Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Respir Res ; 23(1): 43, 2022 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-35241091

RESUMEN

BACKGROUND: The prevalence of age-associated diseases, such as chronic obstructive pulmonary disease (COPD), is increasing as the average life expectancy increases around the world. We previously identified a gene signature for ageing in the human lung which included genes involved in apical and tight junction assembly, suggesting a role for airway epithelial barrier dysfunction with ageing. AIM: To investigate the association between genes involved in epithelial barrier function and age both in silico and in vitro in the airway epithelium. METHODS: We curated a gene signature of 274 genes for epithelial barrier function and tested the association with age in two independent cohorts of bronchial brushings from healthy individuals with no respiratory disease, using linear regression analysis (FDR < 0.05). Protein-protein interactions were identified using STRING©. The barrier function of primary bronchial epithelial cells at air-liquid interface and CRISPR-Cas9-induced knock-down of target genes in human bronchial 16HBE14o-cells was assessed using Trans epithelial resistance (TER) measurement and Electric cell-surface impedance sensing (ECIS) respectively. RESULTS: In bronchial brushings, we found 55 genes involved in barrier function to be significantly associated with age (FDR < 0.05). EPCAM was most significantly associated with increasing age and TRPV4 with decreasing age. Protein interaction analysis identified CDH1, that was negatively associated with higher age, as potential key regulator of age-related epithelial barrier function changes. In vitro, barrier function was lower in bronchial epithelial cells from subjects > 45 years of age and significantly reduced in CDH1-deficient 16HBE14o-cells. CONCLUSION: The significant association between genes involved in epithelial barrier function and age, supported by functional studies in vitro, suggest a role for epithelial barrier dysfunction in age-related airway disease.


Asunto(s)
Envejecimiento/fisiología , Bronquios/metabolismo , Células Epiteliales/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Adolescente , Adulto , Anciano , Bronquios/patología , Células Cultivadas , Células Epiteliales/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/patología , Mucosa Respiratoria/metabolismo , Adulto Joven
2.
Physiol Genomics ; 50(9): 705-713, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29932825

RESUMEN

BACKGROUND: Understanding effects of acute smoke exposure (ASE) on airway epithelial gene expression and their relationship with the effects of chronic smoke exposure may provide biological insights into the development of smoking-related respiratory diseases. METHODS: Bronchial airway epithelial cell brushings were collected from 63 individuals without recent cigarette smoke exposure and before and 24 h after smoking three cigarettes. RNA from these samples was profiled on Affymetrix Human Gene 1.0 ST microarrays. RESULTS: We identified 91 genes differentially expressed 24 h after ASE (false discovery rate < 0.25). ASE induced genes involved in xenobiotic metabolism, oxidative stress, and inflammation and repressed genes related to cilium morphogenesis and cell cycle. While many genes altered by ASE are altered similarly in chronic smokers, metallothionein genes are induced by ASE and suppressed in chronic smokers. Metallothioneins are also suppressed in current and former smokers with lung cancer relative to those without lung cancer. CONCLUSIONS: Acute exposure to as little as three cigarettes and chronic smoking induce largely concordant changes in airway epithelial gene expression. Differences in short-term and long-term effects of smoking on metallothionein expression and their relationship to lung cancer requires further study given these enzymes' role in the oxidative stress response.


Asunto(s)
Bronquios/metabolismo , Bronquios/patología , Regulación de la Expresión Génica , Fumar/efectos adversos , Adolescente , Adulto , Anciano , Femenino , Humanos , Masculino , Metalotioneína/metabolismo , Persona de Mediana Edad , Cese del Hábito de Fumar , Adulto Joven
3.
Pulm Pharmacol Ther ; 50: 47-56, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29627483

RESUMEN

BACKGROUND: COPD patients have increased risk of pneumonia when treated with fluticasone propionate (FP), whereas this is generally not the case with budesonide (BUD) treatment. We hypothesized that BUD and FP differentially affect the expression of immune defense genes. METHODS: Human bronchial epithelial 16HBE cells and air-liquid interface (ALI)-cultured primary bronchial epithelial cells (PBECs) were pre-treated with clinically equipotent concentrations of BUD or FP (0.16-16 nM BUD and 0.1-10 nM FP), and the expression of immune defense genes was studied at baseline and after exposure to rhinovirus (RV16). RESULTS: Using microfluidic cards, we observed that both BUD and FP significantly suppressed CXCL8, IFNB1 and S100A8 mRNA expression in unstimulated 16HBE cells. Interestingly, BUD, but not FP, significantly increased lactotransferrin (LTF) expression. The difference between the effect of BUD and FP on LTF expression was statistically significant and confirmed by qPCR and at the protein level by western blotting. RV16 infection of ALI-cultured PBECs significantly increased the expression of CCL20, IFNB1 and S100A8, but not of LTF or CAMP/LL-37. In these RV16-exposed cells, LTF expression was again significantly higher upon pre-treatment with BUD than with FP. The same was observed for S100A8, but not for CCL20, IFNB1 or CAMP/LL-37 expression. CONCLUSIONS: Treatment of human bronchial epithelial cells with BUD results in significantly higher expression of specific immune defense genes than treatment with FP. The differential regulation of these immune defense genes may help to explain the clinical observation that BUD and FP treatment differ with respect to the risk of developing pneumonia in COPD.


Asunto(s)
Bronquios/efectos de los fármacos , Bronquios/inmunología , Broncodilatadores/farmacología , Budesonida/farmacología , Citocinas/genética , Fluticasona/farmacología , Bronquios/citología , Línea Celular , Citocinas/biosíntesis , Citocinas/inmunología , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Expresión Génica/efectos de los fármacos , Expresión Génica/inmunología , Humanos , Lactoferrina/biosíntesis , Lactoferrina/genética , Lactoferrina/inmunología , Poli I-C/farmacología , ARN Mensajero/biosíntesis , ARN Mensajero/genética
4.
Respir Res ; 17: 2, 2016 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-26739349

RESUMEN

BACKGROUND: COPD patients have a higher risk of pneumonia when treated with fluticasone propionate (FP) than with placebo, and a lower risk with budesonide (BUD). We hypothesized that BUD and FP differentially affect the mucosal barrier in response to viral infection and/or cigarette smoke. METHODS: We assessed protective effects of equivalent concentrations of BUD and FP on cytokine production and barrier function (electrical resistance) in human bronchial epithelial 16HBE cells and primary bronchial epithelial cells (PBECs) upon exposure to viral mimetic poly-(I:C) and/or cigarette smoke extract (CSE) or epidermal growth factor (EGF). RESULTS: BUD and FP were equally effective in suppressing poly-(I:C)- and/or CSE-induced IL-8 secretion in 16HBE and PBECs. Poly-(I:C) substantially decreased electrical resistance in 16HBE cells and both BUD and FP fully counteracted this effect. However, FP hardly affected 16HBE barrier dysfunction induced by CSE with/without poly-(I:C), whereas BUD (16 nM) provided full protection, an effect likely mediated by affecting EGFR-downstream target GSK-3ß. Similarly, BUD, but not FP, significantly improved CSE-induced barrier dysfunction in PBECs. Finally, BUD, but not FP, exerted a modest but significant protective effect against Streptococcus Pneumoniae-induced barrier dysfunction, and BUD, but not FP, prevented cellular adhesion and/or internalization of these bacteria induced by poly-(I:C) in 16HBE. CONCLUSIONS: Collectively, both BUD and FP efficiently control epithelial pro-inflammatory responses and barrier function upon mimicry of viral infection. Of potential clinical relevance, BUD more effectively counteracted CSE-induced barrier dysfunction, reinforcing the epithelial barrier and potentially limiting access of pathogens upon smoking in vivo.


Asunto(s)
Bronquios/inmunología , Budesonida/administración & dosificación , Células Epiteliales/inmunología , Células Epiteliales/virología , Fluticasona/administración & dosificación , Poli C/inmunología , Bronquios/efectos de los fármacos , Bronquios/virología , Broncodilatadores/administración & dosificación , Línea Celular , Permeabilidad de la Membrana Celular/efectos de los fármacos , Permeabilidad de la Membrana Celular/inmunología , Citocinas/inmunología , Relación Dosis-Respuesta a Droga , Células Epiteliales/efectos de los fármacos , Humanos , Rhinovirus/efectos de los fármacos , Rhinovirus/fisiología , Breas
5.
Am J Physiol Lung Cell Mol Physiol ; 309(11): L1344-53, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26453516

RESUMEN

Most patients with allergic asthma are sensitized to house dust mite (HDM). The allergenicity of HDM largely depends on disruption of the integrity and proinflammatory activation of the airway epithelium. In this study, we hypothesized that Pim1 kinase activity attenuates HDM-induced asthma by preserving airway epithelial integrity. The effects of Pim1 kinase activity on barrier function and release of the proinflammatory mediators IL-1α and CCL20 were studied in vitro in 16HBE and primary bronchial epithelial cells (PBECs). Pim1-proficient and -deficient mice were exposed to a HDM-driven model of allergic asthma, and airway hyperresponsiveness (AHR) was measured upon methacholine challenge. Airway inflammation and proinflammatory mediators in lung tissue and BAL fluid were determined. We observed that inhibition of Pim1 kinase prolongs the HDM-induced loss of barrier function in 16HBE cells and sensitizes PBECs to HDM-induced barrier dysfunction. Additionally, inhibition of Pim1 kinase increased the HDM-induced proinflammatory activity of 16HBE cells as measured by IL-1α secretion. In line herewith, HDM exposure induced an enhanced production of the proinflammatory chemokines CCL17 and CCL20 in Pim1-deficient mice compared with wild-type controls. While we observed a marked increase in eosinophilic and neutrophilic granulocytes as well as mucus cell metaplasia and AHR to methacholine in mice exposed to HDM, these parameters were independent of Pim1 kinase activity. In contrast, levels of the Th2-cytokines IL-5 and IL-10 were significantly augmented in HDM-treated Pim1-deficient mice. Taken together, our study shows that Pim1 kinase activity maintains airway epithelial integrity and protects against HDM-induced proinflammatory activation of the airway epithelium.


Asunto(s)
Bronquios/patología , Células Epiteliales/enzimología , Células Epiteliales/parasitología , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Pyroglyphidae/fisiología , Adulto , Anciano , Animales , Líquido del Lavado Bronquioalveolar/citología , Línea Celular , Quimiocinas/metabolismo , Células Epiteliales/patología , Femenino , Humanos , Inflamación/parasitología , Inflamación/patología , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Neumonía/patología , Proteínas Proto-Oncogénicas c-pim-1/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-pim-1/deficiencia , Hipersensibilidad Respiratoria/enzimología , Hipersensibilidad Respiratoria/parasitología , Hipersensibilidad Respiratoria/patología , Células Th2/inmunología , Adulto Joven
6.
Allergy ; 70(12): 1545-52, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26296735

RESUMEN

BACKGROUND: House dust mite (HDM) acts on the airway epithelium to induce airway inflammation in asthma. We previously showed that the ability of HDM to induce allergic sensitization in mice is related to airway epithelial CCL20 secretion. OBJECTIVE: As a disintegrin and metalloprotease (ADAM)s have been implicated in chemokine shedding, we sought to determine their involvement in HDM-induced release of chemokines, including CCL20, by airway epithelial cells. METHODS: We studied the effects of pharmacological ADAM inhibitors as well as ADAM10 and ADAM17 siRNA downregulation on chemokine release using (multiplex) ELISA in supernatants from HDM-exposed human bronchial epithelial 16HBE cells and primary normal human bronchial epithelial cells (NHBE) at 4-24 h. RESULTS: House dust) mite markedly increased CCL20 levels in both 16HBE and NHBE cells (16-24 h). In 16HBE cells, the HDM-induced increase was observed as early as 4 h upon exposure and the use of specific inhibitors indicated the involvement of ADAM10/17-mediated shedding. siRNA knockdown of ADAM10, but not of ADAM17, significantly reduced the HDM-induced release of CCL20 in both 16HBE and NHBE cells. A similar effect was observed for HDM-induced CCL2, CCL5, and CXCL8 release in NHBE cells. The HDM-induced increase in CCL20 levels was not affected by protein synthesis inhibitor cycloheximide nor protein transport inhibitor monensin, indicating that HDM induces surface shedding of chemokines. CONCLUSION: Our data show for the first time that ADAM10 activity contributes to HDM-induced shedding of chemokines, including CCL20. The ADAM10/CCL20 axis may be a target for novel therapeutic strategies in asthma.


Asunto(s)
Proteínas ADAM/inmunología , Secretasas de la Proteína Precursora del Amiloide/inmunología , Asma/inmunología , Quimiocina CCL20/metabolismo , Proteínas de la Membrana/inmunología , Pyroglyphidae/inmunología , Hipersensibilidad Respiratoria/inmunología , Mucosa Respiratoria/inmunología , Proteínas ADAM/metabolismo , Proteína ADAM10 , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Antígenos Dermatofagoides/inmunología , Asma/metabolismo , Western Blotting , Células Cultivadas , Quimiocina CCL20/inmunología , Humanos , Proteínas de la Membrana/metabolismo , ARN Interferente Pequeño , Hipersensibilidad Respiratoria/metabolismo , Mucosa Respiratoria/metabolismo , Transfección
7.
Am J Physiol Lung Cell Mol Physiol ; 307(3): L240-51, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24816488

RESUMEN

Exposure to cigarette smoke (CS) is the main risk factor for developing chronic obstructive pulmonary disease and can induce airway epithelial cell damage, innate immune responses, and airway inflammation. We hypothesized that cell survival factors might decrease the sensitivity of airway epithelial cells to CS-induced damage, thereby protecting the airways against inflammation upon CS exposure. Here, we tested whether Pim survival kinases could protect from CS-induced inflammation. We determined expression of Pim kinases in lung tissue, airway inflammation, and levels of keratinocyte-derived cytokine (KC) and several damage-associated molecular patterns in bronchoalveolar lavage in mice exposed to CS or air. Human bronchial epithelial BEAS-2B cells were treated with CS extract (CSE) in the presence or absence of Pim1 inhibitor and assessed for loss of mitochondrial membrane potential, induction of cell death, and release of heat shock protein 70 (HSP70). We observed increased expression of Pim1, but not of Pim2 and Pim3, in lung tissue after exposure to CS. Pim1-deficient mice displayed a strongly enhanced neutrophilic airway inflammation upon CS exposure compared with wild-type controls. Inhibition of Pim1 activity in BEAS-2B cells increased the loss of mitochondrial membrane potential and reduced cell viability upon CSE treatment, whereas release of HSP70 was enhanced. Interestingly, we observed release of S100A8 but not of double-strand DNA or HSP70 in Pim1-deficient mice compared with wild-type controls upon CS exposure. In conclusion, we show that expression of Pim1 protects against CS-induced cell death in vitro and neutrophilic airway inflammation in vivo. Our data suggest that the underlying mechanism involves CS-induced release of S100A8 and KC.


Asunto(s)
Células Epiteliales/metabolismo , Inflamación/metabolismo , Pulmón/metabolismo , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Fumar/efectos adversos , Fumar/metabolismo , Animales , Líquido del Lavado Bronquioalveolar , Muerte Celular/fisiología , Células Cultivadas , Quimiocinas/metabolismo , Células Epiteliales/patología , Femenino , Proteínas HSP70 de Choque Térmico/metabolismo , Inflamación/patología , Pulmón/patología , Potencial de la Membrana Mitocondrial/fisiología , Ratones , Ratones Endogámicos BALB C , Neutrófilos/metabolismo , Neutrófilos/patología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/patología , Fumar/patología
8.
Clin Exp Allergy ; 44(5): 620-30, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24612268

RESUMEN

The integrity of the airway epithelium in patients with asthma is often disrupted, with loss of epithelial cell-cell contacts. Airway epithelial barrier dysfunction may have important implications for asthma, because structural epithelial barrier function is tightly interwoven with the ability of the epithelium to regulate the immune system. We propose that changes at the airway epithelial barrier play a central role in the sensitisation to allergens and pathogenesis of allergic asthma. Many of the recently identified susceptibility genes for asthma are expressed in airway epithelium. However, the exact mechanisms by which the expression of epithelial susceptibility genes translates into a functionally altered response to aeroallergens in asthma are still unknown. In this review, we will focus on the role of airway epithelial barrier function in the susceptibility to develop allergic asthma and discuss therapeutic strategies aimed at the epithelial barrier.


Asunto(s)
Asma/inmunología , Asma/metabolismo , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Animales , Antiasmáticos/farmacología , Antiasmáticos/uso terapéutico , Asma/tratamiento farmacológico , Asma/genética , Comunicación Celular , Exposición a Riesgos Ambientales/efectos adversos , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/patología , Factores de Riesgo
9.
Allergy ; 68(9): 1117-25, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23915187

RESUMEN

BACKGROUND: House dust mite (HDM) affects the immunological and physical barrier function of airway epithelium, leading to allergic sensitization, airway remodeling, and eosinophilic inflammation in mouse models, although the mechanisms are still largely unknown. OBJECTIVE: Given the implications for adenosine triphosphate (ATP)-dependent Ca(2+) signaling in allergic sensitization in mice, we sought to determine the role of intracellular Ca(2+) concentration ([Ca(2+)](i)) in HDM-induced barrier dysfunction and pro-inflammatory activity of bronchial epithelium. METHODS: We investigated the effect of HDM on accumulation of [Ca(2+)](i) levels, barrier function, and CCL20 release in human bronchial epithelial 16HBE cells and primary bronchial epithelial cells (PBECs) from healthy subjects and asthma patients. Involvement of ATP-dependent activation of purinergic receptors and downstream Ca(2+) influx was studied, using the ATP hydrolyzing agent apyrase, the purinergic receptor agonist PPADS, the calcium chelator BAPTA-AM, and calpain inhibitors. RESULTS: Asthma PBECs were more susceptible to HDM-induced barrier dysfunction, CCL20 secretion, and Ca(2+) influx than healthy PBECs. Furthermore, we show that the HDM-induced increase in CCL20 in PBECs and 16HBE cells and the HDM-induced barrier dysfunction in 16HBE cells are dependent on [Ca(2+)](i) accumulation. Additionally, we demonstrate that [Ca(2+)](i) accumulation is initiated partly through the activation of purinergic receptors, which contributes to HDM-induced epithelial barrier dysfunction by disruption of cell-cell contacts, but not CCL20 secretion. CONCLUSION: Our data show for the first time that Ca(2+) signaling plays a crucial role in barrier dysfunction and the pro-inflammatory response of bronchial epithelium upon HDM exposure and may thus have important implications for the development of allergic asthma.


Asunto(s)
Señalización del Calcio , Quimiocina CCL20/biosíntesis , Pyroglyphidae/inmunología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Adenosina Trifosfato/metabolismo , Adulto , Animales , Asma/inmunología , Asma/fisiopatología , Cadherinas/metabolismo , Calcio/metabolismo , Estudios de Casos y Controles , Línea Celular , Femenino , Humanos , Masculino , Transporte de Proteínas , Receptores Purinérgicos/metabolismo , Mucosa Respiratoria/fisiopatología , Adulto Joven
10.
Thorax ; 67(6): 488-95, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22167364

RESUMEN

BACKGROUND: House dust mite (HDM) allergens have been reported to increase airway epithelial permeability, thereby facilitating access of allergens and allergic sensitisation. OBJECTIVES: The authors aimed to understand which biochemical properties of HDM are critical for epithelial immune and barrier responses as well as T helper 2-driven experimental asthma in vivo. METHODS: Three commercially available HDM extracts were analysed for endotoxin levels, protease and chitinase activities and effects on transepithelial resistance, junctional proteins and pro-inflammatory cytokine release in the bronchial epithelial cell line 16HBE and normal human bronchial cells. Furthermore, the effects on epithelial remodelling and airway inflammation were investigated in a mouse model. RESULTS: The different HDM extracts varied extensively in their biochemical properties and induced divergent responses in vitro and in vivo. Importantly, the Greer extract, with the lowest serine protease activity, induced the most pronounced effects on epithelial barrier function and CCL20 release in vitro. In vivo, this extract induced the most profound epithelial E-cadherin delocalisation and increase in CCL20, CCL17 and interleukin 5 levels, accompanied by the most pronounced induction of HDM-specific IgE, goblet cell hyperplasia, eosinophilic inflammation and airway hyper-reactivity. CONCLUSIONS: This study shows the ability of HDM extracts to alter epithelial immune and barrier responses is related to allergic sensitisation but independent of serine/cysteine protease activity.


Asunto(s)
Antígenos Dermatofagoides/inmunología , Asma/inmunología , Pyroglyphidae/inmunología , Mucosa Respiratoria/inmunología , Animales , Asma/fisiopatología , Biomarcadores/metabolismo , Cadherinas/inmunología , Quimiocina CCL17/inmunología , Quimiocina CCL20/inmunología , Citocinas/inmunología , Dermatophagoides pteronyssinus/inmunología , Modelos Animales de Enfermedad , Humanos , Hipersensibilidad/inmunología , Técnicas In Vitro , Interleucina-5/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Mucosa Respiratoria/fisiopatología
11.
Eur Respir J ; 39(2): 419-28, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21778164

RESUMEN

Cigarette smoking, the major cause of chronic obstructive pulmonary disease (COPD), induces aberrant airway epithelial structure and function. The underlying mechanisms are unresolved so far. We studied effects of cigarette smoke extract (CSE) on epithelial barrier function and wound regeneration in human bronchial epithelial 16HBE cells and primary bronchial epithelial cells (PBECs) from COPD patients, nonsmokers and healthy smokers. We demonstrate that CSE rapidly and transiently impairs 16HBE barrier function, largely due to disruption of cell-cell contacts. CSE induced a similar, but stronger and more sustained, defect in PBECs. Application of the specific epidermal growth factor receptor (EGFR) inhibitor AG1478 showed that EGFR activation contributes to the CSE-induced defects in both 16HBE cells and PBECs. Furthermore, our data indicate that the endogenous protease calpain mediates these defects through tight junction protein degradation. CSE also delayed the reconstitution of 16HBE intercellular contacts during wound healing and attenuated PBEC barrier function upon wound regeneration. These findings were comparable between PBECs from smokers, healthy smokers and COPD patients. In conclusion, we demonstrate for the first time that CSE reduces epithelial integrity, probably by EGFR and calpain-dependent disruption of intercellular contacts. This may increase susceptibility to environmental insults, e.g. inhaled pathogens. Thus, EGFR may be a promising target for therapeutic strategies to improve mucosal barrier function in cigarette smoking-related disease.


Asunto(s)
Comunicación Celular/fisiología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/patología , Mucosa Respiratoria/citología , Fumar/efectos adversos , División Celular/fisiología , Células Cultivadas , Impedancia Eléctrica , Electroporación , Inhibidores Enzimáticos/farmacología , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Permeabilidad , Enfermedad Pulmonar Obstructiva Crónica/etiología , Quinazolinas/farmacología , Mucosa Respiratoria/efectos de los fármacos , Uniones Estrechas/fisiología , Tirfostinos/farmacología , Cicatrización de Heridas/fisiología
12.
Eur Respir J ; 39(2): 439-45, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21828034

RESUMEN

A subset of asthma patients suffer from glucocorticoid (GC) insensitivity. T-helper cell type 17 cells have an emerging role in GC insensitivity, although the mechanisms are still poorly understood. We investigated whether interleukin (IL)-17A induces GC insensitivity in airway epithelium by studying its effects on responsiveness of tumour necrosis factor (TNF)-α-induced IL-8 production to budesonide in human bronchial epithelial 16HBE cells. We unravelled the underlying mechanism by the use of specific pathway inhibitors, reporter and overexpression constructs and a histone deacetylase (HDAC) activity assay. We demonstrated that IL-17A-induced IL-8 production is normally sensitive to GCs, while IL-17A pre-treatment significantly reduced the sensitivity of TNF-α-induced IL-8 production to budesonide. IL-17A activated the p38, extracellular signal-related kinase (ERK) and phosphoinositide-3-kinase (PI3K) pathways, and the latter appeared to be involved in IL-17A-induced GC insensitivity. Furthermore, IL-17A reduced HDAC activity, and overexpression of HDAC2 reversed IL-17A-induced GC insensitivity. In contrast, IL-17A did not affect budesonide-induced transcriptional activity of the GC receptor, suggesting that IL-17A does not impair the actions of the ligated GC receptor. In conclusion, we have shown for the first time that IL-17A induces GC insensitivity in airway epithelium, which is probably mediated by PI3K activation and subsequent reduction of HDAC2 activity. Thus, blockade of IL-17A or downstream signalling molecule PI3K may offer new strategies for therapeutic intervention in GC-insensitive asthma.


Asunto(s)
Asma/inmunología , Resistencia a Medicamentos/inmunología , Glucocorticoides/farmacología , Interleucina-17/inmunología , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/inmunología , Asma/tratamiento farmacológico , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Histona Desacetilasa 2/metabolismo , Humanos , Interleucina-17/metabolismo , Interleucina-8/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Glucocorticoides/inmunología , Receptores de Glucocorticoides/metabolismo , Mucosa Respiratoria/citología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Células Th17/efectos de los fármacos , Células Th17/inmunología , Transcripción Genética/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
Eur Respir J ; 36(5): 1016-26, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20351035

RESUMEN

Impaired airway epithelial barrier function has emerged as a key factor in the pathogenesis of allergic asthma. We aimed to discern the involvement of the epidermal growth factor receptor (EGFR) in allergen-induced epithelial barrier impairment, as we previously observed that house dust mite (HDM) signals through EGFR. We investigated the junctional integrity of human bronchial epithelial cells using electric cell-substrate impedance sensing and immunofluorescent staining. HDM induced a rapid, transient fall in epithelial resistance, concomitant with delocalisation of E-cadherin and zona occludens (ZO)-1, and proteolytic cleavage of the latter. EGFR inhibition by AG1478 reduced the HDM-triggered decrease in epithelial resistance and improved restoration of epithelial junctions. Similarly, AG1478 increased epithelial barrier recovery upon electroporation-induced injury, although it delayed the migration phase of the wound healing response. HDM-promoted redistribution of E-cadherin was mediated via EGFR-dependent activation of protease-activated receptor (PAR)-2, while the concomitant ZO-1 degradation was PAR-2/EGFR-independent. Importantly, the fibrogenic cytokine transforming growth factor (TGF)-ß prolonged HDM-induced EGFR phosphorylation and inhibited ligand-induced EGFR internalisation/degradation, which resulted in sustained E-cadherin and ZO-1 redistribution. Thus, allergen-induced, PAR-2/EGFR-mediated signalling decreases epithelial resistance and promotes junction disassembly. The TGF-ß-enhanced EGFR signalling may be an important contributor to barrier dysfunction and increased epithelial vulnerability in response to HDM.


Asunto(s)
Receptores ErbB/metabolismo , Pyroglyphidae/inmunología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Transducción de Señal/inmunología , Animales , Bronquios/citología , Cadherinas/metabolismo , Comunicación Celular/inmunología , Línea Celular , Electroporación , Receptores ErbB/inmunología , Humanos , Uniones Intercelulares/inmunología , Uniones Intercelulares/metabolismo , Proteínas de la Membrana/metabolismo , Permeabilidad , Fosfoproteínas/metabolismo , Mucosa Respiratoria/citología , Factor de Crecimiento Transformador beta/metabolismo , Cicatrización de Heridas/fisiología , Proteína de la Zonula Occludens-1
14.
Eur Respir J ; 35(4): 894-903, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19741028

RESUMEN

Research on epithelial cell lines and primary epithelium is required to dissect the mechanisms underlying the structural abnormalities in airway epithelium observed for respiratory diseases, including asthma and chronic obstructive pulmonary disease. The novel electric cell-substrate impedance sensing technique was used to monitor cell adhesion/spreading, barrier function and wound healing. Primary bronchial epithelium was compared with airway epithelial cell lines 16HBE14o-, BEAS-2B, NCI-H292 and A549. BEAS-2B, A549 and primary cells form a confluent monolayer more rapidly than do 16HBE14o- cells. In contrast, 16HBE14o- cells form stronger intercellular contacts, with a 10-fold higher resistance than BEAS-2B, A549 and NCI-H292 cells and a five-fold increase over primary cells. Accordingly, expression of the adhesion molecules zona occludens-1 and E-cadherin was highest in 16HBE14o- cells. These molecules were localised in intercellular junctions in both 16HBE14o- and primary cells. Finally, restoration of barrier function upon injury was impaired in BEAS-2B compared to 16HBE14o- cells. In conclusion, epithelial cell types display remarkable phenotypic differences and should, accordingly, be used to address specific research questions. 16HBE14o- cells appear most suitable for studies on barrier formation, whereas resemblance in attachment of primary and BEAS-2B and A549 cells makes the latter more important for translational research on cell-matrix contact.


Asunto(s)
Adhesión Celular/fisiología , Impedancia Eléctrica , Células Epiteliales/citología , Células Epiteliales/fisiología , Mucosa Respiratoria/citología , Cadherinas/metabolismo , Comunicación Celular/fisiología , Movimiento Celular/fisiología , Células Cultivadas , Humanos , Uniones Intercelulares/metabolismo , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Proteína de la Zonula Occludens-1
15.
Sci Rep ; 9(1): 15047, 2019 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-31636329

RESUMEN

COPD is characterized by chronic lung inflammation and irreversible lung tissue damage. Inhaled noxious gases, including cigarette smoke, are the major risk factor for COPD. Inhaled smoke first encounters the epithelial lining of the lungs, causing oxidative stress and mitochondrial dysfunction. We investigated whether a mitochondrial defect may contribute to increased lung epithelial pro-inflammatory responses, impaired epithelial repair and reduced corticosteroid sensitivity as observed in COPD. We used wild-type alveolar epithelial cells A549 and mitochondrial DNA-depleted A549 cells (A549 Rho-0) and studied pro-inflammatory responses using (multiplex) ELISA as well as epithelial barrier function and repair (real-time impedance measurements), in the presence and absence of the inhaled corticosteroid budesonide. We observed that A549 Rho-0 cells secrete higher levels of pro-inflammatory cytokines than wild-type A549 cells and display impaired repair upon wounding. Budesonide strongly suppressed the production of neutrophil attractant CXCL8, and promoted epithelial integrity in A549 wild-type cells, while A549 Rho-0 cells displayed reduced corticosteroid sensitivity compared to wild-type cells. The reduced corticosteroid responsiveness may be mediated by glycolytic reprogramming, specifically glycolysis-associated PI3K signaling, as PI3K inhibitor LY294002 restored the sensitivity of CXCL8 secretion to corticosteroids in A549 Rho-0 cells. In conclusion, mitochondrial defects may lead to increased lung epithelial pro-inflammatory responses, reduced epithelial repair and reduced corticosteroid responsiveness in lung epithelium, thus potentially contributing to the pathogenesis of COPD.


Asunto(s)
Corticoesteroides/farmacología , Citocinas/biosíntesis , Epitelio/patología , Mediadores de Inflamación/metabolismo , Pulmón/patología , Mitocondrias/patología , Cicatrización de Heridas/efectos de los fármacos , Células A549 , Quimiocinas/metabolismo , ADN Mitocondrial/genética , Epitelio/efectos de los fármacos , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Modelos Biológicos
16.
Sci Rep ; 8(1): 13275, 2018 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-30185803

RESUMEN

The airway epithelium regulates responses to aeroallergens, acting as a physical and immunological barrier. In asthma, epithelial barrier function and the expression of adherens junction protein E-cadherin is compromised, but it is unknown whether this is cause or consequence of the disease. We hypothesized that airway epithelial loss of E-cadherin is a critical step in the development of manifestations of asthma. We generated a transgenic mouse model with conditional loss of E-cadherin in lung epithelial cells at birth and onwards. We observed normal lung development at the time of birth in mice lacking E-cadherin in the lung epithelium. However, E-cadherin deficiency led to progressive epithelial damage in mice growing into adulthood, as evidenced by airway epithelial denudation, decreased zonula occludens (ZO)-1 expression, loss of ciliated cells, and enlarged alveolar spaces. In addition, spontaneous goblet cell metaplasia with mucus production was observed. These epithelial changes were accompanied by elevated levels of the epithelial-derived chemokine CCL17, infiltration of eosinophils and dendritic cells, and mucus production. In conclusion, loss of E-cadherin induces features in the lung reminiscent of those observed in asthma, indicating that the disruption of E-cadherin-mediated cell-cell contacts may play a key role in the development of asthma manifestations.


Asunto(s)
Cadherinas/metabolismo , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Uniones Adherentes/metabolismo , Animales , Asma/metabolismo , Cadherinas/genética , Cadherinas/fisiología , Quimiocina CCL17/metabolismo , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Eosinófilos/metabolismo , Células Epiteliales/metabolismo , Epitelio/metabolismo , Células Caliciformes/metabolismo , Pulmón/patología , Metaplasia/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Uniones Estrechas/metabolismo , Proteína de la Zonula Occludens-1/metabolismo
17.
Pharmacol Ther ; 112(2): 489-500, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16814862

RESUMEN

T helper (Th) 2 lymphocytes play a crucial role in the initiation, progression and persistence of allergic diseases, including asthma. Drugs that interfere with the activation of T-cells or more selectively Th2-specific signaling molecules and drugs that prevent the selective migration into lung tissue are promising novel strategies for the treatment of allergic asthma. Although the mainstay asthma therapy of inhaled glucocorticoids is rather effective, targeting Th2 cells may be an important alternative in childhood. Regulatory T-cells (Treg cells) have a physiological role in protection of unwanted immune responses to auto-antigens and allergens. Literature data indicate that an imbalance between Th2 and Treg cells may underlie development and disease expression of allergic asthma. Drugs or immunotherapies that stimulate these counter-Treg cells may limit aberrant Th2 responses leading to suppression of symptoms. Furthermore, these types of treatments may offer the perspective of disease modification and long-term relief of symptoms.


Asunto(s)
Asma/tratamiento farmacológico , Hipersensibilidad/tratamiento farmacológico , Linfocitos T/inmunología , Asma/inmunología , AMP Cíclico , Glucocorticoides/uso terapéutico , Humanos , Hipersensibilidad/inmunología , Factores Inmunológicos/uso terapéutico , Inflamación , Transducción de Señal , Linfocitos T/efectos de los fármacos
18.
Clin Epigenetics ; 9: 42, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28450970

RESUMEN

BACKGROUND: Goblet cell metaplasia, a common feature of chronic obstructive pulmonary disease (COPD), is associated with mucus hypersecretion which contributes to the morbidity and mortality among patients. Transcription factors SAM-pointed domain-containing Ets-like factor (SPDEF) and forkhead box protein A2 (FOXA2) regulate goblet cell differentiation. This study aimed to (1) investigate DNA methylation and expression of SPDEF and FOXA2 during goblet cell differentiation and (2) compare this in airway epithelial cells from patients with COPD and controls during mucociliary differentiation. METHODS: To assess DNA methylation and expression of SPDEF and FOXA2 during goblet cell differentiation, primary airway epithelial cells, isolated from trachea (non-COPD controls) and bronchial tissue (patients with COPD), were differentiated by culture at the air-liquid interface (ALI) in the presence of cytokine interleukin (IL)-13 to promote goblet cell differentiation. RESULTS: We found that SPDEF expression was induced during goblet cell differentiation, while FOXA2 expression was decreased. Importantly, CpG number 8 in the SPDEF promoter was hypermethylated upon differentiation, whereas DNA methylation of FOXA2 promoter was not changed. In the absence of IL-13, COPD-derived ALI-cultured cells displayed higher SPDEF expression than control-derived ALI cultures, whereas no difference was found for FOXA2 expression. This was accompanied with hypomethylation of CpG number 6 in the SPDEF promoter and also hypomethylation of CpG numbers 10 and 11 in the FOXA2 promoter. CONCLUSIONS: These findings suggest that aberrant DNA methylation of SPDEF and FOXA2 is one of the factors underlying mucus hypersecretion in COPD, opening new avenues for epigenetic-based inhibition of mucus hypersecretion.


Asunto(s)
Bronquios/citología , Metilación de ADN , Factor Nuclear 3-beta del Hepatocito/genética , Proteínas Proto-Oncogénicas c-ets/genética , Enfermedad Pulmonar Obstructiva Crónica/genética , Tráquea/citología , Bronquios/efectos de los fármacos , Diferenciación Celular , Células Cultivadas , Islas de CpG , Células Epiteliales/citología , Femenino , Regulación de la Expresión Génica , Células Caliciformes/citología , Humanos , Interleucina-13/farmacología , Masculino , Persona de Mediana Edad , Regiones Promotoras Genéticas , Tráquea/efectos de los fármacos
19.
Mucosal Immunol ; 7(2): 215-26, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24150257

RESUMEN

Chronic obstructive pulmonary disease (COPD), a progressive lung disease characterized by sustained neutrophilic airway inflammation, is caused by chronic exposure to noxious stimuli, e.g., cigarette smoke. This chronic exposure can induce immunogenic cell death of structural airway cells, inducing the release of damage-associated molecular patterns (DAMPs). Levels of several DAMPs, including S100 proteins, defensins, and high-mobility group box-1 (HMGB1), are increased in extracellular lung fluids of COPD patients. As DAMPs can attract and activate immune cells upon binding to pattern recognition receptors, we propose that their release may contribute to neutrophilic airway inflammation. In this review, we discuss the novel role of DAMPs in COPD pathogenesis. Relevant DAMPs are categorized based on their subcellular origin, i.e. cytoplasm, endoplasmic reticulum, nucleus, and mitochondria. Furthermore, their potential role in the pathophysiology of COPD will be discussed.


Asunto(s)
Inmunidad Adaptativa , Proteínas Portadoras/metabolismo , Inmunidad Innata , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Animales , Espacio Extracelular , Humanos , Espacio Intracelular , Unión Proteica , Transducción de Señal
20.
Clin Exp Allergy ; 34(9): 1356-63, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15347367

RESUMEN

BACKGROUND: Airway inflammation in asthma is orchestrated by recruitment of T helper (Th)2 lymphocytes to the lung and subsequent production of Th2-like cytokines upon allergen challenge. OBJECTIVE: To examine whether allergen-induced dysfunction of the beta2-adrenergic receptor (beta2-AR) contributes to the enhanced T(h2) cell activity in asthma. METHODS: Beta2-adrenergic regulation of cytokine mRNA expression was studied in alpha-CD3/alpha-CD28-activated peripheral blood lymphocytes from seven asthma patients before and 6 h after allergen challenge, in conjunction with the effects of beta2-agonist fenoterol on T cell chemotaxis and signalling pathways. RESULTS: A complete loss of beta2-AR control over expression of the Th2 cytokines IL-4, IL-5 and IL-13, but not of the Th1 cytokine IFN-gamma, was observed after allergen challenge. Furthermore, we found impaired beta2-AR regulation of T cell migration as well as signal transduction pathways, i.e. the phosphorylation of cyclic adenosine monophosphate-responsive element binding protein and the inhibition of the mitogen-activated protein kinase pathway. The loss of beta2-AR control was associated with increased beta-adrenergic receptor kinase expression, which might be involved in beta2-AR desensitization. In addition, we demonstrate for the first time that T cells exposed to the chemokine thymus and activation-regulated chemokine show hyporesponsiveness to fenoterol. CONCLUSION: Our results suggest that allergen-induced loss of beta2-AR control, possibly mediated by chemokine release, plays an important role in enhanced Th2-like activity in asthma.


Asunto(s)
Alérgenos/inmunología , Asma/inmunología , Receptores Adrenérgicos beta 2/inmunología , Células Th2/inmunología , Adolescente , Agonistas Adrenérgicos beta/inmunología , Adulto , Antígenos CD/inmunología , Células Cultivadas , Quimiotaxis de Leucocito/inmunología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/inmunología , Proteína Quinasa Tipo II Dependiente de AMP Cíclico , Proteínas Quinasas Dependientes de AMP Cíclico/inmunología , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Fenoterol/inmunología , Humanos , Interferón gamma/inmunología , Interleucina-4/inmunología , Interleucina-5/inmunología , Masculino , Persona de Mediana Edad , Fosforilación , ARN Mensajero/análisis , Células TH1/inmunología , Regulación hacia Arriba , Quinasas de Receptores Adrenérgicos beta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA