Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
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.
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
3.
Nat Commun ; 11(1): 4659, 2020 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-32938936

RESUMEN

The αvß6 integrin plays a key role in the activation of transforming growth factor-ß (TGFß), a pro-fibrotic mediator that is pivotal to the development of idiopathic pulmonary fibrosis (IPF). We identified a selective small molecule αvß6 RGD-mimetic, GSK3008348, and profiled it in a range of disease relevant pre-clinical systems. To understand the relationship between target engagement and inhibition of fibrosis, we measured pharmacodynamic and disease-related end points. Here, we report, GSK3008348 binds to αvß6 with high affinity in human IPF lung and reduces downstream pro-fibrotic TGFß signaling to normal levels. In human lung epithelial cells, GSK3008348 induces rapid internalization and lysosomal degradation of the αvß6 integrin. In the murine bleomycin-induced lung fibrosis model, GSK3008348 engages αvß6, induces prolonged inhibition of TGFß signaling and reduces lung collagen deposition and serum C3M, a marker of IPF disease progression. These studies highlight the potential of inhaled GSK3008348 as an anti-fibrotic therapy.


Asunto(s)
Butiratos/farmacología , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Integrinas/antagonistas & inhibidores , Naftiridinas/farmacología , Pirazoles/farmacología , Pirrolidinas/farmacología , Administración por Inhalación , Animales , Antígenos de Neoplasias/metabolismo , Bleomicina/toxicidad , Butiratos/administración & dosificación , Butiratos/metabolismo , Butiratos/farmacocinética , Colágeno/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Humanos , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/patología , Integrinas/metabolismo , Masculino , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Naftiridinas/administración & dosificación , Naftiridinas/metabolismo , Naftiridinas/farmacocinética , Pirazoles/administración & dosificación , Pirazoles/metabolismo , Pirazoles/farmacocinética , Pirrolidinas/administración & dosificación , Pirrolidinas/metabolismo , Pirrolidinas/farmacocinética , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Tomografía Computarizada de Emisión de Fotón Único , Factor de Crecimiento Transformador beta/metabolismo , Investigación Biomédica Traslacional
4.
Int J Biochem Cell Biol ; 120: 105668, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31877385

RESUMEN

ETS domain-containing protein-1 (ELK1) is a transcription factor important in regulating αvß6 integrin expression. αvß6 integrins activate the profibrotic cytokine Transforming Growth Factor ß1 (TGFß1) and are increased in the alveolar epithelium in idiopathic pulmonary fibrosis (IPF). IPF is a disease associated with aging and therefore we hypothesised that aged animals lacking Elk1 globally would develop spontaneous fibrosis in organs where αvß6 mediated TGFß activation has been implicated. Here we identify that Elk1-knockout (Elk1-/0) mice aged to one year developed spontaneous fibrosis in the absence of injury in both the lung and the liver but not in the heart or kidneys. The lungs of Elk1-/0 aged mice demonstrated increased collagen deposition, in particular collagen 3α1, located in small fibrotic foci and thickened alveolar walls. Despite the liver having relatively low global levels of ELK1 expression, Elk1-/0 animals developed hepatosteatosis and fibrosis. The loss of Elk1 also had differential effects on Itgb1, Itgb5 and Itgb6 expression in the four organs potentially explaining the phenotypic differences in these organs. To understand the potential causes of reduced ELK1 in human disease we exposed human lung epithelial cells and murine lung slices to cigarette smoke extract, which lead to reduced ELK1 expression andmay explain the loss of ELK1 in human disease. These data support a fundamental role for ELK1 in protecting against the development of progressive fibrosis via transcriptional regulation of beta integrin subunit genes, and demonstrate that loss of ELK1 can be caused by cigarette smoke.


Asunto(s)
Bronquios/patología , Pulmón/patología , Proteína Elk-1 con Dominio ets/deficiencia , Factores de Edad , Animales , Bronquios/metabolismo , Fibrosis/metabolismo , Fibrosis/patología , Humanos , Pulmón/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteína Elk-1 con Dominio ets/metabolismo
5.
Am J Respir Cell Mol Biol ; 58(5): 594-603, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29053339

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is characterized by accumulation of extracellular matrix (ECM) proteins and fibroblast proliferation. ECM cross-linking enzymes have been implicated in fibrotic diseases, and we hypothesized that the ECM in IPF is abnormally cross-linked, which enhances fibroblast growth and resistance to normal ECM turnover. We used a combination of in vitro ECM preparations and in vivo assays to examine the expression of cross-linking enzymes and the effect of their inhibitors on fibroblast growth and ECM turnover. Lysyl oxidase-like 1 (LOXL1), LOXL2, LOXL3, and LOXL4 were expressed equally in control and IPF-derived fibroblasts. Transglutaminase 2 was more strongly expressed in IPF fibroblasts. LOXL2-, transglutaminase 2-, and transglutaminase-generated cross-links were strongly expressed in IPF lung tissue. Fibroblasts grown on IPF ECM had higher LOXL3 protein expression and transglutaminase activity than those grown on control ECM. IPF-derived ECM also enhanced fibroblast adhesion and proliferation compared with control ECM. Inhibition of lysyl oxidase and transglutaminase activity during ECM formation affected ECM structure as visualized by electron microscopy, and it reduced the enhanced fibroblast adhesion and proliferation of IPF ECM to control levels. Inhibition of transglutaminase, but not of lysyl oxidase, activity enhanced the turnover of ECM in vitro. In bleomycin-treated mice, during the postinflammatory fibrotic phase, inhibition of transglutaminases was associated with a reduction in whole-lung collagen. Our findings suggest that the ECM in IPF may enhance pathological cross-linking, which contributes to increased fibroblast growth and resistance to normal ECM turnover to drive lung fibrosis.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias) , Proliferación Celular , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/metabolismo , Aminoácido Oxidorreductasas/metabolismo , Animales , Bleomicina , Adhesión Celular , Células Cultivadas , Cistamina/farmacología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/ultraestructura , Proteínas de Unión al GTP/antagonistas & inhibidores , Proteínas de Unión al GTP/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/patología , Fibrosis Pulmonar Idiopática/prevención & control , Pulmón/efectos de los fármacos , Pulmón/ultraestructura , Ratones Endogámicos C57BL , Proteína Glutamina Gamma Glutamiltransferasa 2 , Proteolisis , Transglutaminasas/antagonistas & inhibidores , Transglutaminasas/metabolismo
6.
Lancet Respir Med ; 5(12): 946-955, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29150411

RESUMEN

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a progressive, fatal disorder with a variable disease trajectory. The aim of this study was to assess potential biomarkers to predict outcomes for people with IPF. METHODS: PROFILE is a large prospective longitudinal cohort of treatment-naive patients with IPF. We adopted a two-stage discovery and validation design using patients from the PROFILE cohort. For the discovery analysis, we examined 106 patients and 50 age and sex matched healthy controls from Nottingham University Hospitals NHS Trust and the Royal Brompton Hospital. We did an unbiased, multiplex immunoassay assessment of 123 biomarkers. We further investigated promising novel markers by immunohistochemical assessment of IPF lung tissue. In the validation analysis, we examined samples from 206 people with IPF from among the remaining 212 patients recruited to PROFILE Central England. We used the samples to attempt to replicate the biomarkers identified from the discovery analysis by use of independent immunoassays for each biomarker. We investigated the predictive power of the selected biomarkers to identify individuals with IPF who were at risk of progression or death. The PROFILE studies are registered on ClinicalTrials.gov, numbers NCT01134822 (PROFILE Central England) and NCT01110694 (PROFILE Royal Brompton Hospital). FINDINGS: In the discovery analysis, we identified four serum biomarkers (surfactant protein D, matrix metalloproteinase 7, CA19-9, and CA-125) that were suitable for replication. Histological assessment of CA19-9 and CA-125 suggested that these proteins were markers of epithelial damage. Replication analysis showed that baseline values of surfactant protein D (46·6 ng/mL vs 34·6 ng/mL, p=0·0018) and CA19-9 (53·7 U/mL vs 22·2 U/mL; p<0·0001) were significantly higher in patients with progressive disease than in patients with stable disease, and rising concentrations of CA-125 over 3 months were associated with increased risk of mortality (HR 2·542, 95% CI 1·493-4·328, p=0·00059). INTERPRETATION: We have identified serum proteins secreted from metaplastic epithelium that can be used to predict disease progression and death in IPF. FUNDING: GlaxoSmithKline R&D and the UK Medical Research Council.


Asunto(s)
Antígeno Ca-125/sangre , Antígeno CA-19-9/sangre , Progresión de la Enfermedad , Fibrosis Pulmonar Idiopática/sangre , Metaloproteinasa 7 de la Matriz/sangre , Proteína D Asociada a Surfactante Pulmonar/sangre , Biomarcadores/sangre , Estudios de Casos y Controles , Ensayo de Inmunoadsorción Enzimática , Epitelio/metabolismo , Epitelio/patología , Femenino , Humanos , Fibrosis Pulmonar Idiopática/mortalidad , Estudios Longitudinales , Pulmón/metabolismo , Pulmón/patología , Masculino , Valor Predictivo de las Pruebas , Estudios Prospectivos
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
J Nucl Med ; 54(12): 2146-52, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24167080

RESUMEN

UNLABELLED: Transforming growth factor ß activation by the αvß6 integrin is central to the pathogenesis of idiopathic pulmonary fibrosis. Expression of the αvß6 integrin is increased in fibrotic lung tissue and is a promising therapeutic target for treatment of the disease. Currently, measurement of αvß6 integrin levels in the lung requires immunohistochemical analysis of biopsy samples. This procedure is clinically impractical for many patients with pulmonary fibrosis, and a noninvasive strategy for measuring αvß6 integrin levels in the lungs is urgently required to facilitate monitoring of disease progression and therapeutic responses. METHODS: Using a murine model of bleomycin-induced lung injury, we assessed the binding of intravenously administered (111)In-labeled αvß6-specific (diethylenetriamine pentaacetate-tetra [DTPA]-A20FMDV2) or control (DTPA-A20FMDVran) peptide by nanoSPECT/CT imaging. Development of fibrosis was assessed by lung hydroxyproline content, and αvß6 protein and itgb6 messenger RNA were measured in the lungs. RESULTS: Maximal binding of (111)In-labeled A20FMDV2 peptide to αvß6 integrins was detected in the lungs 1 h after intravenous administration. No significant binding was detected in mice injected with control peptide. Integrin binding was increased in the lungs of bleomycin-, compared with saline-, exposed mice and was attenuated by pretreatment with αvß6-blocking antibodies. Levels of (111)In-labeled A20FMDV2 peptide correlated positively with hydroxyproline, αvß6 protein, and itgb6 messenger RNA levels. CONCLUSION: We have developed a highly sensitive, quantifiable, and noninvasive technique for measuring αvß6 integrin levels within the lung. Measurement of αvß6 integrins by SPECT/CT scanning has the potential for use in stratifying therapy for patients with pulmonary fibrosis.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Fibrosis Pulmonar Idiopática/diagnóstico por imagen , Integrinas/metabolismo , Imagen Multimodal , Tomografía Computarizada de Emisión de Fotón Único , Tomografía Computarizada por Rayos X , Animales , Antígenos de Neoplasias/genética , Biomarcadores/metabolismo , Determinación de Punto Final , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Radioisótopos de Indio , Integrinas/genética , Pulmón/diagnóstico por imagen , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Péptidos , Pronóstico , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal
14.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...