Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
J Med Microbiol ; 70(10)2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34596013

RESUMEN

Introduction. Pseudomonas aeruginosa produces quorum sensing signalling molecules including 2-alkyl-4-quinolones (AQs), which regulate virulence factor production in the cystic fibrosis (CF) airways.Hypothesis/Gap statement. Culture can lead to condition-dependent artefacts which may limit the potential insights and applications of AQs as minimally-invasive biomarkers of bacterial load.Aim. We aimed to use culture-independent methods to explore the correlations between AQ levels and live P. aeruginosa load in adults with CF.Methodology. Seventy-five sputum samples at clinical stability and 48 paired sputum samples obtained at the beginning and end of IV antibiotics for a pulmonary exacerbation in adults with CF were processed using a viable cell separation technique followed by quantitative P. aeruginosa polymerase chain reaction (qPCR). Live P. aeruginosa qPCR load was compared with the concentrations of three AQs (HHQ, NHQ and HQNO) detected in sputum, plasma and urine.Results. At clinical stability and the beginning of IV antibiotics for pulmonary exacerbation, HHQ, NHQ and HQNO measured in sputum, plasma and urine were consistently positively correlated with live P. aeruginosa qPCR load in sputum, compared to culture. Following systemic antibiotics live P. aeruginosa qPCR load decreased significantly (P<0.001) and was correlated with a reduction in plasma NHQ (plasma: r=0.463, P=0.003).Conclusion. In adults with CF, AQ concentrations correlated more strongly with live P. aeruginosa bacterial load measured by qPCR compared to traditional culture. Prospective studies are required to assess the potential of systemic AQs as biomarkers of P. aeruginosa bacterial burden.


Asunto(s)
4-Quinolonas/aislamiento & purificación , Fibrosis Quística/complicaciones , Infecciones por Pseudomonas/complicaciones , Pseudomonas aeruginosa/aislamiento & purificación , Percepción de Quorum , 4-Quinolonas/sangre , 4-Quinolonas/orina , Adolescente , Adulto , Carga Bacteriana , Biomarcadores , Fibrosis Quística/microbiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Infecciones por Pseudomonas/microbiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Esputo/química , Adulto Joven
2.
Clin Epigenetics ; 12(1): 145, 2020 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-33008450

RESUMEN

BACKGROUND: Mesenchymal fibroblasts are ubiquitous cells that maintain the extracellular matrix of organs. Within the lung, airway and parenchymal fibroblasts are crucial for lung development and are altered with disease, but it has been difficult to understand their roles due to the lack of distinct molecular markers. We studied genome-wide DNA methylation and gene expression in airway and parenchymal lung fibroblasts from healthy and asthmatic donors, to identify a robust cell marker and to determine if these cells are molecularly distinct in asthma. RESULTS: Airway (N = 8) and parenchymal (N = 15) lung fibroblasts from healthy individuals differed in the expression of 158 genes, and DNA methylation of 3936 CpGs (Bonferroni adjusted p value < 0.05). Differential DNA methylation between cell types was associated with differential expression of 42 genes, but no single DNA methylation CpG feature (location, effect size, number) defined the interaction. Replication of gene expression and DNA methylation in a second cohort identified TWIST1 gene expression, DNA methylation and protein expression as a cell marker of airway and parenchymal lung fibroblasts, with DNA methylation having 100% predictive discriminatory power. DNA methylation was differentially altered in parenchymal (112 regions) and airway fibroblasts (17 regions) with asthmatic status, with no overlap between regions. CONCLUSIONS: Differential methylation of TWIST1 is a robust cell marker of airway and parenchymal lung fibroblasts. Airway and parenchymal fibroblast DNA methylation are differentially altered in individuals with asthma, and the role of both cell types should be considered in the pathogenesis of asthma.


Asunto(s)
Asma/genética , Metilación de ADN/genética , Fibroblastos/metabolismo , Proteínas Nucleares/metabolismo , Tejido Parenquimatoso/citología , Proteína 1 Relacionada con Twist/metabolismo , Anciano , Remodelación de las Vías Aéreas (Respiratorias)/genética , Asma/patología , Biomarcadores/metabolismo , Estudios de Casos y Controles , Islas de CpG/genética , Femenino , Expresión Génica , Estudio de Asociación del Genoma Completo/métodos , Humanos , Pulmón/patología , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas
3.
Sci Rep ; 9(1): 14409, 2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31595000

RESUMEN

The airway epithelium forms the interface between the inhaled environment and the lung. The airway epithelium is dysfunctional in asthma and epigenetic mechanisms are considered a contributory factor. We hypothesised that the DNA methylation profiles of cultured primary airway epithelial cells (AECs) would differ between cells isolated from individuals with asthma (n = 17) versus those without asthma (n = 16). AECs were isolated from patients by two different isolation techniques; pronase digestion (9 non-asthmatic, 8 asthmatic) and bronchial brushings (7 non-asthmatic and 9 asthmatic). DNA methylation was assessed using an Illumina Infinium HumanMethylation450 BeadChip array. DNA methylation of AECs clustered by isolation technique and linear regression identified 111 CpG sites differentially methylated between isolation techniques in healthy individuals. As a consequence, the effect of asthmatic status on DNA methylation was assessed within AEC samples isolated using the same technique. In pronase isolated AECs, 15 DNA regions were differentially methylated between asthmatics and non-asthmatics. In bronchial brush isolated AECs, 849 differentially methylated DNA regions were identified with no overlap to pronase regions. In conclusion, regardless of cell isolation technique, differential DNA methylation was associated with asthmatic status in AECs, providing further evidence for aberrant DNA methylation as a signature of epithelial dysfunction in asthma.


Asunto(s)
Asma/genética , Metilación de ADN/genética , Epigénesis Genética , Pulmón/metabolismo , Adulto , Asma/patología , Bronquios/metabolismo , Bronquios/patología , Células Cultivadas , Islas de CpG/genética , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Regulación de la Expresión Génica/genética , Humanos , Pulmón/patología , Masculino , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología
4.
Chest ; 155(4): 816-824, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30414795

RESUMEN

Epigenetic changes are heritable changes in gene expression, without changing the DNA sequence. Epigenetic processes provide a critical link between environmental insults to the airway and functional changes that determine how airway cells respond to future stimuli. There are three primary epigenetic processes: histone modifications, DNA modification, and noncoding RNAs. Airway smooth muscle has several important roles in the development and maintenance of the pathologic processes occurring in asthma, including inflammation, remodeling, and contraction/hyperresponsiveness. In this review, we describe the evidence for the role of epigenetic changes in driving these processes in airway smooth muscle cells in asthma, with a particular focus on histone modifications. We also discuss how existing therapies may target some of these changes and how epigenetic processes provide targets for the development of novel asthma therapeutics. Epigenetic marks may also provide a biomarker to assess phenotype and treatment responses.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/fisiología , Asma/fisiopatología , Epigénesis Genética , Asma/genética , Humanos , Músculo Liso/fisiopatología
5.
Biochim Biophys Acta Gene Regul Mech ; 1861(5): 463-472, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29555582

RESUMEN

Cyclooxygenase-2 (COX-2), with its main antifibrotic metabolite PGE2, is regarded as an antifibrotic gene. Repressed COX-2 expression and deficient PGE2 have been shown to contribute to the activation of lung fibroblasts and excessive deposition of collagen in pulmonary fibrosis. We have previously demonstrated that COX-2 expression in lung fibroblasts from patients with idiopathic pulmonary fibrosis (IPF) is epigenetically silenced and can be restored by epigenetic inhibitors. This study aimed to investigate whether COX-2 downregulation induced by the profibrotic cytokine transforming growth factor-ß1 (TGF-ß1) in normal lung fibroblasts could be prevented by epigenetic inhibitors. We found that COX-2 protein expression and PGE2 production were markedly reduced by TGF-ß1 and this was prevented by the pan-histone deacetylase inhibitor suberanilohydroxamic acid (SAHA) and to a lesser extent by the DNA demethylating agent Decitabine (DAC), but not by the G9a histone methyltransferase (HMT) inhibitor BIX01294 or the EZH2 HMT inhibitor 3-deazaneplanocin A (DZNep). However, chromatin immunoprecipitation assay revealed that the effect of SAHA was unlikely mediated by histone modifications. Instead 3'-untranslated region (3'-UTR) luciferase reporter assay indicated the involvement of post-transcriptional mechanisms. This was supported by the downregulation by SAHA of the 3'-UTR mRNA binding protein TIA-1 (T-cell intracellular antigen-1), a negative regulator of COX-2 translation. Furthermore, TIA-1 knockdown by siRNA mimicked the effect of SAHA on COX-2 expression. These findings suggest SAHA can prevent TGF-ß1-induced COX-2 repression in lung fibroblasts post-transcriptionally through a novel TIA-1-dependent mechanism and provide new insights into the mechanisms underlying its potential antifibrotic activity.


Asunto(s)
Ciclooxigenasa 2/genética , Inhibidores de Histona Desacetilasas/administración & dosificación , Antígeno Intracelular 1 de las Células T/genética , Factor de Crecimiento Transformador beta1/genética , Adenosina/administración & dosificación , Adenosina/análogos & derivados , Azacitidina/administración & dosificación , Azacitidina/análogos & derivados , Línea Celular , Ciclooxigenasa 1/genética , Metilación de ADN/genética , Decitabina , Proteína Potenciadora del Homólogo Zeste 2/genética , Fibroblastos/metabolismo , Regulación de la Expresión Génica/genética , Humanos , Ácidos Hidroxámicos/administración & dosificación , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Regiones Promotoras Genéticas , Vorinostat
6.
Clin Epigenetics ; 10: 32, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29527240

RESUMEN

Background: Chronic obstructive pulmonary disease (COPD) is a heterogeneous disease of the lungs that is currently the fourth leading cause of death worldwide. Genetic factors account for only a small amount of COPD risk, but epigenetic mechanisms, including DNA methylation, have the potential to mediate the interactions between an individual's genetics and environmental exposure. DNA methylation is highly cell type-specific, and individual cell type studies of DNA methylation in COPD are sparse. Fibroblasts are present within the airway and parenchyma of the lung and contribute to the aberrant deposition of extracellular matrix in COPD. No assessment or comparison of genome-wide DNA methylation profiles in the airway and parenchymal fibroblasts from individuals with and without COPD has been undertaken. These data provide valuable insight into the molecular mechanisms contributing to COPD and the differing pathologies of small airways disease and emphysema in COPD. Methods: Genome-wide DNA methylation was evaluated at over 485,000 CpG sites using the Illumina Infinium HumanMethylation450 BeadChip array in the airway (non-COPD n = 8, COPD n = 7) and parenchymal fibroblasts (non-COPD n = 17, COPD n = 29) isolated from individuals with and without COPD. Targeted gene expression was assessed by qPCR in matched RNA samples. Results: Differentially methylated DNA regions were identified between cells isolated from individuals with and without COPD in both airway and parenchymal fibroblasts. Only in parenchymal fibroblasts was differential DNA methylation associated with differential gene expression. A second analysis of differential DNA methylation variability identified 359 individual differentially variable CpG sites in parenchymal fibroblasts. No differentially variable CpG sites were identified in the airway fibroblasts. Five differentially variable-methylated CpG sites, associated with three genes, were subsequently assessed for gene expression differences. Two genes (OAT and GRIK2) displayed significantly increased gene expression in cells isolated from individuals with COPD. Conclusions: Differential and variable DNA methylation was associated with COPD status in the parenchymal fibroblasts but not airway fibroblasts. Aberrant DNA methylation was associated with altered gene expression imparting biological function to DNA methylation changes. Changes in DNA methylation are therefore implicated in the molecular mechanisms underlying COPD pathogenesis and may represent novel therapeutic targets.


Asunto(s)
Metilación de ADN , Pulmón/química , Ornitina-Oxo-Ácido Transaminasa/genética , Tejido Parenquimatoso/química , Enfermedad Pulmonar Obstructiva Crónica/genética , Receptores de Ácido Kaínico/genética , Regulación hacia Arriba , Anciano , Células Cultivadas , Islas de CpG , Epigénesis Genética , Femenino , Fibroblastos/química , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Humanos , Pulmón/citología , Masculino , Persona de Mediana Edad , Especificidad de Órganos , Tejido Parenquimatoso/citología , Análisis de Secuencia de ADN , Receptor de Ácido Kaínico GluK2
7.
J Immunol ; 200(4): 1283-1294, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29305433

RESUMEN

Airway smooth muscle (ASM) cells contribute to asthmatic lung pathology with chemokine hypersecretion and increased ASM cell mass. With little recent progress in the development of asthma therapies, a greater understanding of lung inflammation mechanisms has become a priority. Chemokine gene expression in ASM cells is dependent upon NF-κB transcription factor activity. The telomerase/shelterin complex maintains chromosomal telomere ends during cell division. Telomerase is a possible cofactor for NF-κB activity, but its role in NF-κB activity in airway tissue inflammation is not known. In this study, we sought to address two key questions: whether telomerase is involved in inflammation in ASM cells, and whether components of the shelterin complex are also required for an inflammatory response in ASM cells. Telomerase inhibitors and telomerase small interfering RNA (siRNA) reduced TNF-α-induced chemokine expression in ASM cells. Telomerase siRNA and inhibitors reduced NF-κB activity. An siRNA screen of shelterin components identified a requirement for PIN2/TERF1 interacting-telomerase inhibitor 1 (PINX1) in chemokine gene expression. High-level PINX1 overexpression reduced NF-κB reporter activity, but low-level expression amplified NF-κB activity. Coimmunoprecipitation studies showed association of PINX1 and p65. Overexpression of the N terminus (2-252 aa) of PINX1, but not the C-terminal telomerase-inhibitor domain (253-328 aa), amplified TNF-α-induced NF-κB activity. GST pull-downs demonstrated that the N terminus of PINX1 bound more p65 than the C-terminal telomerase-inhibitor domain; these observations were confirmed in whole cells with N-terminal and C-terminal PINX1 immunoprecipitation. We conclude that telomerase and PINX1 are required for chemokine expression in ASM cells and represent significant new targets for future anti-inflammatory therapies for lung diseases, such as asthma.


Asunto(s)
Quimiocinas/biosíntesis , Regulación de la Expresión Génica/fisiología , Miocitos del Músculo Liso/inmunología , Telomerasa/biosíntesis , Proteínas Supresoras de Tumor/biosíntesis , Proteínas de Ciclo Celular , Línea Celular , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Miocitos del Músculo Liso/metabolismo , Telomerasa/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Proteínas Supresoras de Tumor/inmunología
8.
Am J Respir Cell Mol Biol ; 58(4): 449-460, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29053336

RESUMEN

Selective repression of the antifibrotic gene CXCL10 contributes to tissue remodeling in idiopathic pulmonary fibrosis (IPF). We have previously reported that histone deacetylation and histone H3 lysine 9 (H3K9) methylation are involved in CXCL10 repression. In this study, we explored the role of H3K27 methylation and the interplay between the two histone lysine methyltransferases enhancer of zest homolog 2 (EZH2) and G9a in CXCL10 repression in IPF. By applying chromatin immunoprecipitation, Re-ChIP, and proximity ligation assays, we demonstrated that, like G9a-mediated H3K9 methylation, EZH2-mediated histone H3 lysine 27 trimethylation (H3K27me3) was significantly enriched at the CXCL10 promoter in fibroblasts from IPF lungs (F-IPF) compared with fibroblasts from nonfibrotic lungs, and we also found that EZH2 and G9a physically interacted with each other. EZH2 knockdown reduced not only EZH2 and H3K27me3 but also G9a and H3K9me3, and G9a knockdown reduced not only G9 and H3K9me3 but also EZH2 and H3K27me3. Depletion and inhibition of EZH2 and G9a also reversed histone deacetylation and restored CXCL10 expression in F-IPF. Furthermore, treatment of fibroblasts from nonfibrotic lungs with the profibrotic cytokine transforming growth factor-ß1 increased EZH2, G9a, H3K27me3, H3K9me3, and histone deacetylation at the CXCL10 promoter, similar to that observed in F-IPF, which was correlated with CXCL10 repression and was prevented by EZH2 and G9a knockdown. These findings suggest that a novel and functionally interdependent interplay between EZH2 and G9a regulates histone methylation-mediated epigenetic repression of the antifibrotic CXCL10 gene in IPF. This interdependent interplay may prove to be a target for epigenetic intervention to restore the expression of CXCL10 and other antifibrotic genes in IPF.


Asunto(s)
Quimiocina CXCL10/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Fibroblastos/enzimología , Antígenos de Histocompatibilidad/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Fibrosis Pulmonar Idiopática/enzimología , Pulmón/enzimología , Estudios de Casos y Controles , Células Cultivadas , Quimiocina CXCL10/genética , Metilación de ADN , Regulación hacia Abajo , Proteína Potenciadora del Homólogo Zeste 2/genética , Represión Epigenética , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Antígenos de Histocompatibilidad/genética , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/patología , Pulmón/efectos de los fármacos , Pulmón/patología , Regiones Promotoras Genéticas , Transducción de Señal , Factor de Crecimiento Transformador beta1/farmacología
9.
Am J Respir Cell Mol Biol ; 56(6): 772-783, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28225294

RESUMEN

Fibrosis is a major cause of progressive organ dysfunction in several chronic pulmonary diseases. Rho-associated coiled-coil forming kinase (ROCK) has been shown to be involved in myofibroblast differentiation driven by altered matrix stiffness in a fibrotic state. There are two known ROCK isoforms in humans, ROCK1 and ROCK2, but the specific role of each isoform in myofibroblast differentiation in lung fibrosis remains unknown. To study this, we developed a gelatin methacryloyl hydrogel-based culture system with different stiffness levels relevant to healthy and fibrotic lungs. We have shown that stiff matrix, but not soft matrix, can induce myofibroblast differentiation with high smooth muscle actin isoform (αSMA) expression. Furthermore, our data confirmed that the inhibition of ROCK signaling by a pharmacological inhibitor (i.e., Y27632) attenuates stiffness-induced αSMA expression and fiber assembly in myofibroblasts. To assess the role of ROCK isoforms in this process, we used short interfering RNA to knock down the expression of each isoform. Our data showed that knocking down either ROCK1 or ROCK2 did not result in a reduction in αSMA expression in myofibroblasts on stiff matrix, as opposed to soft matrix, where αSMA expression was reduced significantly. Paradoxically, on stiff matrix, the absence of one isoform (particularly ROCK2) exaggerated αSMA expression and led to thick fiber assembly. Moreover, complete loss of αSMA fiber assembly was seen only in the absence of both ROCK isoforms, suggesting that both isoforms are implicated in this process. Overall, our results indicate the differential role of ROCK isoforms in myofibroblast differentiation on soft and stiff matrices.


Asunto(s)
Diferenciación Celular , Miofibroblastos/enzimología , Miofibroblastos/patología , Fibrosis Pulmonar/enzimología , Fibrosis Pulmonar/patología , Estrés Mecánico , Quinasas Asociadas a rho/metabolismo , Actinas/metabolismo , Amidas/farmacología , Fenómenos Biomecánicos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Gelatina/farmacología , Silenciador del Gen/efectos de los fármacos , Humanos , Hidrogeles/farmacología , Isoenzimas/metabolismo , Metacrilatos/farmacología , Polimerizacion/efectos de los fármacos , Piridinas/farmacología , Transducción de Señal/efectos de los fármacos , Fibras de Estrés/efectos de los fármacos , Fibras de Estrés/metabolismo , Especificidad por Sustrato/efectos de los fármacos , Andamios del Tejido/química , Transactivadores/metabolismo
10.
J Cyst Fibros ; 16(2): 230-238, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27773591

RESUMEN

BACKGROUND: Pulmonary P. aeruginosa infection is associated with poor outcomes in cystic fibrosis (CF) and early diagnosis is challenging, particularly in those who are unable to expectorate sputum. Specific P. aeruginosa 2-alkyl-4-quinolones are detectable in the sputum, plasma and urine of adults with CF, suggesting that they have potential as biomarkers for P. aeruginosa infection. AIM: To investigate systemic 2-alkyl-4-quinolones as potential biomarkers for pulmonary P. aeruginosa infection. METHODS: A multicentre observational study of 176 adults and 68 children with CF. Cross-sectionally, comparisons were made between current P. aeruginosa infection using six 2-alkyl-4-quinolones detected in sputum, plasma and urine against hospital microbiological culture results. All participants without P. aeruginosa infection at baseline were followed up for one year to determine if 2-alkyl-4-quinolones were early biomarkers of pulmonary P. aeruginosa infection. RESULTS: Cross-sectional analysis: the most promising biomarker with the greatest diagnostic accuracy was 2-heptyl-4-hydroxyquinoline (HHQ). In adults, areas under the ROC curves (95% confidence intervals) for HHQ analyses were 0.82 (0.75-0.89) in sputum, 0.76 (0.69-0.82) in plasma and 0.82 (0.77-0.88) in urine. In children, the corresponding values for HHQ analyses were 0.88 (0.77-0.99) in plasma and 0.83 (0.68-0.97) in urine. Longitudinal analysis: Ten adults and six children had a new positive respiratory culture for P. aeruginosa in follow-up. A positive plasma HHQ test at baseline was significantly associated with a new positive culture for P. aeruginosa in both adults and children in follow-up (odds ratio (OR)=6.67;-95% CI:-1.48-30.1;-p=0.01 and OR=70; 95% CI: 5-956;-p<0.001 respectively). CONCLUSIONS: AQs measured in sputum, plasma and urine may be used to diagnose current infection with P. aeruginosa in adults and children with CF. These preliminary data show that plasma HHQ may have potential as an early biomarker of pulmonary P. aeruginosa. Further studies are necessary to evaluate if HHQ could be used in clinical practice to aid early diagnosis of P. aeruginosa infection in the future.


Asunto(s)
Fibrosis Quística , Infecciones por Pseudomonas , Pseudomonas aeruginosa , Quinolonas , Infecciones del Sistema Respiratorio , Adulto , Biomarcadores/análisis , Biomarcadores/metabolismo , Niño , Estudios Transversales , Fibrosis Quística/diagnóstico por imagen , Fibrosis Quística/microbiología , Diagnóstico Precoz , Femenino , Humanos , Masculino , Valor Predictivo de las Pruebas , Pronóstico , Infecciones por Pseudomonas/diagnóstico , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/aislamiento & purificación , Pseudomonas aeruginosa/fisiología , Quinolonas/análisis , Quinolonas/metabolismo , Reproducibilidad de los Resultados , Infecciones del Sistema Respiratorio/diagnóstico , Infecciones del Sistema Respiratorio/microbiología , Reino Unido
11.
Sci Signal ; 9(451): ra104, 2016 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-27811142

RESUMEN

Heterotrimeric guanine nucleotide-binding protein (G protein) signaling links hundreds of G protein-coupled receptors with four G protein signaling pathways. Two of these, one mediated by Gq and G11 (Gq/11) and the other by G12 and G13 (G12/13), are implicated in the force-dependent activation of transforming growth factor-ß (TGFß) in lung epithelial cells. Reduced TGFß activation in alveolar cells leads to emphysema, whereas enhanced TGFß activation promotes acute lung injury and idiopathic pulmonary fibrosis. Therefore, precise control of alveolar TGFß activation is essential for alveolar homeostasis. We investigated the involvement of the Gq/11 and G12/13 pathways in epithelial cells in generating active TGFß and regulating alveolar inflammation. Mice deficient in both Gαq and Gα11 developed inflammation that was primarily caused by alternatively activated (M2-polarized) macrophages, enhanced matrix metalloproteinase 12 (MMP12) production, and age-related alveolar airspace enlargement consistent with emphysema. Mice with impaired Gq/11 signaling had reduced stretch-mediated generation of TGFß by epithelial cells and enhanced macrophage MMP12 synthesis but were protected from the effects of ventilator-induced lung injury. Furthermore, synthesis of the cytokine interleukin-33 (IL-33) was increased in these alveolar epithelial cells, resulting in the M2-type polarization of alveolar macrophages independently of the effect on TGFß. Our results suggest that alveolar Gq/11 signaling maintains alveolar homeostasis and likely independently increases TGFß activation in response to the mechanical stress of the epithelium and decreases epithelial IL-33 synthesis. Together, these findings suggest that disruption of Gq/11 signaling promotes inflammatory emphysema but protects against mechanically induced lung injury.


Asunto(s)
Enfisema/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Interleucina-33/metabolismo , Macrófagos Alveolares/metabolismo , Mucosa Respiratoria/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Enfisema/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Interleucina-33/genética , Metaloproteinasa 12 de la Matriz/genética , Metaloproteinasa 12 de la Matriz/metabolismo , Ratones , Ratones Transgénicos , Mucosa Respiratoria/patología , Factor de Crecimiento Transformador beta/genética , Lesión Pulmonar Inducida por Ventilación Mecánica/genética , Lesión Pulmonar Inducida por Ventilación Mecánica/metabolismo
12.
Biochem Biophys Res Commun ; 476(4): 431-437, 2016 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-27240956

RESUMEN

Airway epithelial cells in cystic fibrosis (CF) overexpress Interleukin 8 (CXCL8) through poorly defined mechanisms. CXCL8 transcription is dependent on coordinated binding of CCAAT/enhancer binding protein (C/EBP)ß, nuclear factor (NF)-κB, and activator protein (AP)-1 to the promoter. Here we show abnormal epigenetic regulation is responsible for CXCL8 overexpression in CF cells. Under basal conditions CF cells had increased bromodomain (Brd)3 and Brd4 recruitment and enhanced NF-κB and C/EBPß binding to the CXCL8 promoter compared to non-CF cells due to trimethylation of histone H3 at lysine 4 (H3K4me3) and DNA hypomethylation at CpG6. IL-1ß increased NF-κB, C/EBPß and Brd4 binding. Furthermore, inhibitors of bromodomain and extra-terminal domain family (BET) proteins reduced CXCL8 production in CF cells suggesting a therapeutic target for the BET pathway.


Asunto(s)
Fibrosis Quística/genética , Epigénesis Genética , Interleucina-8/genética , Azepinas/farmacología , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteínas de Ciclo Celular , Células Cultivadas , Fibrosis Quística/patología , Metilación de ADN , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Interleucina-1beta/farmacología , Interleucina-8/metabolismo , Mutación , FN-kappa B/genética , FN-kappa B/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Triazoles/farmacología
13.
J Biol Chem ; 291(18): 9540-53, 2016 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-26861876

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic lung disease with high mortality. Active TGFß1 is considered central to the pathogenesis of IPF. A major mechanism of TGFß1 activation in the lung involves the epithelially restricted αvß6 integrin. Expression of the αvß6 integrin is dramatically increased in IPF. How αvß6 integrin expression is regulated in the pulmonary epithelium is unknown. Here we identify a region in the ß6 subunit gene (ITGB6) promoter acting to markedly repress basal gene transcription, which responds to both the Ets domain-containing protein Elk1 (Elk1) and the glucocorticoid receptor (GR). Both Elk1 and GR can regulate αvß6 integrin expression in vitro We demonstrate Elk1 binding to the ITGB6 promoter basally and that manipulation of Elk1 or Elk1 binding alters ITGB6 promoter activity, gene transcription, and αvß6 integrin expression. Crucially, we find that loss of Elk1 causes enhanced Itgb6 expression and exaggerated lung fibrosis in an in vivo model of fibrosis, whereas the GR agonist dexamethasone inhibits Itgb6 expression. Moreover, Elk1 dysregulation is present in epithelium from patients with IPF. These data reveal a novel role for Elk1 regulating ITGB6 expression and highlight how dysregulation of Elk1 can contribute to human disease.


Asunto(s)
Antígenos de Neoplasias/biosíntesis , Regulación de la Expresión Génica , Integrinas/biosíntesis , Fibrosis Pulmonar/metabolismo , Transducción de Señal , Transcripción Genética , Proteína Elk-1 con Dominio ets/metabolismo , Animales , Antígenos de Neoplasias/genética , Línea Celular Transformada , Humanos , Integrinas/genética , Ratones , Ratones Noqueados , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Proteína Elk-1 con Dominio ets/genética
14.
COPD ; 13(1): 26-34, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26418236

RESUMEN

Matrix metalloproteinases (MMPs) are elevated in the airways and blood of COPD patients, contributing to disease pathogenesis and tissue remodelling. However, it is not clear if MMP levels in airways, blood and urine are related or if MMP levels are related to disease severity or presence of exacerbations requiring hospitalisation. Seventy-two patients requiring hospitalisation for COPD exacerbations had serum, urine and sputum MMP-8, -9 and active MMP-9 measured by ELISA and gelatin zymography on day one, five and four weeks later (recovery). Clinical history, spirometry, COPD Assessment Test and MRC dyspnoea score were obtained. Twenty-two stable COPD patients had MMP measurements one week apart. During exacerbations, serum and urine MMP-9 were slightly elevated by 17% and 30% compared with recovery values respectively (p = 0.001 and p = 0.026). MMP-8 was not significantly changed. These MMP levels related to serum neutrophil numbers but not to outcome of exacerbations, disease severity measures or smoking status. In clinically stable patients, serum MMP levels did not vary significantly over 7 days, whereas urine MMPs varied by up to nine fold for MMP-8 (p = 0.003). Sputum, serum and urine contained different MMP species and complexes. Median values for sputum active MMP-9 were significantly different from serum (p = 0.035) and urine (p = 0.024). Serum and urine MMPs are only modestly elevated during exacerbations of COPD and unlikely to be useful biomarkers in this clinical setting. Airway, serum and urine MMP levels are independent of each other in COPD patients. Further, MMP levels are variable between patients and do not reflect airflow obstruction.


Asunto(s)
Metaloproteinasa 8 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Anciano , Anciano de 80 o más Años , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Volumen Espiratorio Forzado , Humanos , Masculino , Metaloproteinasa 8 de la Matriz/sangre , Metaloproteinasa 8 de la Matriz/orina , Metaloproteinasa 9 de la Matriz/sangre , Metaloproteinasa 9 de la Matriz/orina , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Índice de Severidad de la Enfermedad , Fumar/metabolismo , Espirometría , Esputo/metabolismo , Capacidad Vital
15.
Pediatr Pulmonol ; 51(3): 253-7, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26709241

RESUMEN

RATIONALE: Pulmonary infection and malnutrition in cystic fibrosis are associated with decreased survival. Glutamine has a possible anti-microbial effect, with a specific impact against Pseudomonas aeruginosa. We aimed to test the hypothesis that oral glutamine supplementation (21 g/day) for 8 weeks in adults with cystic fibrosis would decrease pulmonary inflammation and improve clinical status. METHODS: The study design was a randomized double-blind placebo-controlled study design with an iso-nitrogenous placebo. The primary analysis was intention to treat, and the primary outcome was change in induced sputum neutrophils. RESULTS: Thirty-nine individuals were recruited and thirty-six completed the study. Glutamine supplementation had no impact on any of the outcome measures in the intention-to-treat analysis. In the per protocol analysis, glutamine supplementation was associated with an increase in induced sputum neutrophils (P = 0.046), total cells (P = 0.03), and in Pseudomonas isolation agar colony forming units (P = 0.04) compared to placebo. CONCLUSIONS: There was no effect of glutamine supplementation on markers of pulmonary inflammation in the intention-to-treat analysis.


Asunto(s)
Fibrosis Quística/complicaciones , Suplementos Dietéticos , Glutamina/uso terapéutico , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Adulto , Antibacterianos/uso terapéutico , Método Doble Ciego , Femenino , Humanos , Masculino , Pseudomonas aeruginosa/aislamiento & purificación , Infecciones del Sistema Respiratorio/complicaciones , Resultado del Tratamiento , Adulto Joven
16.
Eur Respir J ; 46(5): 1308-21, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26357963

RESUMEN

U-BIOPRED is a European Union consortium of 20 academic institutions, 11 pharmaceutical companies and six patient organisations with the objective of improving the understanding of asthma disease mechanisms using a systems biology approach.This cross-sectional assessment of adults with severe asthma, mild/moderate asthma and healthy controls from 11 European countries consisted of analyses of patient-reported outcomes, lung function, blood and airway inflammatory measurements.Patients with severe asthma (nonsmokers, n=311; smokers/ex-smokers, n=110) had more symptoms and exacerbations compared to patients with mild/moderate disease (n=88) (2.5 exacerbations versus 0.4 in the preceding 12 months; p<0.001), with worse quality of life, and higher levels of anxiety and depression. They also had a higher incidence of nasal polyps and gastro-oesophageal reflux with lower lung function. Sputum eosinophil count was higher in severe asthma compared to mild/moderate asthma (median count 2.99% versus 1.05%; p=0.004) despite treatment with higher doses of inhaled and/or oral corticosteroids.Consistent with other severe asthma cohorts, U-BIOPRED is characterised by poor symptom control, increased comorbidity and airway inflammation, despite high levels of treatment. It is well suited to identify asthma phenotypes using the array of "omic" datasets that are at the core of this systems medicine approach.


Asunto(s)
Corticoesteroides/administración & dosificación , Antiasmáticos/administración & dosificación , Asma/complicaciones , Fumar/efectos adversos , Adulto , Ansiedad/epidemiología , Asma/tratamiento farmacológico , Asma/epidemiología , Estudios de Casos y Controles , Comorbilidad , Estudios Transversales , Depresión/epidemiología , Europa (Continente) , Femenino , Reflujo Gastroesofágico/epidemiología , Humanos , Masculino , Persona de Mediana Edad , Óxido Nítrico/análisis , Estudios Prospectivos , Calidad de Vida , Índice de Severidad de la Enfermedad , Fumar/epidemiología , Espirometría , Encuestas y Cuestionarios , Biología de Sistemas
17.
J Vis Exp ; (101): e52986, 2015 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-26275100

RESUMEN

Electrospinning is a highly adaptable method producing porous 3D fibrous scaffolds that can be exploited in in vitro cell culture. Alterations to intrinsic parameters within the process allow a high degree of control over scaffold characteristics including fiber diameter, alignment and porosity. By developing scaffolds with similar dimensions and topographies to organ- or tissue-specific extracellular matrices (ECM), micro-environments representative to those that cells are exposed to in situ can be created. The airway bronchiole wall, comprised of three main micro-environments, was selected as a model tissue. Using decellularized airway ECM as a guide, we electrospun the non-degradable polymer, polyethylene terephthalate (PET), by three different protocols to produce three individual electrospun scaffolds optimized for epithelial, fibroblast or smooth muscle cell-culture. Using a commercially available bioreactor system, we stably co-cultured the three cell-types to provide an in vitro model of the airway wall over an extended time period. This model highlights the potential for such methods being employed in in vitro diagnostic studies investigating important inter-cellular cross-talk mechanisms or assessing novel pharmaceutical targets, by providing a relevant platform to allow the culture of fully differentiated adult cells within 3D, tissue-specific environments.


Asunto(s)
Bronquiolos/citología , Ingeniería de Tejidos/métodos , Andamios del Tejido , Adulto , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/fisiología , Técnicas de Cocultivo/métodos , Matriz Extracelular/fisiología , Fibroblastos/citología , Humanos , Miocitos del Músculo Liso/citología , Tereftalatos Polietilenos , Polímeros , Porosidad
18.
Respir Res ; 16: 69, 2015 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-26062683

RESUMEN

BACKGROUND: Exacerbations of chronic obstructive pulmonary disease (COPD) contribute significantly to disease progression. However, the effect on tissue structure and turnover is not well described. There is an urgent clinical need for biomarkers of disease activity associated with disease progression. Extracellular matrix (ECM) turnover reflects activity in tissues and consequently assessment of ECM turnover may serve as biomarkers of disease activity. We hypothesized that the turnover of lung ECM proteins were altered during exacerbations of COPD. METHODS: 69 patients with COPD hospitalised for an exacerbation were recruited at admission and returned for a 4 weeks follow-up. Competitive ELISAs measuring circulating protein fragments in serum or plasma assessed the formation and degradation of collagen types III (Pro-C3 and C3M, respectively), IV (P4NP 7S and C4M, respectively), and VI (Pro-C6 and C6M, respectively), and degradation of elastin (ELM7 and EL-NE) and versican (VCANM). RESULTS: Circulating levels of C3M, C4M, C6M, ELM7, and EL-NE were elevated during an exacerbation of COPD as compared to follow-up (all P <0.0001), while VCANM levels were decreased (P <0.0001). Pro-C6 levels were decreased and P4NP 7S levels were elevated during exacerbation (P <0.0001). Pro-C3 levels were unchanged. At time of exacerbation, degradation/formation ratios were increased for collagen types III and VI and decreased for collagen type IV. CONCLUSIONS: Exacerbations of COPD resulted in elevated levels of circulating fragments of structural proteins, which may serve as markers of disease activity. This suggests that patients with COPD have accelerated ECM turnover during exacerbations which may be related to disease progression.


Asunto(s)
Progresión de la Enfermedad , Matriz Extracelular/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/sangre , Enfermedad Pulmonar Obstructiva Crónica/diagnóstico , Anciano , Biomarcadores/sangre , Femenino , Estudios de Seguimiento , Hospitalización/tendencias , Humanos , Masculino , Persona de Mediana Edad
20.
Am J Physiol Lung Cell Mol Physiol ; 309(3): L237-49, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26047642

RESUMEN

Human airway smooth muscle cells (HASMC) contribute to asthma pathophysiology through an increased smooth muscle mass and elevated cytokine/chemokine output. Little is known about how HASMC and the airway epithelium interact to regulate chronic airway inflammation and remodeling. Amphiregulin is a member of the family of epidermal growth factor receptor (EGFR) agonists with cell growth and proinflammatory roles and increased expression in the lungs of asthma patients. Here we show that bradykinin (BK) stimulation of HASMC increases amphiregulin secretion in a mechanism dependent on BK-induced COX-2 expression, increased PGE2 output, and the stimulation of HASMC EP2 and EP4 receptors. Conditioned medium from BK treated HASMC induced CXCL8, VEGF, and COX-2 mRNA and protein accumulation in airway epithelial cells, which were blocked by anti-amphiregulin antibodies and amphiregulin siRNA, suggesting a paracrine effect of HASMC-derived amphiregulin on airway epithelial cells. Consistent with this, recombinant amphiregulin induced CXCL8, VEGF, and COX-2 in airway epithelial cells. Finally, we found that conditioned media from amphiregulin-stimulated airway epithelial cells induced amphiregulin expression in HASMC and that this was dependent on airway epithelial cell COX-2 activity. Our study provides evidence of a dynamic axis of interaction between HASMC and epithelial cells that amplifies CXCL8, VEGF, COX-2, and amphiregulin production.


Asunto(s)
Ciclooxigenasa 2/genética , Familia de Proteínas EGF/metabolismo , Células Epiteliales/metabolismo , Miocitos del Músculo Liso/metabolismo , Anfirregulina , Asma/metabolismo , Bradiquinina/fisiología , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Expresión Génica , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Transducción de Señal , Activación Transcripcional , Factor A de Crecimiento Endotelial Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...