Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Development ; 150(9)2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-37102682

RESUMEN

Alveolar development and repair require tight spatiotemporal regulation of numerous signalling pathways that are influenced by chemical and mechanical stimuli. Mesenchymal cells play key roles in numerous developmental processes. Transforming growth factor-ß (TGFß) is essential for alveologenesis and lung repair, and the G protein α subunits Gαq and Gα11 (Gαq/11) transmit mechanical and chemical signals to activate TGFß in epithelial cells. To understand the role of mesenchymal Gαq/11 in lung development, we generated constitutive (Pdgfrb-Cre+/-;Gnaqfl/fl;Gna11-/-) and inducible (Pdgfrb-Cre/ERT2+/-;Gnaqfl/fl;Gna11-/-) mesenchymal Gαq/11 deleted mice. Mice with constitutive Gαq/11 gene deletion exhibited abnormal alveolar development, with suppressed myofibroblast differentiation, altered mesenchymal cell synthetic function, and reduced lung TGFß2 deposition, as well as kidney abnormalities. Tamoxifen-induced mesenchymal Gαq/11 gene deletion in adult mice resulted in emphysema associated with reduced TGFß2 and elastin deposition. Cyclical mechanical stretch-induced TGFß activation required Gαq/11 signalling and serine protease activity, but was independent of integrins, suggesting an isoform-specific role for TGFß2 in this model. These data highlight a previously undescribed mechanism of cyclical stretch-induced Gαq/11-dependent TGFß2 signalling in mesenchymal cells, which is imperative for normal alveologenesis and maintenance of lung homeostasis.


Asunto(s)
Receptor beta de Factor de Crecimiento Derivado de Plaquetas , Factor de Crecimiento Transformador beta , Ratones , Animales , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Transducción de Señal , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Homeostasis
2.
Immunol Rev ; 302(1): 228-240, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34028807

RESUMEN

The COVID-19 pandemic rapidly spread around the world following the first reports in Wuhan City, China in late 2019. The disease, caused by the novel SARS-CoV-2 virus, is primarily a respiratory condition that can affect numerous other bodily systems including the cardiovascular and gastrointestinal systems. The disease ranges in severity from asymptomatic through to severe acute respiratory distress requiring intensive care treatment and mechanical ventilation, which can lead to respiratory failure and death. It has rapidly become evident that COVID-19 patients can develop features of interstitial pulmonary fibrosis, which in many cases persist for as long as we have thus far been able to follow the patients. Many questions remain about how such fibrotic changes occur within the lung of COVID-19 patients, whether the changes will persist long term or are capable of resolving, and whether post-COVID-19 pulmonary fibrosis has the potential to become progressive, as in other fibrotic lung diseases. This review brings together our existing knowledge on both COVID-19 and pulmonary fibrosis, with a particular focus on lung epithelial cells and fibroblasts, in order to discuss common pathways and processes that may be implicated as we try to answer these important questions in the months and years to come.


Asunto(s)
COVID-19/patología , Células Epiteliales/patología , Fibroblastos/patología , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/virología , Mucosa Respiratoria/patología , COVID-19/complicaciones , Humanos , SARS-CoV-2
3.
Am J Physiol Lung Cell Mol Physiol ; 324(3): L285-L296, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36625900

RESUMEN

As survival of extremely preterm infants continues to improve, there is also an associated increase in bronchopulmonary dysplasia (BPD), one of the most significant complications of preterm birth. BPD development is multifactorial resulting from exposure to multiple antenatal and postnatal stressors. BPD has both short-term health implications and long-term sequelae including increased respiratory, cardiovascular, and neurological morbidity. Transforming growth factor ß (TGF-ß) is an important signaling pathway in lung development, organ injury, and fibrosis and is implicated in the development of BPD. This review provides a detailed account on the role of TGF-ß in antenatal and postnatal lung development, the effect of known risk factors for BPD on the TGF-ß signaling pathway, and how medications currently in use or under development, for the prevention or treatment of BPD, affect TGF-ß signaling.


Asunto(s)
Displasia Broncopulmonar , Nacimiento Prematuro , Lactante , Recién Nacido , Femenino , Humanos , Embarazo , Displasia Broncopulmonar/metabolismo , Recien Nacido Prematuro , Nacimiento Prematuro/metabolismo , Pulmón/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Transducción de Señal
4.
Am J Physiol Lung Cell Mol Physiol ; 324(3): L271-L284, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36594851

RESUMEN

Airway remodeling occurs in chronic asthma leading to increased airway smooth muscle (ASM) mass and extracellular matrix (ECM) deposition. Although extensively studied in murine airways, studies report only selected larger airways at one time-point meaning the spatial distribution and resolution of remodeling are poorly understood. Here we use a new method allowing comprehensive assessment of the spatial and temporal changes in ASM, ECM, and epithelium in large numbers of murine airways after allergen challenge. Using image processing to analyze 20-50 airways per mouse from a whole lung section revealed increases in ASM and ECM after allergen challenge were greater in small and large rather than intermediate airways. ASM predominantly accumulated adjacent to the basement membrane, whereas ECM was distributed across the airway wall. Epithelial hyperplasia was most marked in small and intermediate airways. After challenge, ASM changes resolved over 7 days, whereas ECM and epithelial changes persisted. The new method suggests large and small airways remodel differently, and the long-term consequences of airway inflammation may depend more on ECM and epithelial changes than ASM. The improved quantity and quality of unbiased data provided by the method reveals important spatial differences in remodeling and could set new analysis standards for murine asthma models.


Asunto(s)
Asma , Pulmón , Ratones , Animales , Músculo Liso , Matriz Extracelular/fisiología , Remodelación de las Vías Aéreas (Respiratorias)/fisiología , Alérgenos
5.
Eur Respir J ; 60(1)2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34996828

RESUMEN

BACKGROUND: Airway smooth muscle (ASM) cells are fundamental to asthma pathogenesis, influencing bronchoconstriction, airway hyperresponsiveness and airway remodelling. The extracellular matrix (ECM) can influence tissue remodelling pathways; however, to date no study has investigated the effect of ASM ECM stiffness and cross-linking on the development of asthmatic airway remodelling. We hypothesised that transforming growth factor-ß (TGF-ß) activation by ASM cells is influenced by ECM in asthma and sought to investigate the mechanisms involved. METHODS: This study combines in vitro and in vivo approaches: human ASM cells were used in vitro to investigate basal TGF-ß activation and expression of ECM cross-linking enzymes. Human bronchial biopsies from asthmatic and nonasthmatic donors were used to confirm lysyl oxidase like 2 (LOXL2) expression in ASM. A chronic ovalbumin (OVA) model of asthma was used to study the effect of LOXL2 inhibition on airway remodelling. RESULTS: We found that asthmatic ASM cells activated more TGF-ß basally than nonasthmatic controls and that diseased cell-derived ECM influences levels of TGF-ß activated. Our data demonstrate that the ECM cross-linking enzyme LOXL2 is increased in asthmatic ASM cells and in bronchial biopsies. Crucially, we show that LOXL2 inhibition reduces ECM stiffness and TGF-ß activation in vitro, and can reduce subepithelial collagen deposition and ASM thickness, two features of airway remodelling, in an OVA mouse model of asthma. CONCLUSION: These data are the first to highlight a role for LOXL2 in the development of asthmatic airway remodelling and suggest that LOXL2 inhibition warrants further investigation as a potential therapy to reduce remodelling of the airways in severe asthma.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias) , Aminoácido Oxidorreductasas/metabolismo , Asma , Remodelación de las Vías Aéreas (Respiratorias)/fisiología , Animales , Asma/metabolismo , Ratones , Músculo Liso/patología , Proteína-Lisina 6-Oxidasa/metabolismo , Proteína-Lisina 6-Oxidasa/farmacología , Factor de Crecimiento Transformador beta/metabolismo
6.
J Math Biol ; 82(5): 35, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33721103

RESUMEN

Precision-cut lung-slices (PCLS), in which viable airways embedded within lung parenchyma are stretched or induced to contract, are a widely used ex vivo assay to investigate bronchoconstriction and, more recently, mechanical activation of pro-remodelling cytokines in asthmatic airways. We develop a nonlinear fibre-reinforced biomechanical model accounting for smooth muscle contraction and extracellular matrix strain-stiffening. Through numerical simulation, we describe the stresses and contractile responses of an airway within a PCLS of finite thickness, exposing the importance of smooth muscle contraction on the local stress state within the airway. We then consider two simplifying limits of the model (a membrane representation and an asymptotic reduction in the thin-PCLS-limit), that permit analytical progress. Comparison against numerical solution of the full problem shows that the asymptotic reduction successfully captures the key elements of the full model behaviour. The more tractable reduced model that we develop is suitable to be employed in investigations to elucidate the time-dependent feedback mechanisms linking airway mechanics and cytokine activation in asthma.


Asunto(s)
Pulmón , Modelos Teóricos , Fenómenos Biomecánicos , Broncoconstricción , Simulación por Computador , Citocinas/química , Matriz Extracelular/química , Humanos , Pulmón/química , Contracción Muscular/fisiología
7.
Am J Respir Cell Mol Biol ; 56(5): 667-679, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28459387

RESUMEN

Numerous compounds have shown efficacy in limiting development of pulmonary fibrosis using animal models, yet few of these compounds have replicated these beneficial effects in clinical trials. Given the challenges associated with performing clinical trials in patients with idiopathic pulmonary fibrosis (IPF), it is imperative that preclinical data packages be robust in their analyses and interpretations to have the best chance of selecting promising drug candidates to advance to clinical trials. The American Thoracic Society has convened a group of experts in lung fibrosis to discuss and formalize recommendations for preclinical assessment of antifibrotic compounds. The panel considered three major themes (choice of animal, practical considerations of fibrosis modeling, and fibrotic endpoints for evaluation). Recognizing the need for practical considerations, we have taken a pragmatic approach. The consensus view is that use of the murine intratracheal bleomycin model in animals of both genders, using hydroxyproline measurements for collagen accumulation along with histologic assessments, is the best-characterized animal model available for preclinical testing. Testing of antifibrotic compounds in this model is recommended to occur after the acute inflammatory phase has subsided (generally after Day 7). Robust analyses may also include confirmatory studies in human IPF specimens and validation of results in a second system using in vivo or in vitro approaches. The panel also strongly encourages the publication of negative results to inform the lung fibrosis community. These recommendations are for preclinical therapeutic evaluation only and are not intended to dissuade development of emerging technologies to better understand IPF pathogenesis.


Asunto(s)
Congresos como Asunto , Modelos Animales de Enfermedad , Fibrosis Pulmonar/terapia , Sociedades Médicas , Animales , Determinación de Punto Final , Femenino , Humanos , Masculino , Organismos Modificados Genéticamente , Reproducibilidad de los Resultados
8.
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
10.
Lab Invest ; 96(6): 623-31, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26974397

RESUMEN

Idiopathic pulmonary fibrosis is a progressive, fatal disease with limited treatment options. Protease-mediated transforming growth factor-ß (TGF-ß) activation has been proposed as a pathogenic mechanism of lung fibrosis. Protease activity in the lung is tightly regulated by protease inhibitors, particularly secretory leukocyte protease inhibitor (SLPI). The bleomycin model of lung fibrosis was used to determine the effect of increased protease activity in the lungs of Slpi(-/-) mice following injury. Slpi(-/-), and wild-type, mice received oropharyngeal administration of bleomycin (30 IU) and the development of pulmonary fibrosis was assessed. Pro and active forms of matrix metalloproteinase (MMP)-2 and MMP-9 were measured. Lung fibrosis was determined by collagen subtype-specific gene expression, hydroxyproline concentration, and histological assessment. Alveolar TGF-ß activation was measured using bronchoalveolar lavage cell pSmad2 levels and global TGF-ß activity was assessed by pSmad2 immunohistochemistry. The active-MMP-9 to pro-MMP-9 ratio was significantly increased in Slpi(-/-) animals compared with wild-type animals, demonstrating enhanced metalloproteinase activity. Wild-type animals showed an increase in TGF-ß activation following bleomycin, with a progressive and sustained increase in collagen type I, alpha 1 (Col1α1), III, alpha 1(Col3α1), IV, alpha 1(Col4α1) mRNA expression, and a significant increase in total lung collagen 28 days post bleomycin. In contrast Slpi(-/-) mice showed no significant increase of alveolar TGF-ß activity following bleomycin, above their already elevated levels, although global TGF-ß activity did increase. Slpi(-/-) mice had impaired collagen gene expression but animals demonstrated minimal reduction in lung fibrosis compared with wild-type animals. These data suggest that enhanced proteolysis does not further enhance TGF-ß activation, and inhibits sustained Col1α1, Col3α1, and Col4α1 gene expression following lung injury. However, these changes do not prevent the development of lung fibrosis. Overall, these data suggest that the absence of Slpi does not markedly modify the development of lung fibrosis following bleomycin-induced lung injury.


Asunto(s)
Fibrosis Pulmonar Idiopática/etiología , Lesión Pulmonar/etiología , Inhibidor Secretorio de Peptidasas Leucocitarias/deficiencia , Animales , Bleomicina/toxicidad , Colágeno/genética , Colágeno/metabolismo , Eliminación de Gen , Hidroxiprolina/metabolismo , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Lesión Pulmonar/genética , Lesión Pulmonar/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , ARN Mensajero/genética , ARN Mensajero/metabolismo , Inhibidor Secretorio de Peptidasas Leucocitarias/genética , Factor de Crecimiento Transformador beta/metabolismo
11.
Thorax ; 71(6): 565-7, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26911575

RESUMEN

Caffeine is a commonly used food additive found naturally in many products. In addition to potently stimulating the central nervous system caffeine is able to affect various systems within the body including the cardiovascular and respiratory systems. Importantly, caffeine is used clinically to treat apnoea and bronchopulmonary dysplasia in premature babies. Recently, caffeine has been shown to exhibit antifibrotic effects in the liver in part through reducing collagen expression and deposition, and reducing expression of the profibrotic cytokine TGFß. The potential antifibrotic effects of caffeine in the lung have not previously been investigated. Using a combined in vitro and ex vivo approach we have demonstrated that caffeine can act as an antifibrotic agent in the lung by acting on two distinct cell types, namely epithelial cells and fibroblasts. Caffeine inhibited TGFß activation by lung epithelial cells in a concentration-dependent manner but had no effect on TGFß activation in fibroblasts. Importantly, however, caffeine abrogated profibrotic responses to TGFß in lung fibroblasts. It inhibited basal expression of the α-smooth muscle actin gene and reduced TGFß-induced increases in profibrotic genes. Finally, caffeine reduced established bleomycin-induced fibrosis after 5 days treatment in an ex vivo precision-cut lung slice model. Together, these findings suggest that there is merit in further investigating the potential use of caffeine, or its analogues, as antifibrotic agents in the lung.


Asunto(s)
Cafeína/farmacología , Células Epiteliales/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Biomarcadores/metabolismo , Células Cultivadas , Células Epiteliales/metabolismo , Fibroblastos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Fibrosis Pulmonar/genética , Transducción de Señal/efectos de los fármacos
12.
Magn Reson Med ; 76(4): 1224-35, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26507239

RESUMEN

PURPOSE: Asthma is a disease of increasing worldwide importance that calls for new investigative methods. Ex vivo lung tissue is being increasingly used to study functional respiratory parameters independent of confounding systemic considerations but also to reduce animal numbers and associated research costs. In this work, a straightforward laboratory method is advanced to probe dynamic changes in gas inhalation patterns by using an ex vivo small animal ovalbumin (OVA) model of human asthma. METHODS: Hyperpolarized (hp) (129) Xe was actively inhaled by the excised lungs exposed to a constant pressure differential that mimicked negative pleural cavity pressure. The method enabled hp (129) Xe MRI of airway responsiveness to intravenous methacholine (MCh) and airway challenge reversal through salbutamol. RESULTS: Significant differences were demonstrated between control and OVA challenged animals on global lung hp (129) Xe gas inhalation with P < 0.05 at MCh dosages above 460 µg. Spatial mapping of the regional hp gas distribution revealed an approximately three-fold increase in heterogeneity for the asthma model organs. CONCLUSION: The experimental results from this proof of concept work suggest that the ex vivo hp noble gas imaging arrangement and the applied image analysis methodology may be useful as an adjunct to current diagnostic techniques. Magn Reson Med 76:1224-1235, 2016. © 2015 The Authors. Magnetic Resonance in Medicine published by Wiley Periodicals, Inc. on behalf of International Society for Magnetic Resonance in Medicine. This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.


Asunto(s)
Asma/diagnóstico por imagen , Asma/fisiopatología , Pulmón/diagnóstico por imagen , Pulmón/fisiopatología , Imagen por Resonancia Magnética/métodos , Intercambio Gaseoso Pulmonar , Isótopos de Xenón/farmacocinética , Administración por Inhalación , Animales , Simulación por Computador , Aumento de la Imagen/métodos , Interpretación de Imagen Asistida por Computador/métodos , Masculino , Modelos Biológicos , Imagen Molecular/métodos , Radiofármacos/administración & dosificación , Radiofármacos/farmacocinética , Ratas , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Distribución Tisular , Isótopos de Xenón/administración & dosificación
13.
J Biol Chem ; 289(51): 35246-63, 2014 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-25339175

RESUMEN

Influenza infection exacerbates chronic pulmonary diseases, including idiopathic pulmonary fibrosis. A central pathway in the pathogenesis of idiopathic pulmonary fibrosis is epithelial injury leading to activation of transforming growth factor ß (TGFß). The mechanism and functional consequences of influenza-induced activation of epithelial TGFß are unclear. Influenza stimulates toll-like receptor 3 (TLR3), which can increase RhoA activity, a key event prior to activation of TGFß by the αvß6 integrin. We hypothesized that influenza would stimulate TLR3 leading to activation of latent TGFß via αvß6 integrin in epithelial cells. Using H1152 (IC50 6.1 µm) to inhibit Rho kinase and 6.3G9 to inhibit αvß6 integrins, we demonstrate their involvement in influenza (A/PR/8/34 H1N1) and poly(I:C)-induced TGFß activation. We confirm the involvement of TLR3 in this process using chloroquine (IC50 11.9 µm) and a dominant negative TLR3 construct (pZERO-hTLR3). Examination of lungs from influenza-infected mice revealed augmented levels of collagen deposition, phosphorylated Smad2/3, αvß6 integrin, and apoptotic cells. Finally, we demonstrate that αvß6 integrin-mediated TGFß activity following influenza infection promotes epithelial cell death in vitro and enhanced collagen deposition in vivo and that this response is diminished in Smad3 knock-out mice. These data show that H1N1 and poly(I:C) can induce αvß6 integrin-dependent TGFß activity in epithelial cells via stimulation of TLR3 and suggest a novel mechanism by which influenza infection may promote collagen deposition in fibrotic lung disease.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Colágeno/metabolismo , Células Epiteliales/metabolismo , Integrinas/metabolismo , Infecciones por Orthomyxoviridae/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Animales , Antígenos de Neoplasias/genética , Antivirales/farmacología , Apoptosis , Línea Celular Transformada , Perros , Células Epiteliales/efectos de los fármacos , Células Epiteliales/virología , Interacciones Huésped-Patógeno , Humanos , Immunoblotting , Subtipo H1N1 del Virus de la Influenza A/fisiología , Integrinas/genética , Pulmón/metabolismo , Pulmón/virología , Células de Riñón Canino Madin Darby , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/virología , Fosforilación/efectos de los fármacos , Poli I-C/farmacología , Proteína smad3/genética , Proteína smad3/metabolismo , Receptor Toll-Like 3/metabolismo , Factor de Crecimiento Transformador beta/genética , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
14.
Am J Physiol Lung Cell Mol Physiol ; 308(9): L962-72, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25713319

RESUMEN

Asthma is characterized by airway inflammation and remodeling and CXCL8 is a CXC chemokine that drives steroid-resistant neutrophilic airway inflammation. We have shown that airway smooth muscle (ASM) cells isolated from asthmatic individuals secrete more CXCL8 than cells from nonasthmatic individuals. Here we investigated chromatin modifications at the CXCL8 promoter in ASM cells from nonasthmatic and asthmatic donors to further understand how CXCL8 is dysregulated in asthma. ASM cells from asthmatic donors had increased histone H3 acetylation, specifically histone H3K18 acetylation, and increased binding of histone acetyltransferase p300 compared with nonasthmatic donors but no differences in CXCL8 DNA methylation. The acetylation reader proteins Brd3 and Brd4 were bound to the CXCL8 promoter and Brd inhibitors inhibited CXCL8 secretion from ASM cells by disrupting Brd4 and RNA polymerase II binding to the CXCL8 promoter. Our results show a novel dysregulation of CXCL8 transcriptional regulation in asthma characterized by a promoter complex that is abnormal in ASM cells isolated from asthmatic donors and can be modulated by Brd inhibitors. Brd inhibitors may provide a new therapeutic strategy for steroid-resistant inflammation.


Asunto(s)
Asma/metabolismo , Interleucina-8/metabolismo , Músculo Liso/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Acetilación , Remodelación de las Vías Aéreas (Respiratorias)/inmunología , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteínas de Ciclo Celular , Células Cultivadas , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Histonas/metabolismo , Humanos , Inflamación/inmunología , Interleucina-8/antagonistas & inhibidores , Interleucina-8/genética , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Cultivo Primario de Células , Regiones Promotoras Genéticas , Unión Proteica , ARN Polimerasa II/metabolismo , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/metabolismo , Factor de Transcripción ReIA/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Transcripción Genética , Factores de Transcripción p300-CBP/metabolismo
16.
Pulm Pharmacol Ther ; 29(2): 96-107, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25062835

RESUMEN

Asthma is an obstructive airway disease, with a heterogeneous and multifactorial pathogenesis. Although generally considered to be a disease principally driven by chronic inflammation, it is becoming increasingly recognised that the immune component of the pathology poorly correlates with the clinical symptoms of asthma, thus highlighting a potentially central role for non-immune cells. In this context airway smooth muscle (ASM) may be a key player, as it comprises a significant proportion of the airway wall and is the ultimate effector of acute airway narrowing. Historically, the contribution of ASM to asthma pathogenesis has been contentious, yet emerging evidence suggests that ASM contractile activation imparts chronic effects that extend well beyond the temporary effects of bronchoconstriction. In this review article we describe the effects that ASM contraction, in combination with cellular mechanotransduction and novel contraction-inflammation synergies, contribute to asthma pathogenesis. Specific emphasis will be placed on the effects that ASM contraction exerts on the mechanical properties of the airway wall, as well as novel mechanisms by which ASM contraction may contribute to more established features of asthma such as airway wall remodelling.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/fisiología , Asma/fisiopatología , Mecanotransducción Celular/fisiología , Contracción Muscular/fisiología , Músculo Liso/fisiopatología , Animales , Asma/etiología , Broncoconstricción/fisiología , Humanos , Fenómenos Fisiológicos Respiratorios
17.
J Immunol ; 187(11): 6094-107, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22025551

RESUMEN

Severe asthma is associated with airway remodeling, characterized by structural changes including increased smooth muscle mass and matrix deposition in the airway, leading to deteriorating lung function. TGF-ß is a pleiotropic cytokine leading to increased synthesis of matrix molecules by human airway smooth muscle (HASM) cells and is implicated in asthmatic airway remodeling. TGF-ß is synthesized as a latent complex, sequestered in the extracellular matrix, and requires activation for functionality. Activation of latent TGF-ß is the rate-limiting step in its bioavailability. This study investigated the effect of the contraction agonists LPA and methacholine on TGF-ß activation by HASM cells and its role in the development of asthmatic airway remodeling. The data presented show that LPA and methacholine induced TGF-ß activation by HASM cells via the integrin αvß5. Our findings highlight the importance of the ß5 cytoplasmic domain because a polymorphism in the ß5 subunit rendered the integrin unable to activate TGF-ß. To our knowledge, this is the first description of a biologically relevant integrin that is unable to activate TGF-ß. These data demonstrate that murine airway smooth muscle cells express αvß5 integrins and activate TGF-ß. Finally, these data show that inhibition, or genetic loss, of αvß5 reduces allergen-induced increases in airway smooth muscle thickness in two models of asthma. These data highlight a mechanism of TGF-ß activation in asthma and support the hypothesis that bronchoconstriction promotes airway remodeling via integrin mediated TGF-ß activation.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/inmunología , Asma/metabolismo , Miocitos del Músculo Liso/metabolismo , Receptores de Vitronectina/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Asma/inmunología , Asma/patología , Western Blotting , Línea Celular , Separación Celular , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Inmunohistoquímica , Inmunoprecipitación , Ratones , Miocitos del Músculo Liso/inmunología , Miocitos del Músculo Liso/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Vitronectina/inmunología , Sistema Respiratorio , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Factor de Crecimiento Transformador beta/inmunología
18.
Pharmacol Ther ; 249: 108504, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37482099

RESUMEN

Pulmonary fibrosis is a debilitating and life-limiting lung condition in which the damage- response mechanisms of mixed-population cells within the lungs go awry. The tissue microenvironment is drastically remodelled by aberrantly activated fibroblasts which deposit ECM components into the surrounding lung tissue, detrimentally affecting lung function and capacity for gas exchange. Growing evidence suggests a role for adenosine signalling in the pathology of tissue fibrosis in a variety of organs, including the lung, but the molecular pathways through which this occurs remain largely unknown. This review explores the role of adenosine in fibrosis and evaluates the contribution of the different adenosine receptors to fibrogenesis. Therapeutic targeting of the adenosine receptors is also considered, along with clinical observations pointing towards a role for adenosine in fibrosis. In addition, the interaction between adenosine signalling and other profibrotic signalling pathways, such as TGFß1 signalling, is discussed.


Asunto(s)
Fibrosis Pulmonar Idiopática , Humanos , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Pulmón/metabolismo , Fibrosis , Fibroblastos/metabolismo , Adenosina/metabolismo , Receptores Purinérgicos P1/metabolismo
19.
J Asthma Allergy ; 15: 595-610, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35592385

RESUMEN

Airway remodeling is a complex clinical feature of asthma that involves long-term disruption and modification of airway architecture, which contributes significantly to airway hyperresponsiveness (AHR) and lung function decline. It is characterized by thickening of the airway smooth muscle layer, deposition of a matrix below the airway epithelium, resulting in subepithelial fibrosis, changes within the airway epithelium, leading to disruption of the barrier, and excessive mucous production and angiogenesis within the airway wall. Airway remodeling contributes to stiffer and less compliant airways in asthma and leads to persistent, irreversible airflow obstruction. Current asthma treatments aim to reduce airway inflammation and exacerbations but none are targeted towards airway remodeling. Inhibiting the development of airway remodeling or reversing established remodeling has the potential to dramatically improve symptoms and disease burden in asthmatic patients. Integrins are a family of transmembrane heterodimeric proteins that serve as the primary receptors for extracellular matrix (ECM) components, mediating cell-cell and cell-ECM interactions to initiate intracellular signaling cascades. Cells present within the lungs, including structural and inflammatory cells, express a wide and varying range of integrin heterodimer combinations and permutations. Integrins are emerging as an important regulator of inflammation, repair, remodeling, and fibrosis in the lung, particularly in chronic lung diseases such as asthma. Here, we provide a comprehensive summary of the current state of knowledge on integrins in the asthmatic airway and how these integrins promote the remodeling process, and emphasize their potential involvement in airway disease.

20.
Methods Mol Biol ; 2299: 99-108, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34028736

RESUMEN

Myofibroblasts are critical to processes involved in normal wound healing and during pathological fibrosis. They transdifferentiate from fibroblasts, and in doing so become contractile and capable of secreting large amounts of extracellular matrix proteins. Transforming growth factor-beta (TGFß) is a key cytokine involved in wound healing and fibrogenesis. TGFß signaling has long been the subject of experimental therapeutic approaches to inhibit fibrosis in a variety of organ systems. Inhibition of TGFß can reduce myofibroblast transdifferentiation, contractility, and matrix production. Importantly, TGFß is released from cells and sequestered in the extracellular matrix in a latent form that requires activation for biological function. There have been multiple mechanisms of TGFß activation described in a variety of cell types and in cell free systems; however, myofibroblasts have previously been shown to activate TGFß via cell surface integrins, particularly αvß5 integrins. This chapter will provide detailed protocols for accurately measuring activation of TGFß by myofibroblasts in vitro. Levels of active TGFß usually represent a small proportion of the total amount of latent TGFß present in the matrix. Methods to measure active TGFß therefore need to be sensitive and specific to detect the active cytokine only.


Asunto(s)
Miofibroblastos/citología , Receptores de Vitronectina/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular , Células Cultivadas , Proteínas de la Matriz Extracelular/metabolismo , Humanos , Miofibroblastos/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA