , Chemokine CXCL1/metabolism , Animals , Respiratory Mucosa/metabolism , Respiratory Mucosa/microbiology , Mice , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Chemokines/metabolism , Mice, Inbred C57BL
9.
Int J Mol Sci ; 25(13)2024 Jul 05.
Article in English | MEDLINE | ID: mdl-39000481

ABSTRACT

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


Subject(s)
Betula , Bronchi , Epithelial Cells , Molecular Weight , Pollen , Humans , Bronchi/metabolism , Bronchi/cytology , Bronchi/drug effects , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Cell Line , Allergens , Cytokines/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/drug effects
10.
Sci Rep ; 14(1): 16568, 2024 07 17.
Article in English | MEDLINE | ID: mdl-39019950

ABSTRACT

Mucus stasis is a pathologic hallmark of muco-obstructive diseases, including cystic fibrosis (CF). Mucins, the principal component of mucus, are extensively modified with hydroxyl (O)-linked glycans, which are largely terminated by sialic acid. Sialic acid is a negatively charged monosaccharide and contributes to the biochemical/biophysical properties of mucins. Reports suggest that mucin sialylation may be altered in CF; however, the consequences of reduced sialylation on mucus clearance have not been fully determined. Here, we investigated the consequences of reduced sialylation on the charge state and conformation of the most prominent airway mucin, MUC5B, and defined the functional consequences of reduced sialylation on mucociliary transport (MCT). Reduced sialylation contributed to a lower charged MUC5B form and decreased polymer expansion. The inhibition of total mucin sialylation de novo impaired MCT in primary human bronchial epithelial cells and rat airways, and specific α-2,3 sialylation blockade was sufficient to recapitulate these findings. Finally, we show that ST3 beta-galactoside alpha-2,3-sialyltransferase (ST3Gal1) expression is downregulated in CF and partially restored by correcting CFTR via Elexacaftor/Tezacaftor/Ivacaftor treatment. Overall, this study demonstrates the importance of mucin sialylation in mucus clearance and identifies decreased sialylation by ST3Gal1 as a possible therapeutic target in CF and potentially other muco-obstructive diseases.


Subject(s)
Mucin-5B , Mucus , Humans , Animals , Mucin-5B/metabolism , Rats , Mucus/metabolism , Sialyltransferases/metabolism , N-Acetylneuraminic Acid/metabolism , Mucociliary Clearance , Respiratory Mucosa/metabolism , Cystic Fibrosis/metabolism , Mucins/metabolism , Epithelial Cells/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Bronchi/metabolism
11.
Free Radic Biol Med ; 222: 588-600, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38996820

ABSTRACT

Emerging evidence has reported that acute lung injury (ALI), characterized by inflammation and oxidative stress in airway epithelium, is regulated by programmed cell death. Ferroptosis, a regulated form of cell death spurred by uncontrolled lipid peroxidation, has been proven to implicate various diseases. Inhibiting ferroptosis represents a feasible strategy for ALI through the suppression of lipid peroxidation, while the mechanism remains to be further elucidated. Here, we identified Sequestosome 1 (SQSTM1) as a negative regulator of airway epithelium ferroptosis during ALI. SQSTM1 knockdown cells manifested higher sensitivity to ferroptosis. Mechanistically, SQSTM1 was found to directly interact with vitamin D receptor (VDR) through its nuclear receptor (NR) box motif, facilitating its nuclear translocation and initiating autophagy at the transcriptional level. To further validate these findings, an in vivo preventive model utilizing spermidine, a proven inducer of SQSTM1 was established. The results consistently demonstrated that spermidine supplementation significantly induced SQSTM1 and ameliorated ALI by mitigating airway epithelial ferroptosis. Notably, these effects were abrogated in the absence of SQSTM1. Taken together, this study identified SQSTM1 as a negative regulator of airway epithelium ferroptosis in a VDR-mediated autophagy manner, making it a potential therapeutic target for the treatment of ALI.


Subject(s)
Acute Lung Injury , Autophagy , Ferroptosis , Receptors, Calcitriol , Sequestosome-1 Protein , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/genetics , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Acute Lung Injury/genetics , Ferroptosis/genetics , Ferroptosis/drug effects , Receptors, Calcitriol/metabolism , Receptors, Calcitriol/genetics , Animals , Humans , Mice , Male , Mice, Inbred C57BL , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Oxidative Stress , Lipid Peroxidation/drug effects
12.
Int J Mol Sci ; 25(14)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39063127

ABSTRACT

The production of nanoparticles has recently surged due to their varied applications in the biomedical, pharmaceutical, textile, and electronic sectors. However, this rapid increase in nanoparticle manufacturing has raised concerns about environmental pollution, particularly its potential adverse effects on human health. Among the various concerns, inhalation exposure to nanoparticles poses significant risks, especially affecting the respiratory system. Airway epithelial cells play a crucial role as the primary defense against inhaled particulate matter and pathogens. Studies have shown that nanoparticles can disrupt the airway epithelial barrier, triggering inflammatory responses, generating reactive oxygen species, and compromising cell viability. However, our understanding of how different types of nanoparticles specifically impact the airway epithelial barrier remains limited. Both in vitro cell culture and in vivo murine models are commonly utilized to investigate nanoparticle-induced cellular responses and barrier dysfunction. This review discusses the methodologies frequently employed to assess nanoparticle toxicity and barrier disruption. Furthermore, we analyze and compare the distinct effects of various nanoparticle types on the airway epithelial barrier. By elucidating the diverse responses elicited by different nanoparticles, we aim to provide insights that can guide future research endeavors in assessing and mitigating the potential risks associated with nanoparticle exposure.


Subject(s)
Epithelial Cells , Nanoparticles , Humans , Animals , Nanoparticles/toxicity , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Toxicity Tests/methods , Reactive Oxygen Species/metabolism
13.
mSphere ; 9(8): e0029724, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39041808

ABSTRACT

Interaction between viruses and bacteria during the development of infectious diseases is a complex question that requires continuous study. In this study, we explored the interactions between pseudorabies virus (PRV) and Pasteurella multocida (PM), which are recognized as the primary and secondary agents of porcine respiratory disease complex (PRDC), respectively. In vivo tests using mouse models demonstrated that intranasal inoculation with PRV at a sublethal dose induced disruption of murine respiratory barrier and promoted the invasion and damages caused by PM through respiratory infection. Inoculation with PRV also disrupted the barrier function of murine and porcine respiratory epithelial cells, and accelerated the adherence and invasion of PM to the cells. In mechanism, PRV infection resulted in decreased expression of tight junction proteins (ZO-1, occludin) and adherens junction proteins (ß-catenin, E-cadherin) between neighboring respiratory epithelial cells. Additionally, PRV inoculation at an early stage downregulated multiple biological processes contributing to epithelial adhesion and barrier functions while upregulating signals beneficial for respiratory barrier disruption (e.g., the HIF-1α signaling). Furthermore, PRV infection also stimulated the upregulation of cellular receptors (CAM5, ICAM2, ACAN, and DSCAM) that promote bacterial adherence. The data presented in this study provide insights into the understanding of virus-bacteria interactions in PRDC and may also contribute to understanding the mechanisms of secondary infections caused by different respiratory viruses (e.g., influenza virus and SARS-CoV-2) in both medical and veterinary medicine. IMPORTANCE: Co-infections caused by viral and bacterial agents are common in both medical and veterinary medicine, but the related mechanisms are not fully understood. This study investigated the interactions between the zoonotic pathogens PRV and PM during the development of respiratory infections in both cell and mouse models, and reported the possible mechanisms which included: (i) the primary infection of PRV may induce the disruption and/or damage of mammal respiratory barrier, thereby contributing to the invasion of PM; (ii) PRV infection at early stage accelerates the transcription and/or expression of several cellular receptors that are beneficial for bacterial adherence. This study may shed a light on understanding the mechanisms on the secondary infection of PM promoted by different respiratory viruses (e.g., influenza virus and SARS-CoV-2) in both medical and veterinary medicine.


Subject(s)
Herpesvirus 1, Suid , Pasteurella Infections , Pasteurella multocida , Pseudorabies , Animals , Pasteurella multocida/pathogenicity , Pasteurella multocida/physiology , Mice , Pasteurella Infections/microbiology , Herpesvirus 1, Suid/physiology , Swine , Pseudorabies/virology , Pseudorabies/metabolism , Coinfection/microbiology , Coinfection/virology , Epithelial Cells/virology , Epithelial Cells/microbiology , Permeability , Female , Swine Diseases/microbiology , Swine Diseases/virology , Swine Diseases/metabolism , Mice, Inbred BALB C , Disease Models, Animal , Respiratory Mucosa/virology , Respiratory Mucosa/microbiology , Respiratory Mucosa/metabolism
15.
Nucleic Acids Res ; 52(16): 9369-9383, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39077931

ABSTRACT

A fundamental challenge for cystic fibrosis (CF) gene therapy is ensuring sufficient transduction of airway epithelia to achieve therapeutic correction. Hypertonic saline (HTS) is frequently administered to people with CF to enhance mucus clearance. HTS transiently disrupts epithelial cell tight junctions, but its ability to improve gene transfer has not been investigated. Here, we asked if increasing the concentration of NaCl enhances the transduction efficiency of three gene therapy vectors: adenovirus, AAV, and lentiviral vectors. Vectors formulated with 3-7% NaCl exhibited markedly increased transduction for all three platforms, leading to anion channel correction in primary cultures of human CF epithelial cells and enhanced gene transfer in mouse and pig airways in vivo. The mechanism of transduction enhancement involved tonicity but not osmolarity or pH. Formulating vectors with a high ionic strength solution is a simple strategy to greatly enhance efficacy and immediately improve preclinical or clinical applications.


Subject(s)
Cystic Fibrosis , Genetic Therapy , Genetic Vectors , Transduction, Genetic , Animals , Humans , Genetic Vectors/genetics , Genetic Vectors/chemistry , Mice , Cystic Fibrosis/genetics , Cystic Fibrosis/therapy , Osmolar Concentration , Swine , Genetic Therapy/methods , Adenoviridae/genetics , Dependovirus/genetics , Lentivirus/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Respiratory Mucosa/metabolism , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Gene Transfer Techniques , Saline Solution, Hypertonic
16.
Pulm Pharmacol Ther ; 86: 102314, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38964603

ABSTRACT

Ivacaftor is the first clinically approved monotherapy potentiator to treat CFTR channel dysfunction in people with cystic fibrosis. Ivacaftor (Iva) is a critical component for all current modulator therapies, including highly effective modulator therapies. Clinical studies show that CF patients on ivacaftor-containing therapies present various clinical responses, off-target effects, and adverse reactions, which could be related to metabolites of the compound. In this study, we reported the concentrations of Iva and two of its major metabolites (M1-Iva and M6-Iva) in capillary plasma and estimated M1-Iva and M6-Iva metabolic activity via the metabolite parent ratio in capillary plasma over 12 h. We also used the ratio of capillary plasma versus human nasal epithelial cell concentrations to evaluate entry into epithelial cells in vivo. M6-Iva was rarely detected by LC-MS/MS in epithelial cells from participants taking ivacaftor, although it was detected in plasma. To further explore this discrepancy, we performed in vitro studies, which showed that M1-Iva, but not M6-Iva, readily crossed 16HBE cell membranes. Our studies also suggest that metabolism of these compounds is unlikely to occur in airway epithelia despite evidence of expression of metabolism enzymes. Overall, our data provide evidence that there are differences between capillary and cellular concentrations of these compounds that may inform future studies of clinical response and off-target effects.


Subject(s)
Aminophenols , Cystic Fibrosis , Epithelial Cells , Quinolones , Humans , Quinolones/pharmacology , Aminophenols/pharmacology , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Cystic Fibrosis/drug therapy , Cystic Fibrosis/metabolism , Adult , Male , Female , Tandem Mass Spectrometry , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Chromatography, Liquid , Young Adult , Respiratory Mucosa/metabolism , Respiratory Mucosa/drug effects , Cell Line , Chloride Channel Agonists/pharmacology
17.
J Vis Exp ; (209)2024 Jul 12.
Article in English | MEDLINE | ID: mdl-39072632

ABSTRACT

The preterm neonatal airway epithelium is constantly exposed to environmental stressors. One of these stressors in neonates with lung disease includes oxygen (O2) tension higher than the ambient atmosphere - termed hyperoxia (>21% O2). The effect of hyperoxia on the airway depends on various factors, including the developmental stage of the airway, the degree of hyperoxia, and the duration of exposure, with variable exposures potentially leading to unique phenotypes. While there has been extensive research on the effect of hyperoxia on neonatal lung alveolarization and airway hyperreactivity, little is known about the short and long-term underlying effect of hyperoxia on human neonatal airway epithelial cells. A major reason for this is the scarcity of an effective in vitro model to study human neonatal airway epithelial development and function. Here, we describe a method for isolating and expanding human neonatal tracheal airway epithelial cells (nTAECs) utilizing human neonatal tracheal aspirates and culturing these cells in air-liquid interface (ALI) culture. We demonstrate that nTAECs form a mature polarized cell-monolayer in ALI culture and undergo mucociliary differentiation. We also present a method for moderate hyperoxia exposure of the cell monolayer in ALI culture using a specialized incubator. Additionally, we describe an assay to measure cellular oxidative stress following hyperoxia exposure in ALI culture using fluorescent quantification, which confirms that moderate hyperoxia exposure induces cellular oxidative stress but does not cause significant cell membrane damage or apoptosis. This model can potentially be used to simulate clinically relevant hyperoxia exposure encountered by neonatal airways in the Neonatal Intensive Care Unit (NICU) and used to study the short and long-lasting effects of O2 on neonatal airway epithelial programming. Studies using this model could be utilized to explore ways to mitigate early-life oxidative injury to developing airways, which is implicated in the development of long-term airway diseases in former premature infants.


Subject(s)
Epithelial Cells , Hyperoxia , Humans , Infant, Newborn , Hyperoxia/metabolism , Epithelial Cells/metabolism , Epithelial Cells/cytology , Trachea/cytology , Trachea/metabolism , Cell Culture Techniques, Three Dimensional/methods , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , Cell Culture Techniques/methods
18.
J Vis Exp ; (209)2024 Jul 12.
Article in English | MEDLINE | ID: mdl-39072637

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is largely attributed to tobacco smoke exposure. Investigating how airway epithelial cells functionally adapt to tobacco smoke is crucial for understanding the pathogenesis of COPD. The present study was to set up an in vitro model using primary murine airway epithelial cells to mimic the real-life impact of tobacco smoke. Unlike established cell lines, primary cells retain more in vivo-like properties, including growth patterns, aging, and differentiation. These cells exhibit a sensitive inflammatory response and efficient differentiation, thus closely representing physiological conditions. In this model, primary murine airway epithelial cells were cultured for 28 days under an air-liquid interface with an optimal concentration of cigarette smoke extract (CSE), which led to the transformation of a monolayer of undifferentiated cells into a pseudostratified columnar epithelium, indicative of CSE acclimation. Comprehensive multi-omics analyses were then applied to elucidate the mechanisms by which CSE influences the differentiation of basal airway cells. These insights provide a deeper understanding of the cellular processes underpinning COPD progression in response to tobacco smoke exposure.


Subject(s)
Cell Differentiation , Epithelial Cells , Smoke , Animals , Mice , Cell Differentiation/drug effects , Cell Differentiation/physiology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Smoke/adverse effects , Nicotiana/chemistry , Respiratory Mucosa/cytology , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Tobacco Products , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Disease, Chronic Obstructive/metabolism , Multiomics
19.
Respir Res ; 25(1): 269, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38982492

ABSTRACT

BACKGROUND: Cystic Fibrosis causing mutations in the gene CFTR, reduce the activity of the CFTR channel protein, and leads to mucus aggregation, airway obstruction and poor lung function. A role for CFTR in the pathogenesis of other muco-obstructive airway diseases such as Chronic Obstructive Pulmonary Disease (COPD) has been well established. The CFTR modulatory compound, Ivacaftor (VX-770), potentiates channel activity of CFTR and certain CF-causing mutations and has been shown to ameliorate mucus obstruction and improve lung function in people harbouring these CF-causing mutations. A pilot trial of Ivacaftor supported its potential efficacy for the treatment of mucus obstruction in COPD. These findings prompted the search for CFTR potentiators that are more effective in ameliorating cigarette-smoke (CS) induced mucostasis. METHODS: Small molecule potentiators, previously identified in CFTR binding studies, were tested for activity in augmenting CFTR channel activity using patch clamp electrophysiology in HEK-293 cells, a fluorescence-based assay of membrane potential in Calu-3 cells and in Ussing chamber studies of primary bronchial epithelial cultures. Addition of cigarette smoke extract (CSE) to the solutions bathing the apical surface of Calu-3 cells and primary bronchial airway cultures was used to model COPD. Confocal studies of the velocity of fluorescent microsphere movement on the apical surface of CSE exposed airway epithelial cultures, were used to assess the effect of potentiators on CFTR-mediated mucociliary movement. RESULTS: We showed that SK-POT1, like VX-770, was effective in augmenting the cyclic AMP-dependent channel activity of CFTR. SK-POT-1 enhanced CFTR channel activity in airway epithelial cells previously exposed to CSE and ameliorated mucostasis on the surface of primary airway cultures. CONCLUSION: Together, this evidence supports the further development of SK-POT1 as an intervention in the treatment of COPD.


Subject(s)
Aminophenols , Bronchi , Cystic Fibrosis Transmembrane Conductance Regulator , Epithelial Cells , Quinolones , Humans , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Quinolones/pharmacology , Aminophenols/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Bronchi/drug effects , Bronchi/metabolism , Smoke/adverse effects , Cells, Cultured , HEK293 Cells , Chloride Channel Agonists/pharmacology , Chloride Channel Agonists/therapeutic use , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism
20.
Biochem J ; 481(14): 959-980, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38941070

ABSTRACT

While IκB-kinase-ε (IKKε) induces immunomodulatory genes following viral stimuli, its up-regulation by inflammatory cytokines remains under-explored. Since airway epithelial cells respond to airborne insults and potentiate inflammation, IKKε expression was characterized in pulmonary epithelial cell lines (A549, BEAS-2B) and primary human bronchial epithelial cells grown as submersion or differentiated air-liquid interface cultures. IKKε expression was up-regulated by the pro-inflammatory cytokines, interleukin-1ß (IL-1ß) and tumour necrosis factor-α (TNFα). Thus, mechanistic interrogations in A549 cells were used to demonstrate the NF-κB dependence of cytokine-induced IKKε. Furthermore, chromatin immunoprecipitation in A549 and BEAS-2B cells revealed robust recruitment of the NF-κB subunit, p65, to one 5' and two intronic regions within the IKKε locus (IKBKE). In addition, IL-1ß and TNFα induced strong RNA polymerase 2 recruitment to the 5' region, the first intron, and the transcription start site. Stable transfection of the p65-binding regions into A549 cells revealed IL-1ß- and TNFα-inducible reporter activity that required NF-κB, but was not repressed by glucocorticoid. While critical NF-κB motifs were identified in the 5' and downstream intronic regions, the first intronic region did not contain functional NF-κB motifs. Thus, IL-1ß- and TNFα-induced IKKε expression involves three NF-κB-binding regions, containing multiple functional NF-κB motifs, and potentially other mechanisms of p65 binding through non-classical NF-κB binding motifs. By enhancing IKKε expression, IL-1ß may prime, or potentiate, responses to alternative stimuli, as modelled by IKKε phosphorylation induced by phorbol 12-myristate 13-acetate. However, since IKKε expression was only partially repressed by glucocorticoid, IKKε-dependent responses could contribute to glucocorticoid-resistant disease.


Subject(s)
Epithelial Cells , I-kappa B Kinase , Humans , I-kappa B Kinase/metabolism , I-kappa B Kinase/genetics , Epithelial Cells/metabolism , Epithelial Cells/drug effects , A549 Cells , Transcription Factor RelA/metabolism , Transcription Factor RelA/genetics , Interleukin-1beta/pharmacology , Interleukin-1beta/metabolism , Interleukin-1beta/genetics , NF-kappa B/metabolism , NF-kappa B/genetics , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/genetics , Lung/metabolism , Lung/cytology , Respiratory Mucosa/metabolism , Respiratory Mucosa/cytology , Gene Expression Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL