Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Cells ; 10(11)2021 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-34831082

RESUMEN

COPD is characterized by irreversible lung tissue damage. We hypothesized that lung-derived mesenchymal stromal cells (LMSCs) reduce alveolar epithelial damage via paracrine processes, and may thus be suitable for cell-based strategies in COPD. We aimed to assess whether COPD-derived LMSCs display abnormalities. LMSCs were isolated from lung tissue of severe COPD patients and non-COPD controls. Effects of LMSC conditioned-medium (CM) on H2O2-induced, electric field- and scratch-injury were studied in A549 and NCI-H441 epithelial cells. In organoid models, LMSCs were co-cultured with NCI-H441 or primary lung cells. Organoid number, size and expression of alveolar type II markers were assessed. Pre-treatment with LMSC-CM significantly attenuated oxidative stress-induced necrosis and accelerated wound repair in A549. Co-culture with LMSCs supported organoid formation in NCI-H441 and primary epithelial cells, resulting in significantly larger organoids with lower type II-marker positivity in the presence of COPD-derived versus control LMSCs. Similar abnormalities developed in organoids from COPD compared to control-derived lung cells, with significantly larger organoids. Collectively, this indicates that LMSCs' secretome attenuates alveolar epithelial injury and supports epithelial repair. Additionally, LMSCs promote generation of alveolar organoids, with abnormalities in the supportive effects of COPD-derived LMCS, reflective of impaired regenerative responses of COPD distal lung cells.


Asunto(s)
Células Epiteliales Alveolares/patología , Células Madre Mesenquimatosas/patología , Comunicación Paracrina , Anciano , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Femenino , Humanos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Persona de Mediana Edad , Modelos Biológicos , Organoides/metabolismo , Estrés Oxidativo/efectos de los fármacos , Comunicación Paracrina/efectos de los fármacos , Enfermedad Pulmonar Obstructiva Crónica/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Regeneración/efectos de los fármacos , Esferoides Celulares/patología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
3.
Respir Res ; 18(1): 213, 2017 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-29268739

RESUMEN

BACKGROUND: Nasal gene expression profiling is a promising method to characterize COPD non-invasively. We aimed to identify a nasal gene expression profile to distinguish COPD patients from healthy controls. We investigated whether this COPD-associated gene expression profile in nasal epithelium is comparable with the profile observed in bronchial epithelium. METHODS: Genome wide gene expression analysis was performed on nasal epithelial brushes of 31 severe COPD patients and 22 controls, all current smokers, using Affymetrix Human Gene 1.0 ST Arrays. We repeated the gene expression analysis on bronchial epithelial brushes in 2 independent cohorts of mild-to-moderate COPD patients and controls. RESULTS: In nasal epithelium, 135 genes were significantly differentially expressed between severe COPD patients and controls, 21 being up- and 114 downregulated in COPD (false discovery rate < 0.01). Gene Set Enrichment Analysis (GSEA) showed significant concordant enrichment of COPD-associated nasal and bronchial gene expression in both independent cohorts (FDRGSEA < 0.001). CONCLUSION: We identified a nasal gene expression profile that differentiates severe COPD patients from controls. Of interest, part of the nasal gene expression changes in COPD mimics differentially expressed genes in the bronchus. These findings indicate that nasal gene expression profiling is potentially useful as a non-invasive biomarker in COPD. TRIAL REGISTRATION: ClinicalTrials.gov registration number NCT01351792 (registration date May 10, 2011), ClinicalTrials.gov registration number NCT00848406 (registration date February 19, 2009), ClinicalTrials.gov registration number NCT00807469 (registration date December 11, 2008).


Asunto(s)
Bronquios/metabolismo , Mucosa Nasal/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/diagnóstico , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Adulto , Anciano , Bronquios/patología , Estudios de Cohortes , Femenino , Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Mucosa Nasal/patología , Enfermedad Pulmonar Obstructiva Crónica/genética
5.
Eur Respir J ; 48(2): 504-15, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27126693

RESUMEN

Wingless/integrase-1 (WNT) signalling is associated with lung inflammation and repair, but its role in chronic obstructive pulmonary disease (COPD) pathogenesis is unclear. We investigated whether cigarette smoke-induced dysregulation of WNT-5B contributes to airway remodelling in COPD.We analysed WNT-5B protein expression in the lung tissue of COPD patients and (non)smoking controls, and investigated the effects of cigarette smoke exposure on WNT-5B expression in COPD and control-derived primary bronchial epithelial cells (PBECs). Additionally, we studied downstream effects of WNT-5B on remodelling related genes fibronectin, matrix metalloproteinase (MMP)-2, MMP-9 and SnaiI in BEAS-2B and air-liquid interface (ALI)-cultured PBECs.We observed that airway epithelial WNT-5B expression is significantly higher in lung tissue from COPD patients than controls. Cigarette smoke extract significantly increased mRNA expression of WNT-5B in COPD, but not control-derived PBECs. Exogenously added WNT-5B augmented the expression of remodelling related genes in BEAS-2B cells, which was mediated by transforming growth factor (TGF)-ß/Smad3 signalling. In addition, WNT-5B upregulated the expression of these genes in ALI-cultured PBECs, particularly PBECs from COPD patients.Together, our results provide evidence that exaggerated WNT-5B expression upon cigarette smoke exposure in the bronchial epithelium of COPD patients leads to TGF-ß/Smad3-dependent expression of genes related to airway remodelling.


Asunto(s)
Epitelio/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Humo/efectos adversos , Proteínas Wnt/metabolismo , Anciano , Anciano de 80 o más Años , Bronquios/metabolismo , Estudios de Casos y Controles , Citocinas/metabolismo , Células Epiteliales/metabolismo , Femenino , Fibronectinas , Humanos , Inflamación , Pulmón/metabolismo , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/inducido químicamente , Enfermedad Pulmonar Obstructiva Crónica/patología , Mucosa Respiratoria/citología , Transducción de Señal , Proteína smad3/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo , Nicotiana , Productos de Tabaco , Factor de Crecimiento Transformador beta/metabolismo
6.
Am J Physiol Lung Cell Mol Physiol ; 309(10): L1112-23, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26320152

RESUMEN

In chronic obstructive pulmonary disease (COPD), oxidative stress regulates the inflammatory response of bronchial epithelium and monocytes/macrophages through kinase modulation and has been linked to glucocorticoid unresponsiveness. Glycogen synthase-3ß (GSK3ß) inactivation plays a key role in mediating signaling processes upon reactive oxygen species (ROS) exposure. We hypothesized that GSK3ß is involved in oxidative stress-induced glucocorticoid insensitivity in COPD. We studied levels of phospho-GSK3ß-Ser9, a marker of GSK3ß inactivation, in lung sections and cultured monocytes and bronchial epithelial cells of COPD patients, control smokers, and nonsmokers. We observed increased levels of phospho-GSK3ß-Ser9 in monocytes, alveolar macrophages, and bronchial epithelial cells from COPD patients and control smokers compared with nonsmokers. Pharmacological inactivation of GSK3ß did not affect CXCL8 or granulocyte-macrophage colony-stimulating factor (GM-CSF) expression but resulted in glucocorticoid insensitivity in vitro in both inflammatory and structural cells. Further mechanistic studies in monocyte and bronchial epithelial cell lines showed that GSK3ß inactivation is a common effector of oxidative stress-induced activation of the MEK/ERK-1/2 and phosphatidylinositol 3-kinase/Akt signaling pathways leading to glucocorticoid unresponsiveness. In primary monocytes, the mechanism involved modulation of histone deacetylase 2 (HDAC2) activity in response to GSK3ß inactivation. In conclusion, we demonstrate for the first time that ROS-induced glucocorticoid unresponsiveness in COPD is mediated through GSK3ß, acting as a ROS-sensitive hub.


Asunto(s)
Dexametasona/farmacología , Glucocorticoides/farmacología , Glucógeno Sintasa Quinasa 3/fisiología , Enfermedad Pulmonar Obstructiva Crónica/enzimología , Anciano , Células Cultivadas , Dexametasona/uso terapéutico , Femenino , Expresión Génica/efectos de los fármacos , Glucocorticoides/uso terapéutico , Glucógeno Sintasa Quinasa 3 beta , Histona Desacetilasa 2/metabolismo , Humanos , Leucocitos Mononucleares/metabolismo , Macrófagos Alveolares/enzimología , Masculino , Persona de Mediana Edad , Estrés Oxidativo , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , Mucosa Respiratoria/enzimología , Transducción de Señal
7.
Am J Respir Cell Mol Biol ; 52(5): 554-62, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25192219

RESUMEN

Cigarette smoking, the major causative factor for the development of chronic obstructive pulmonary disease, is associated with neutrophilic airway inflammation. Cigarette smoke (CS) exposure can induce a switch from apoptotic to necrotic cell death in airway epithelium. Therefore, we hypothesized that CS promotes neutrophil necrosis with subsequent release of damage-associated molecular patterns (DAMPs), including high mobility group box 1 (HMGB1), alarming the innate immune system. We studied the effect of smoking two cigarettes on sputum neutrophils in healthy individuals and of 5-day CS or air exposure on neutrophil counts, myeloperoxidase, and HMGB1 levels in bronchoalveolar lavage fluid of BALB/c mice. In human peripheral blood neutrophils, mitochondrial membrane potential, apoptosis/necrosis markers, caspase activity, and DAMP release were studied after CS exposure. Finally, we assessed the effect of neutrophil-derived supernatants on the release of chemoattractant CXCL8 in normal human bronchial epithelial cells. Cigarette smoking caused a significant decrease in sputum neutrophil numbers after 3 hours. In mice, neutrophil counts were significantly increased 16 hours after repeated CS exposure but reduced 2 hours after an additional exposure. In vitro, CS induced necrotic neutrophil cell death, as indicated by mitochondrial dysfunction, inhibition of apoptosis, and DAMP release. Supernatants from CS-treated neutrophils significantly increased the release of CXCL8 in normal human bronchial epithelial cells. Together, these observations show, for the first time, that CS exposure induces neutrophil necrosis, leading to DAMP release, which may amplify CS-induced airway inflammation by promoting airway epithelial proinflammatory responses.


Asunto(s)
Mediadores de Inflamación/metabolismo , Pulmón/metabolismo , Neutrófilos/metabolismo , Neumonía/etiología , Humo/efectos adversos , Fumar/efectos adversos , Animales , Apoptosis , Líquido del Lavado Bronquioalveolar/inmunología , Células Cultivadas , Estudios Cruzados , Femenino , Proteína HMGB1/metabolismo , Humanos , Inmunidad Innata , Mediadores de Inflamación/inmunología , Exposición por Inhalación/efectos adversos , Interleucina-8/metabolismo , Pulmón/inmunología , Pulmón/patología , Potencial de la Membrana Mitocondrial , Ratones Endogámicos C57BL , Necrosis , Neutrófilos/inmunología , Peroxidasa/metabolismo , Fenotipo , Neumonía/inmunología , Neumonía/metabolismo , Neumonía/patología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Transducción de Señal , Fumar/inmunología , Fumar/metabolismo , Esputo/inmunología , Esputo/metabolismo , Factores de Tiempo
8.
PLoS One ; 9(3): e91206, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24651123

RESUMEN

Aeroallergens such as house dust mite (HDM), cockroach, and grass or tree pollen are innocuous substances that can induce allergic sensitization upon inhalation. The serine proteases present in these allergens are thought to activate the protease-activated receptor (PAR)-2, on the airway epithelium, thereby potentially inducing allergic sensitization at the expense of inhalation tolerance. We hypothesized that the proteolytic activity of allergens may play an important factor in the allergenicity to house dust mite and is essential to overcome airway tolerance. Here, we aimed to investigate the role of PAR-2 activation in allergic sensitization and HDM-induced allergic airway inflammation. In our study, Par-2 deficient mice were treated with two different HDM extracts containing high and low serine protease activities twice a week for a period of 5 weeks. We determined airway inflammation through quantification of percentages of mononuclear cells, eosinophils and neutrophils in the bronchial alveolar lavage fluid and measured total IgE and HDM-specific IgE and IgG1 levels in serum. Furthermore, Th2 and pro-inflammatory cytokines including IL-5, IL-13, Eotaxin-1, IL-17, KC, Chemokine (C-C motif) ligand 17 (CCL17) and thymic stromal lymphopoietin (TSLP), were measured in lung tissue homogenates. We observed that independent of the serine protease content, HDM was able to induce elevated levels of eosinophils and neutrophils in the airways of both wild-type (WT) and Par-2 deficient mice. Furthermore, we show that induction of pro-inflammatory cytokines by HDM exposure is independent of Par-2 activation. In contrast, serine protease activity of HDM does contribute to enhanced levels of total IgE, but not HDM-specific IgE. We conclude that, while Par-2 activation contributes to the development of IgE responses, it is largely dispensable for the HDM-induced induction of pro-inflammatory cytokines and airway inflammation in an experimental mouse model of HDM-driven allergic airway disease.


Asunto(s)
Inmunoglobulina E/inmunología , Pyroglyphidae/inmunología , Receptor PAR-2/metabolismo , Animales , Citocinas/biosíntesis , Inmunización , Mediadores de Inflamación/metabolismo , Ratones Endogámicos C57BL , Neumonía/inmunología , Neumonía/parasitología , Neumonía/patología , Receptor PAR-2/deficiencia
9.
Am J Physiol Cell Physiol ; 306(6): C585-97, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24452374

RESUMEN

Airway epithelium, which forms the first barrier towards environmental insults, is disturbed by cigarette smoking, a major risk factor for developing chronic obstructive pulmonary disease (COPD). A-kinase anchoring proteins (AKAP) maintain endothelial barrier function and coordinate subcellular localization of protein kinase A (PKA). However, the role of AKAPs in epithelial barrier function is unknown. We studied the role of AKAPs in regulating human bronchial epithelial (Hogg JC, Timens W. Annu Rev Pathol 4: 435-459, 2009; HBE) barrier. Cigarette smoke extract (CSE) reduced barrier function in 16HBE cells and the expression of the adhesion molecule E-cadherin specifically at the cell membrane. In addition, CSE reduced the protein expression of the AKAP family member AKAP9 at the cell membrane. The expression of AKAP5 and AKAP12 was unaffected by CSE. AKAP9 interacted and colocalized with E-cadherin at the cell membrane, suggesting that the reduction of both proteins may be related. Interestingly, disruption of AKAP-PKA interactions by st-Ht31 prevented the CSE-induced reduction of E-cadherin and AKAP9 protein expression and subsequent loss of barrier function. Silencing of AKAP9 reduced the functional epithelial barrier and prevented the ability of st-Ht31 to restore membrane localization of E-cadherin. Our data suggest the possibility of a specific role for AKAP9 in the maintenance of the epithelial barrier. E-cadherin, but not AKAP9, protein expression was reduced in lung tissue from COPD patients compared with controls. However, AKAP9 mRNA expression was decreased in primary bronchial epithelial cells from current smokers compared with non/ex-smokers. In conclusion, our results indicate that AKAP proteins, most likely AKAP9, maintain the bronchial epithelial barrier by regulating the E-cadherin expression at the cell membrane.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Cadherinas/metabolismo , Proteínas del Citoesqueleto/metabolismo , Nicotiana/efectos adversos , Mucosa Respiratoria/metabolismo , Humo/efectos adversos , Fumar/metabolismo , Proteínas de Anclaje a la Quinasa A/genética , Adulto , Anciano , Anciano de 80 o más Años , Bronquios/efectos de los fármacos , Bronquios/metabolismo , Cadherinas/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Membrana Celular/metabolismo , Proteínas del Citoesqueleto/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Antígenos de Histocompatibilidad Menor , Proteínas Proto-Oncogénicas/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Mucosa Respiratoria/efectos de los fármacos
10.
Respir Res ; 14: 97, 2013 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-24088173

RESUMEN

BACKGROUND: Cigarette smoking is the major risk factor for COPD, leading to chronic airway inflammation. We hypothesized that cigarette smoke induces structural and functional changes of airway epithelial mitochondria, with important implications for lung inflammation and COPD pathogenesis. METHODS: We studied changes in mitochondrial morphology and in expression of markers for mitochondrial capacity, damage/biogenesis and fission/fusion in the human bronchial epithelial cell line BEAS-2B upon 6-months from ex-smoking COPD GOLD stage IV patients to age-matched smoking and never-smoking controls. RESULTS: We observed that long-term CSE exposure induces robust changes in mitochondrial structure, including fragmentation, branching and quantity of cristae. The majority of these changes were persistent upon CSE depletion. Furthermore, long-term CSE exposure significantly increased the expression of specific fission/fusion markers (Fis1, Mfn1, Mfn2, Drp1 and Opa1), oxidative phosphorylation (OXPHOS) proteins (Complex II, III and V), and oxidative stress (Mn-SOD) markers. These changes were accompanied by increased levels of the pro-inflammatory mediators IL-6, IL-8, and IL-1ß. Importantly, COPD primary bronchial epithelial cells (PBECs) displayed similar changes in mitochondrial morphology as observed in long-term CSE-exposure BEAS-2B cells. Moreover, expression of specific OXPHOS proteins was higher in PBECs from COPD patients than control smokers, as was the expression of mitochondrial stress marker PINK1. CONCLUSION: The observed mitochondrial changes in COPD epithelium are potentially the consequence of long-term exposure to cigarette smoke, leading to impaired mitochondrial function and may play a role in the pathogenesis of COPD.


Asunto(s)
Bronquios/patología , Células Epiteliales/patología , Mitocondrias/fisiología , Mitocondrias/ultraestructura , Dinámicas Mitocondriales/fisiología , Recambio Mitocondrial/fisiología , Fumar/efectos adversos , Adulto , Anciano , Bronquios/metabolismo , Estudios de Casos y Controles , Línea Celular , Células Cultivadas , Citocinas/metabolismo , Dinaminas , Células Epiteliales/metabolismo , Femenino , GTP Fosfohidrolasas/metabolismo , Humanos , Técnicas In Vitro , Masculino , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Persona de Mediana Edad , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas Quinasas/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/patología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Factores de Riesgo , Superóxido Dismutasa/metabolismo , Factores de Tiempo
11.
Am J Respir Cell Mol Biol ; 49(4): 662-71, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23742006

RESUMEN

The molecular basis for airway epithelial fragility in asthma has remained unclear. We investigated whether the loss of caveolin-1, the major component of caveolae and a known stabilizer of adherens junctions, contributes to epithelial barrier dysfunction in asthma. We studied the expression of caveolin-1 and adhesion molecules E-cadherin and ß-catenin in airway sections, and we cultured bronchial epithelial cells from patients with asthma and from healthy control subjects. To determine the functional role of caveolin-1, we investigated the effects of caveolin-1 up-regulation and down-regulation on E-cadherin expression, barrier function, and proallergic activity in the human bronchial epithelial cell lines 16HBE and BEAS-2B. The membrane expression of caveolin-1 was significantly lower in airway epithelia from patients with asthma than from subjects without asthma, and this lower expression was maintained in vitro upon air-liquid interface and submerged culturing. Importantly, reduced caveolin-1 expression was accompanied by a loss of junctional E-cadherin and ß-catenin expression, disrupted epithelial barrier function, and increased levels of the proallergic cytokine thymic stromal lymphopoietin (TSLP). Furthermore, E-cadherin redistribution upon exposure to epidermal growth factor or house dust mite was paralleled by the internalization of caveolin-1 in 16HBE cells. These effects appear to be causally related, because the short, interfering RNA down-regulation of caveolin-1 resulted in the delocalization of E-cadherin and barrier dysfunction in 16HBE cells. Moreover, caveolin-1 overexpression improved barrier function and reduced TSLP expression in BEAS-2B cells. Together, our data demonstrate a crucial role for caveolin-1 in epithelial cell-cell adhesion, with important consequences for epithelial barrier function and the promotion of Th2 responses in asthma.


Asunto(s)
Asma/metabolismo , Bronquios/inmunología , Bronquios/metabolismo , Caveolina 1/metabolismo , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Mucosa Respiratoria/metabolismo , Uniones Adherentes/genética , Uniones Adherentes/inmunología , Uniones Adherentes/metabolismo , Adolescente , Adulto , Animales , Asma/genética , Asma/inmunología , Cadherinas/genética , Cadherinas/inmunología , Cadherinas/metabolismo , Caveolina 1/genética , Caveolina 1/inmunología , Adhesión Celular/genética , Adhesión Celular/inmunología , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/inmunología , Moléculas de Adhesión Celular/metabolismo , Niño , Regulación hacia Abajo , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/inmunología , Factor de Crecimiento Epidérmico/metabolismo , Femenino , Humanos , Masculino , Pyroglyphidae/inmunología , Mucosa Respiratoria/inmunología , Células Th2/inmunología , Células Th2/metabolismo , Regulación hacia Arriba , beta Catenina/genética , beta Catenina/inmunología , beta Catenina/metabolismo
12.
Respir Res ; 14: 45, 2013 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-23594194

RESUMEN

BACKGROUND: Cigarette smoking (CS) is the most important risk factor for COPD, which is associated with neutrophilic airway inflammation. We hypothesize, that highly reactive aldehydes are critical for CS-induced neutrophilic airway inflammation. METHODS: BALB/c mice were exposed to CS, water filtered CS (WF-CS) or air for 5 days. Levels of total particulate matter (TPM) and aldehydes in CS and WF-CS were measured. Six hours after the last exposure, inflammatory cells and cytokine levels were measured in lung tissue and bronchoalveolar lavage fluid (BALF). Furthermore, Beas-2b bronchial epithelial cells were exposed to CS extract (CSE) or WF-CS extract (WF-CSE) in the absence or presence of the aldehyde acrolein and IL-8 production was measured after 24 hrs. RESULTS: Compared to CS, in WF-CS strongly decreased (CS; 271.1 ± 41.5 µM, WF-CS; 58.5 ± 8.2 µM) levels of aldehydes were present whereas levels of TPM were only slightly reduced (CS; 20.78 ± 0.59 mg, WF-CS; 16.38 ± 0.36 mg). The numbers of mononuclear cells in BALF (p<0.01) and lung tissue (p<0.01) were significantly increased in the CS- and WF-CS-exposed mice compared to air control mice. Interestingly, the numbers of neutrophils (p<0.001) in BALF and neutrophils and eosinophils (p<0.05) in lung tissue were significantly increased in the CS-exposed but not in WF-CS-exposed mice as compared to air control mice. Levels of the neutrophil and eosinophil chemoattractants KC, MCP-1, MIP-1α and IL-5 were all significantly increased in lung tissue from CS-exposed mice compared to both WF-CS-exposed and air control mice. Interestingly, depletion of aldehydes in WF-CS extract significantly reduced IL-8 production in Beas-2b as compared to CSE, which could be restored by the aldehyde acrolein. CONCLUSION: Aldehydes present in CS play a critical role in inflammatory cytokine production and neutrophilic- but not mononuclear airway inflammation.


Asunto(s)
Aldehídos/toxicidad , Citocinas/inmunología , Activación Neutrófila/inmunología , Neumonía/inducido químicamente , Neumonía/inmunología , Fumar/efectos adversos , Contaminación por Humo de Tabaco/efectos adversos , Animales , Femenino , Ratones , Ratones Endogámicos BALB C , Activación Neutrófila/efectos de los fármacos
13.
Thorax ; 68(8): 709-16, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23370438

RESUMEN

BACKGROUND: WNT signalling is activated during lung tissue damage and inflammation. We investigated whether lung epithelial expression of WNT ligands, receptors (frizzled; FZD) or target genes is dysregulated on cigarette smoking and/or in chronic obstructive pulmonary disease (COPD). METHODS: We studied this in human lung epithelial cell lines and primary bronchial epithelial cells (PBEC) from COPD patients and control (non-)smokers, at baseline and on cigarette smoke extract (CSE) exposure. RESULTS: CSE significantly decreased WNT-4, WNT-10B and FZD2 and increased WNT-5B mRNA expression in 16HBE, but did not affect WNT-4 protein. The mRNA expression of WNT-4, but not other WNT ligands, was lower in PBEC from smokers than non-smokers and downregulated by CSE in PBEC from all groups, yet higher in PBEC from COPD patients than control smokers. Moreover, PBEC from COPD patients displayed higher WNT-4 protein expression than both smokers and non-smokers. Exogenously added WNT-4 significantly increased CXCL8/IL-8, IL-6, CCL5/RANTES, CCL2/MCP-1 and vascular endothelial growth factor (VEGF) secretion in 16HBE, but did not affect the canonical WNT target genes MMP-2, MMP-9, fibronectin, ß-catenin, Dickkopf and axin-2, and induced activation of the non-canonical signalling molecule p38. Moreover, WNT-4 potentiated the CSE-induced upregulation of IL-8 and VEGF. CONCLUSIONS: WNT-4 mRNA and protein levels are higher in PBEC from COPD patients than control (non-)smokers, while cigarette smoke downregulates airway epithelial WNT-4 mRNA, but not protein expression. As WNT-4 further increases CSE-induced pro-inflammatory cytokine release in bronchial epithelium, we propose that higher epithelial WNT-4 levels in combination with cigarette smoking may have important implications for the development of airway inflammation in COPD.


Asunto(s)
Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Enfermedad Pulmonar Obstructiva Crónica/genética , ARN Mensajero/genética , Mucosa Respiratoria/metabolismo , Fumar/efectos adversos , Proteínas Wnt/genética , Adulto , Anciano , Línea Celular , Células Epiteliales/patología , Femenino , Receptores Frizzled/biosíntesis , Receptores Frizzled/genética , Humanos , Masculino , Persona de Mediana Edad , Proteínas Proto-Oncogénicas/biosíntesis , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/patología , Mucosa Respiratoria/patología , Transducción de Señal , Proteínas Wnt/biosíntesis , Proteína Wnt4/biosíntesis , Proteína Wnt4/genética
14.
J Virol ; 86(12): 6595-604, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22514342

RESUMEN

Pellino-1 has recently been identified as a regulator of interleukin-1 (IL-1) signaling, but its roles in regulation of responses of human cells to human pathogens are unknown. We investigated the potential roles of Pellino-1 in the airways. We show for the first time that Pellino-1 regulates responses to a human pathogen, rhinovirus minor group serotype 1B (RV-1B). Knockdown of Pellino-1 by small interfering RNA (siRNA) was associated with impaired production of innate immune cytokines such as CXCL8 from human primary bronchial epithelial cells in response to RV-1B, without impairment in production of antiviral interferons (IFN), and without loss of control of viral replication. Pellino-1 actions were likely to be independent of interleukin-1 receptor-associated kinase-1 (IRAK-1) regulation, since Pellino-1 knockdown in primary epithelial cells did not alter responses to IL-1 but did inhibit responses to poly(I·C), a Toll-like receptor 3 (TLR3) activator that does not signal via IRAK-1 to engender a response. These data indicate that Pellino-1 represents a novel target that regulates responses of human airways to human viral pathogens, independently of IRAK signaling. Neutralization of Pellino-1 may therefore provide opportunities to inhibit potentially harmful neutrophilic inflammation of the airways induced by respiratory viruses, without loss of control of the underlying viral infection.


Asunto(s)
Células Epiteliales/inmunología , Proteínas Nucleares/inmunología , Infecciones por Picornaviridae/inmunología , Rhinovirus/fisiología , Ubiquitina-Proteína Ligasas/inmunología , Adolescente , Adulto , Anciano , Línea Celular , Células Cultivadas , Células Epiteliales/virología , Femenino , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/genética , Quinasas Asociadas a Receptores de Interleucina-1/inmunología , Masculino , Persona de Mediana Edad , Proteínas Nucleares/genética , Infecciones por Picornaviridae/genética , Infecciones por Picornaviridae/virología , Rhinovirus/genética , Rhinovirus/inmunología , Transducción de Señal , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/inmunología , Ubiquitina-Proteína Ligasas/genética , Adulto Joven
15.
Am J Physiol Lung Cell Mol Physiol ; 293(5): L1156-62, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17720877

RESUMEN

In patients with chronic obstructive pulmonary disease (COPD), an imbalance between oxidants and antioxidants is acknowledged to result in disease development and progression. Cigarette smoke (CS) is known to deplete total glutathione (GSH + GSSG) in the airways. We hypothesized that components in the gaseous phase of CS may irreversibly react with GSH to form GSH derivatives that cannot be reduced (GSX), thereby causing this depletion. To understand this phenomenon, we investigated the effect of CS on GSH metabolism and identified the actual GSX compounds. CS and H(2)O(2) (control) deplete reduced GSH in solution [Delta = -54.1 +/- 1.7 microM (P < 0.01) and -39.8 +/- 0.9 microM (P < 0.01), respectively]. However, a significant decrease of GSH + GSSG was observed after CS (Delta = -75.1 +/- 7.6 microM, P < 0.01), but not after H(2)O(2). Exposure of A549 cells and primary bronchial epithelial cells to CS decreased free sulfhydryl (-SH) groups (Delta = -64.2 +/- 14.6 microM/mg protein, P < 0.05) and irreversibly modified GSH + GSSG (Delta = -17.7 +/- 1.9 microM, P < 0.01) compared with nonexposed cells or H(2)O(2) control. Mass spectrometry (MS) showed that GSH was modified to glutathione-aldehyde derivatives. Further MS identification showed that GSH was bound to acrolein and crotonaldehyde and another, yet to be identified, structure. Our data show that CS does not oxidize GSH to GSSG but, rather, reacts to nonreducible glutathione-aldehyde derivatives, thereby depleting the total available GSH pool.


Asunto(s)
Glutatión/metabolismo , Pulmón/metabolismo , Mucosa Respiratoria/metabolismo , Fumar/metabolismo , Bronquios/citología , Células Cultivadas/metabolismo , Disulfuro de Glutatión/metabolismo , Humanos , Espectrometría de Masas , Oxidantes/farmacología , Oxidación-Reducción
16.
Am J Physiol Lung Cell Mol Physiol ; 292(5): L1211-8, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17209140

RESUMEN

Increased lung cell apoptosis and necrosis occur in patients with chronic obstructive pulmonary disease (COPD). Mitochondria are crucially involved in the regulation of these cell death processes. Cigarette smoke is the main risk factor for development of COPD. We hypothesized that cigarette smoke disturbs mitochondrial function, thereby decreasing the capacity of mitochondria for ATP synthesis, leading to cellular necrosis. This hypothesis was tested in both human bronchial epithelial cells and isolated mitochondria. Cigarette smoke extract exposure resulted in a dose-dependent inhibition of complex I and II activities. This inhibition was accompanied by decreases in mitochondrial membrane potential, mitochondrial oxygen consumption, and production of ATP. Cigarette smoke extract abolished the staurosporin-induced caspase-3 and -7 activities and induced a switch from epithelial cell apoptosis into necrosis. Cigarette smoke induced mitochondrial dysfunction, with compounds of cigarette smoke acting as blocking agents of the mitochondrial respiratory chain; loss of ATP generation leading to cellular necrosis instead of apoptosis is a new pathophysiological concept of COPD development.


Asunto(s)
Mitocondrias/metabolismo , Dilatación Mitocondrial/fisiología , Consumo de Oxígeno/fisiología , Enfermedad Pulmonar Obstructiva Crónica/patología , Mucosa Respiratoria/citología , Mucosa Respiratoria/patología , Mucosa Respiratoria/fisiología , Humo/efectos adversos , Fumar/efectos adversos , Células Cultivadas , Humanos , Cinética , Necrosis , Enfermedad Pulmonar Obstructiva Crónica/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA