Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Respir Res ; 24(1): 89, 2023 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-36949463

RESUMO

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a devastating interstitial lung disease (ILD) with limited treatment options. Interleukin-33 (IL-33) is proposed to play a role in the development of IPF however the exclusive use of prophylactic dosing regimens means that the therapeutic benefit of targeting this cytokine in IPF is unclear. METHODS: IL-33 expression was assessed in ILD lung sections and human lung fibroblasts (HLFs) by immunohistochemistry and gene/protein expression and responses of HLFs to IL-33 stimulation measured by qPCR. In vivo, the fibrotic potential of IL-33:ST2 signalling was assessed using a murine model of bleomycin (BLM)-induced pulmonary fibrosis and therapeutic dosing with an ST2-Fc fusion protein. Lung and bronchoalveolar lavage fluid were collected for measurement of inflammatory and fibrotic endpoints. Human precision-cut lung slices (PCLS) were stimulated with transforming growth factor-ß (TGFß) or IL-33 and fibrotic readouts assessed. RESULTS: IL-33 was expressed by fibrotic fibroblasts in situ and was increased by TGFß treatment in vitro. IL-33 treatment of HLFs did not induce IL6, CXCL8, ACTA2 and COL1A1 mRNA expression with these cells found to lack the IL-33 receptor ST2. Similarly, IL-33 stimulation had no effect on ACTA2, COL1A1, FN1 and fibronectin expression by PCLS. Despite having effects on inflammation suggestive of target engagement, therapeutic dosing with the ST2-Fc fusion protein failed to reduce BLM-induced fibrosis measured by hydroxyproline content or Ashcroft score. CONCLUSIONS: Together these findings suggest the IL-33:ST2 axis does not play a central fibrogenic role in the lungs with therapeutic blockade of this pathway unlikely to surpass the current standard of care for IPF.


Assuntos
Fibrose Pulmonar Idiopática , Interleucina-33 , Animais , Humanos , Camundongos , Bleomicina/toxicidade , Fibroblastos/metabolismo , Fibrose Pulmonar Idiopática/induzido quimicamente , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/metabolismo , Proteína 1 Semelhante a Receptor de Interleucina-1/genética , Interleucina-33/metabolismo , Pulmão/metabolismo , Camundongos Endogâmicos C57BL , Fator de Crescimento Transformador beta/metabolismo
2.
J Biol Chem ; 291(18): 9540-53, 2016 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-26861876

RESUMO

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.


Assuntos
Antígenos de Neoplasias/biossíntese , Regulação da Expressão Gênica , Integrinas/biossíntese , Fibrose Pulmonar/metabolismo , Transdução de Sinais , Transcrição Gênica , Proteínas Elk-1 do Domínio ets/metabolismo , Animais , Antígenos de Neoplasias/genética , Linhagem Celular Transformada , Humanos , Integrinas/genética , Camundongos , Camundongos Knockout , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/genética , Fibrose Pulmonar/patologia , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Proteínas Elk-1 do Domínio ets/genética
3.
Lab Invest ; 96(6): 623-31, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26974397

RESUMO

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.


Assuntos
Fibrose Pulmonar Idiopática/etiologia , Lesão Pulmonar/etiologia , Inibidor Secretado de Peptidases Leucocitárias/deficiência , Animais , Bleomicina/toxicidade , Colágeno/genética , Colágeno/metabolismo , Deleção de Genes , Hidroxiprolina/metabolismo , Fibrose Pulmonar Idiopática/genética , Fibrose Pulmonar Idiopática/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Lesão Pulmonar/genética , Lesão Pulmonar/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Inibidor Secretado de Peptidases Leucocitárias/genética , Fator de Crescimento Transformador beta/metabolismo
4.
Magn Reson Med ; 76(4): 1224-35, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-26507239

RESUMO

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.


Assuntos
Asma/diagnóstico por imagem , Asma/fisiopatologia , Pulmão/diagnóstico por imagem , Pulmão/fisiopatologia , Imageamento por Ressonância Magnética/métodos , Troca Gasosa Pulmonar , Isótopos de Xenônio/farmacocinética , Administração por Inalação , Animais , Simulação por Computador , Aumento da Imagem/métodos , Interpretação de Imagem Assistida por Computador/métodos , Masculino , Modelos Biológicos , Imagem Molecular/métodos , Compostos Radiofarmacêuticos/administração & dosagem , Compostos Radiofarmacêuticos/farmacocinética , Ratos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Distribuição Tecidual , Isótopos de Xenônio/administração & dosagem
5.
J Biol Chem ; 289(51): 35246-63, 2014 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-25339175

RESUMO

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.


Assuntos
Antígenos de Neoplasias/metabolismo , Colágeno/metabolismo , Células Epiteliais/metabolismo , Integrinas/metabolismo , Infecções por Orthomyxoviridae/metabolismo , Fator de Crescimento Transformador beta/metabolismo , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Animais , Antígenos de Neoplasias/genética , Antivirais/farmacologia , Apoptose , Linhagem Celular Transformada , Cães , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Interações Hospedeiro-Patógeno , Humanos , Immunoblotting , Vírus da Influenza A Subtipo H1N1/fisiologia , Integrinas/genética , Pulmão/metabolismo , Pulmão/virologia , Células Madin Darby de Rim Canino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/virologia , Fosforilação/efeitos dos fármacos , Poli I-C/farmacologia , Proteína Smad3/genética , Proteína Smad3/metabolismo , Receptor 3 Toll-Like/metabolismo , Fator de Crescimento Transformador beta/genética , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/metabolismo
6.
Eur Respir J ; 46(2): 486-94, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25745053

RESUMO

Idiopathic pulmonary fibrosis (IPF) and fibrotic nonspecific interstitial pneumonitis are progressive interstitial lung diseases (ILDs) with limited treatment options and poor survival. However, the rate of disease progression is variable, implying there may be different endotypes of disease. We hypothesised that immunophenotyping biopsies from ILD patients might reveal distinct endotypes of progressive fibrotic disease, which may facilitate stratification when undertaking clinical trials of novel therapies for IPF.43 paraffin-embedded, formalin-fixed lung tissue sections were immunostained for five molecules implicated in the pathogenesis of the fibrosis: α-smooth muscle actin (αSMA), αvß6 integrin, pro-surfactant protein C (SP-C), hepatocyte growth factor (HGF) and tenascin-C (TenC). Levels of immunostaining and numbers of fibroblastic foci were quantified using operator-dependent and -independent methods. The relationship of all these markers to overall survival was analysed.Staining revealed high levels of αSMA, αvß6 integrin, pro-SP-C, HGF and TenC, and fibroblastic foci. Immunostaining varied across samples for all molecules but only the extent of αvß6 integrin immunostaining was associated with increased mortality. There was no association with the other markers measured.Our data suggest high levels of αvß6 integrin may identify a specific endotype of progressive fibrotic lung disease.


Assuntos
Antígenos de Neoplasias/metabolismo , Integrinas/metabolismo , Doenças Pulmonares Intersticiais/patologia , Pulmão/patologia , Adulto , Idoso , Biomarcadores/metabolismo , Biópsia , Feminino , Humanos , Doenças Pulmonares Intersticiais/cirurgia , Masculino , Pessoa de Meia-Idade , Prognóstico
7.
Lancet Respir Med ; 12(9): 681-692, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39025091

RESUMO

BACKGROUND: Pulmonary fibrosis results from alveolar injury, leading to extracellular matrix remodelling and impaired lung function. This study aimed to classify patients with pulmonary fibrosis according to blood biomarkers to differentiate distinct disease patterns, known as endotypes. METHODS: In this cluster analysis, we first classified patients from the PROFILE study, a multicentre, prospective, observational cohort of individuals with incident idiopathic pulmonary fibrosis or non-specific interstitial pneumonia in the UK (Nottingham University Hospitals, Nottingham; and Royal Brompton Hospital, London). 13 blood biomarkers representing extracellular matrix remodelling, epithelial stress, and thrombosis were measured by ELISA in the PROFILE study. We classified patients by unsupervised consensus clustering. To evaluate generalisability, a machine learning classifier trained on biomarker signatures derived from consensus clustering was applied to a replication dataset from the Australian Idiopathic Pulmonary Fibrosis Registry (AIPFR). Biomarker associations with mortality and change in percentage of predicted forced vital capacity (FVC%) were assessed, adjusting for age, gender, baseline FVC%, and antifibrotic treatment and steroid treatment before and after baseline. Mortality risk associated with the clusters in the PROFILE cohort was evaluated with Cox proportional hazards models, and mixed-effects models were used to analyse how clustering was associated with longitudinal FVC% in the PROFILE and AIPFR cohorts. FINDINGS: 455 of 580 participants from the PROFILE study (348 [76%] men and 107 [24%] women; mean age 72·4 years [SD 8·3]) were included in the analysis. Within this group, three clusters were identified based on blood biomarkers. A basement membrane collagen (BM) cluster (n=248 [55%]) showed high concentrations of PRO-C4, PRO-C28, C3M, and C6M, whereas an epithelial injury (EI) cluster (n=109 [24%]) showed high concentrations of MMP-7, SP-D, CYFRA211, CA19-9, and CA-125. The third cluster (crosslinked fibrin [XF] cluster; n=98 [22%]) had high concentrations of X-FIB. In the replication dataset (117 of 833 patients from AIPFR; 87 [74%] men and 30 [26%] women; mean age 72·9 years [SD 7·9]), we identified the same three clusters (BM cluster, n=93 [79%]; EI cluster, n=8 [7%]; XF cluster, n=16 [14%]). These clusters showed similarities with clusters in the PROFILE dataset regarding blood biomarkers and phenotypic signatures. In the PROFILE dataset, the EI and XF clusters were associated with increased mortality risk compared with the BM cluster (EI vs BM: adjusted hazard ratio [HR] 1·88 [95% CI 1·42-2·49], p<0·0001; XF vs BM: adjusted HR 1·53 [1·13-2·06], p=0·0058). The EI cluster showed the greatest annual FVC% decline, followed by the BM and XF clusters. A similar FVC% decline pattern was observed in these clusters in the AIPFR replication dataset. INTERPRETATION: Blood biomarker clustering in pulmonary fibrosis identified three distinct blood biomarker signatures associated with lung function and prognosis, suggesting unique pulmonary fibrosis biomarker patterns. These findings support the presence of pulmonary fibrosis endotypes with the potential to guide targeted therapy development. FUNDING: None.


Assuntos
Biomarcadores , Fibrose Pulmonar Idiopática , Humanos , Biomarcadores/sangue , Masculino , Feminino , Estudos Prospectivos , Idoso , Análise por Conglomerados , Fibrose Pulmonar Idiopática/sangue , Fibrose Pulmonar Idiopática/mortalidade , Fibrose Pulmonar Idiopática/diagnóstico , Fibrose Pulmonar Idiopática/fisiopatologia , Pessoa de Meia-Idade , Capacidade Vital , Reino Unido
8.
Lancet Digit Health ; 4(12): e862-e872, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36333179

RESUMO

BACKGROUND: Idiopathic pulmonary fibrosis is a progressive fibrotic lung disease with a variable clinical trajectory. Decline in forced vital capacity (FVC) is the main indicator of progression; however, missingness prevents long-term analysis of patterns in lung function. We aimed to identify distinct clusters of lung function trajectory among patients with idiopathic pulmonary fibrosis using machine learning techniques. METHODS: We did a secondary analysis of longitudinal data on FVC collected from a cohort of patients with idiopathic pulmonary fibrosis from the PROFILE study; a multicentre, prospective, observational cohort study. We evaluated the imputation performance of conventional and machine learning techniques to impute missing data and then analysed the fully imputed dataset by unsupervised clustering using self-organising maps. We compared anthropometric features, genomic associations, serum biomarkers, and clinical outcomes between clusters. We also performed a replication of the analysis on data from a cohort of patients with idiopathic pulmonary fibrosis from an independent dataset, obtained from the Chicago Consortium. FINDINGS: 415 (71%) of 581 participants recruited into the PROFILE study were eligible for further analysis. An unsupervised machine learning algorithm had the lowest imputation error among tested methods, and self-organising maps identified four distinct clusters (1-4), which was confirmed by sensitivity analysis. Cluster 1 comprised 140 (34%) participants and was associated with a disease trajectory showing a linear decline in FVC over 3 years. Cluster 2 comprised 100 (24%) participants and was associated with a trajectory showing an initial improvement in FVC before subsequently decreasing. Cluster 3 comprised 113 (27%) participants and was associated with a trajectory showing an initial decline in FVC before subsequent stabilisation. Cluster 4 comprised 62 (15%) participants and was associated with a trajectory showing stable lung function. Median survival was shortest in cluster 1 (2·87 years [IQR 2·29-3·40]) and cluster 3 (2·23 years [1·75-3·84]), followed by cluster 2 (4·74 years [3·96-5·73]), and was longest in cluster 4 (5·56 years [5·18-6·62]). Baseline FEV1 to FVC ratio and concentrations of the biomarker SP-D were significantly higher in clusters 1 and 3. Similar lung function clusters with some shared anthropometric features were identified in the replication cohort. INTERPRETATION: Using a data-driven unsupervised approach, we identified four clusters of lung function trajectory with distinct clinical and biochemical features. Enriching or stratifying longitudinal spirometric data into clusters might optimise evaluation of intervention efficacy during clinical trials and patient management. FUNDING: National Institute for Health and Care Research, Medical Research Council, and GlaxoSmithKline.


Assuntos
Fibrose Pulmonar Idiopática , Humanos , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/genética , Estudos Prospectivos , Capacidade Vital , Estudos de Coortes , Biomarcadores
9.
Immunology ; 134(1): 60-72, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21718315

RESUMO

Interleukin-23 (IL-23) is important for T helper type 17 (Th17) responses and strategies to regulate IL-23 in human dendritic cells (DC) are limited. This study describes a novel means to control IL-23 secretion by conditioning DC with a phosphatidyl inositol 3-kinase inhibitor Wortmannin (WM). Treatment of monocyte-derived DC with WM increased Toll-like receptor (TLR) -dependent IL-23 secretion 10-fold and IL-12p70 twofold, but IL-27 was unaffected. The effect of WM was restricted to TLR3/4 pathways, did not occur through TLR2, TLR7/8 or Dectin-1, and was characterized by increased p19, p35 and p40 transcription. These responses were not solely dependent on phosphatidyl inositol 3-kinase as the alternative inhibitor LY294002 did not modulate IL-23 production. The normal patterns of activation of mitogen-activated protein kinase pathways were unaffected by WM-conditioning but IL-23 secretion required p38, ERK and JNK pathways. Importantly, this effect was manifest in populations of blood DC. Conditioning freshly isolated myeloid DC with WM before TLR3 or TLR4 triggering resulted in high levels of IL-23 secretion and an absence of IL-12p70. These WM-conditioned myeloid DC were highly effective at priming Th17 responses from naive CD4(+) T cells. Our findings provide a novel means to generate IL-23-rich environments and Th17 responses and suggest as yet unidentified regulatory factors, identification of which will provide new approaches to control IL-23-dependent immunity in infectious disease, autoimmunity and malignancy.


Assuntos
Androstadienos/farmacologia , Células Dendríticas/metabolismo , Interleucina-23/metabolismo , Células Th17/imunologia , Regulação para Cima/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Diferenciação Celular/imunologia , Sobrevivência Celular/efeitos dos fármacos , Cromonas/farmacologia , Técnicas de Cocultura , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Humanos , Interferon gama/farmacologia , Interleucina-12/metabolismo , Subunidade p35 da Interleucina-12/genética , Subunidade p40 da Interleucina-12/genética , Interleucina-17/metabolismo , Interleucina-1beta/metabolismo , Subunidade p19 da Interleucina-23/genética , Interleucina-6/metabolismo , Interleucinas/metabolismo , Lipopolissacarídeos/farmacologia , Ativação Linfocitária/imunologia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Morfolinas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células Th17/metabolismo , Receptor 3 Toll-Like/agonistas , Receptor 3 Toll-Like/metabolismo , Receptor 4 Toll-Like/agonistas , Receptor 4 Toll-Like/metabolismo , Receptores Toll-Like/agonistas , Receptores Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Wortmanina
10.
Methods Mol Biol ; 2299: 99-108, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34028736

RESUMO

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.


Assuntos
Miofibroblastos/citologia , Receptores de Vitronectina/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Técnicas de Cultura de Células , Diferenciação Celular , Células Cultivadas , Proteínas da Matriz Extracelular/metabolismo , Humanos , Miofibroblastos/metabolismo , Transdução de Sinais
11.
Am J Pathol ; 174(4): 1264-79, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19147812

RESUMO

Activation of latent transforming growth factor beta (TGF-beta) by alphavbeta6 integrin is critical in the pathogenesis of lung injury and fibrosis. We have previously demonstrated that the stimulation of protease activated receptor 1 promotes alphavbeta6 integrin-mediated TGF-beta activation via RhoA, which is known to modulate cell contraction. However, whether other G protein-coupled receptors can also induce alphavbeta6 integrin-mediated TGF-beta activation is unknown; in addition, the alphavbeta6 integrin signaling pathway has not yet been fully characterized. In this study, we show that lysophosphatidic acid (LPA) induces alphavbeta6-mediated TGF-beta activation in human epithelial cells via both RhoA and Rho kinase. Furthermore, we demonstrate that LPA-induced alphavbeta6 integrin-mediated TGF-beta activity is mediated via the LPA2 receptor, which signals via G alpha(q). Finally, we show that the expression levels of both the LPA2 receptor and alphavbeta6 integrin are up-regulated and are spatially and temporally associated following bleomycin-induced lung injury. Furthermore, both the LPA2 receptor and alphavbeta6 integrin are up-regulated in the overlying epithelial areas of fibrosis in patients with usual interstitial pneumonia. These studies demonstrate that LPA induces alphavbeta6 integrin-mediated TGF-beta activation in epithelial cells via LPA2, G alpha(q), RhoA, and Rho kinase, and that this pathway might be clinically relevant to the development of lung injury and fibrosis.


Assuntos
Antígenos de Neoplasias/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Integrinas/metabolismo , Lesão Pulmonar/metabolismo , Lisofosfolipídeos/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Western Blotting , Técnicas de Cocultura , Fibrose , Citometria de Fluxo , Imunofluorescência , Humanos , Imuno-Histoquímica , Pulmão/metabolismo , Pulmão/patologia , Lesão Pulmonar/fisiopatologia , Lisofosfolipídeos/farmacologia , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
12.
Biochem Biophys Res Commun ; 370(2): 239-42, 2008 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-18359288

RESUMO

Transforming growth factor beta (TGFbeta) is a key remodelling factor in asthma. It is produced as a latent complex and the main limiting step in TGFbeta bioavailability is its activation. Mast cell tryptase has been shown to stimulate the release of functionally active TGFbeta from human airway smooth muscle (ASM) cells [P. Berger, P.O. Girodet, H. Begueret, O. Ousova, D.W. Perng, R. Marthan, A.F. Walls, J.M. Tunon de Lara, Tryptase-stimulated human airway smooth muscle cells induce cytokine synthesis and mast cell chemotaxis, FASEB J. 17 (2003) 2139-2141]. The aim of this study was to determine if tryptase could cause TGFbeta activation as well as expression in ASM cells via its receptor, proteinase-activated receptor 2 (PAR2). Tryptase caused TGFbeta activation without affecting levels of total TGFbeta. This effect was inhibited by the selective tryptase inhibitor FUT175 and leupeptin but not mimicked by the PAR2 activating peptide SLIGKV-NH(2). Furthermore, the ASM cells used in the study did not express PAR2. The results indicate that tryptase activates TGFbeta via a PAR2-independent proteolytic mechanism in human ASM cells and may help understanding the role of tryptase in asthma.


Assuntos
Brônquios/enzimologia , Miócitos de Músculo Liso/enzimologia , Fator de Crescimento Transformador beta/metabolismo , Triptases/metabolismo , Benzamidinas , Brônquios/citologia , Guanidinas/farmacologia , Humanos , Leupeptinas/farmacologia , Oligopeptídeos/farmacologia , Receptor PAR-2/metabolismo , Inibidores da Tripsina/farmacologia , Triptases/antagonistas & inibidores
13.
Methods Mol Biol ; 1627: 351-365, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28836213

RESUMO

The potent and pluripotent cytokine TGFß has important roles in normal homeostasis and disease pathogenesis. Once released from cells, TGFß exists in both latent and functionally active forms. Large amounts of latent TGFß are secreted from cells and sequestered in extracellular matrix, only a small proportion of which is activated at any given time. Accurate assessment of TGFß activity levels is an important measurement in biological research and requires methods distinct from measuring total levels of TGFß expression as small changes in TGFß activity levels could be masked by the large amounts of latent TGFß available to be measured. In this chapter, we describe detailed experimental methods for assessing levels of active TGFß in cells and tissues. This chapter includes methods for the assessment of TGFß activity in cells in vitro, in ex vivo precision cut tissue, and in vivo.


Assuntos
Bioensaio , Fator de Crescimento Transformador beta/metabolismo , Animais , Bioensaio/métodos , Líquido da Lavagem Broncoalveolar/citologia , Linhagem Celular Transformada , Células Cultivadas , Meios de Cultivo Condicionados/metabolismo , Imuno-Histoquímica , Integrinas/metabolismo , Pulmão/citologia , Pulmão/metabolismo , Vison , Fosforilação , Transdução de Sinais , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta/química
14.
Lancet Respir Med ; 5(12): 946-955, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29150411

RESUMO

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.


Assuntos
Antígeno Ca-125/sangue , Antígeno CA-19-9/sangue , Progressão da Doença , Fibrose Pulmonar Idiopática/sangue , Metaloproteinase 7 da Matriz/sangue , Proteína D Associada a Surfactante Pulmonar/sangue , Biomarcadores/sangue , Estudos de Casos e Controles , Ensaio de Imunoadsorção Enzimática , Epitélio/metabolismo , Epitélio/patologia , Feminino , Humanos , Fibrose Pulmonar Idiopática/mortalidade , Estudos Longitudinais , Pulmão/metabolismo , Pulmão/patologia , Masculino , Valor Preditivo dos Testes , Estudos Prospectivos
15.
Lancet Respir Med ; 5(11): 869-880, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29066090

RESUMO

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a chronic progressive lung disease with high mortality, uncertain cause, and few treatment options. Studies have identified a significant genetic risk associated with the development of IPF; however, mechanisms by which genetic risk factors promote IPF remain unclear. We aimed to identify genetic variants associated with IPF susceptibility and provide mechanistic insight using gene and protein expression analyses. METHODS: We used a two-stage approach: a genome-wide association study in patients with IPF of European ancestry recruited from nine different centres in the UK and controls selected from UK Biobank (stage 1) matched for age, sex, and smoking status; and a follow-up of associated genetic variants in independent datasets of patients with IPF and controls from two independent US samples from the Chicago consortium and the Colorado consortium (stage 2). We investigated the effect of novel signals on gene expression in large transcriptomic and genomic data resources, and examined expression using lung tissue samples from patients with IPF and controls. FINDINGS: 602 patients with IPF and 3366 controls were selected for stage 1. For stage 2, 2158 patients with IPF and 5195 controls were selected. We identified a novel genome-wide significant signal of association with IPF susceptibility near A-kinase anchoring protein 13 (AKAP13; rs62025270, odds ratio [OR] 1·27 [95% CI 1·18-1·37], p=1·32 × 10-9) and confirmed previously reported signals, including in mucin 5B (MUC5B; rs35705950, OR 2·89 [2·56-3·26], p=1·12 × 10-66) and desmoplakin (DSP; rs2076295, OR 1·44 [1·35-1·54], p=7·81 × 10-28). For rs62025270, the allele A associated with increased susceptibility to IPF was also associated with increased expression of AKAP13 mRNA in lung tissue from patients who had lung resection procedures (n=1111). We showed that AKAP13 is expressed in the alveolar epithelium and lymphoid follicles from patients with IPF, and AKAP13 mRNA expression was 1·42-times higher in lung tissue from patients with IPF (n=46) than that in lung tissue from controls (n=51). INTERPRETATION: AKAP13 is a Rho guanine nucleotide exchange factor regulating activation of RhoA, which is known to be involved in profibrotic signalling pathways. The identification of AKAP13 as a susceptibility gene for IPF increases the prospect of successfully targeting RhoA pathway inhibitors in patients with IPF. FUNDING: UK Medical Research Council, National Heart, Lung, and Blood Institute of the US National Institutes of Health, Agencia Canaria de Investigación, Innovación y Sociedad de la Información, Spain, UK National Institute for Health Research, and the British Lung Foundation.


Assuntos
Proteínas de Ancoragem à Quinase A/genética , Predisposição Genética para Doença/genética , Variação Genética , Fibrose Pulmonar Idiopática/genética , Antígenos de Histocompatibilidade Menor/genética , Proteínas Proto-Oncogênicas/genética , População Branca/genética , Idoso , Células Epiteliais Alveolares/metabolismo , Estudos de Casos e Controles , Europa (Continente) , Feminino , Estudo de Associação Genômica Ampla , Humanos , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/fisiologia , Transdução de Sinais/genética , Estruturas Linfoides Terciárias/genética , Proteína rhoA de Ligação ao GTP/fisiologia
16.
PLoS One ; 11(8): e0158047, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27494713

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a devastating, progressive disease with poor survival rates and limited treatment options. Upregulation of αvß6 integrins within the alveolar epithelial cells is a characteristic feature of IPF and correlates with poor patient survival. The pro-fibrotic cytokine TGFß1 can upregulate αvß6 integrin expression but the molecular mechanisms driving this effect have not previously been elucidated. We confirm that stimulation with exogenous TGFß1 increases expression of the integrin ß6 subunit gene (ITGB6) and αvß6 integrin cell surface expression in a time- and concentration-dependent manner. TGFß1-induced ITGB6 expression occurs via transcriptional activation of the ITGB6 gene, but does not result from effects on ITGB6 mRNA stability. Basal expression of ITGB6 in, and αvß6 integrins on, lung epithelial cells occurs via homeostatic αvß6-mediated TGFß1 activation in the absence of exogenous stimulation, and can be amplified by TGFß1 activation. Fundamentally, we show for the first time that TGFß1-induced ITGB6 expression occurs via canonical Smad signalling since dominant negative constructs directed against Smad3 and 4 inhibit ITGB6 transcriptional activity. Furthermore, disruption of a Smad binding site at -798 in the ITGB6 promoter abolishes TGFß1-induced ITGB6 transcriptional activity. Using chromatin immunoprecipitation we demonstrate that TGFß1 stimulation of lung epithelial cells results in direct binding of Smad3, and Smad4, to the ITGB6 gene promoter within this region. Finally, using an adenoviral TGFß1 over-expression model of pulmonary fibrosis we demonstrate that Smad3 is crucial for TGFß1-induced αvß6 integrin expression within the alveolar epithelium in vivo. Together, these data confirm that a homeostatic, autocrine loop of αvß6 integrin activated TGFß1-induced ITGB6 gene expression regulates epithelial basal αvß6 integrin expression, and demonstrates that this occurs via Smad-dependent transcriptional regulation at a single Smad binding site in the promoter of the ß6 subunit gene. Active TGFß1 amplifies this pathway both in vitro and in vivo, which may promote fibrosis.


Assuntos
Fibrose Pulmonar Idiopática/patologia , Cadeias beta de Integrinas/metabolismo , Transcrição Gênica/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Sítios de Ligação , Células Cultivadas , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Fibrose Pulmonar Idiopática/metabolismo , Cadeias beta de Integrinas/genética , Integrinas/genética , Integrinas/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Knockout , Mutagênese Sítio-Dirigida , Regiões Promotoras Genéticas , Estabilidade de RNA/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Proteína Smad3/genética , Proteína Smad3/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
17.
Sci Signal ; 9(451): ra104, 2016 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-27811142

RESUMO

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.


Assuntos
Enfisema/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Interleucina-33/metabolismo , Macrófagos Alveolares/metabolismo , Mucosa Respiratória/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais , Enfisema/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Interleucina-33/genética , Metaloproteinase 12 da Matriz/genética , Metaloproteinase 12 da Matriz/metabolismo , Camundongos , Camundongos Transgênicos , Mucosa Respiratória/patologia , Fator de Crescimento Transformador beta/genética , Lesão Pulmonar Induzida por Ventilação Mecânica/genética , Lesão Pulmonar Induzida por Ventilação Mecânica/metabolismo
18.
Pharmacol Res Perspect ; 2(4): e00030, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25505594

RESUMO

Transforming growth factor-ß (TGF-ß) plays an important role in the development of tissue fibrosis, and molecules inhibiting this pathway are attractive therapeutic targets for fibrotic diseases such as idiopathic pulmonary fibrosis (IPF). Activation of TGF-ß is the rate-limiting step in TGF-ß bioavailability, and activation by the αVß6 integrin is important in fibrosis of the lung, liver, and kidney. Activation of TGF-ß by αVß6 requires direct cell-cell contact and measurable release of active TGF-ß in extracellular fluid compartments does not reflect tissue specific activation. The aim of this study was to determine the effect of antifibrotic compounds on both total, and specific αVß6 integrin-mediated TGF-ß activity. Using a transformed mink lung cell (TMLC) TGF-ß reporter, the effects of potential antifibrotic therapies including an activin-like kinase (Alk5) inhibitor, Dexamethasone, Pirfenidone, N-acetylcysteine (NAC), and BIBF1120 were assessed. Effects due to αVß6 integrin-mediated TGF-ß activity were measured using reporter cells cocultured with cells expressing αVß6 integrins. These high-throughput studies were validated using a phosphorylated Smad2 Enzyme-Linked Immunosorbent Assay. Alk5 inhibitors are potent inhibitors of TGF-ß activity, whereas the novel antifibrotics, Pirfenidone, BIBF1120, and NAC are only moderate inhibitors, and Dexamethasone does not specifically affect TGF-ßactivity, but inhibits TGF-ß-induced gene expression. None of the current small molecular inhibitors inhibit αVß6-mediated TGF-ß activity. These results demonstrate the potential of this high-throughput assay of αVß6-specific TGF-ß activity and illustrate that currently available antifibrotics have limited effects on αVß6 integrin-mediated TGF-ß activity.

19.
Eur Cytokine Netw ; 21(4): 319-28, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21112826

RESUMO

Mitogen-activated protein kinases (MAPK) are targets for the immune-modulation of dendritic cells (DC). However, our knowledge of their role in the regulation of IL-12-family cytokines is limited. This study investigated the roles of p38, JNK, p44/42 and PI3K pathways in IL-12/23/27 production by human DC, and their impact on naïve T(H)-responses. We first identified TOP and UBC as robust DC housekeeping genes. Peak transcription of p35 and p40 occurred by 12h, p19 and p28 by 8h and EBI3 by 12-24h. Using selective antagonists, we showed that p38 was a positive regulator of IL-12, 23 and 27, JNK positively regulated IL-12 and IL-27, and inhibition of MEK1/2 had no marked effect. In contrast, the PI3K pathway markedly attenuated IL-23 responses and, to a lesser extent, IL-12, but not IL-27. To identify the role of these soluble factors, we co-stimulated naïve CD4+ T-cells in the presence of DC supernatant. The presence of mature DC supernatant induced not only strong IFNγ responses, but also IL-10 and IL-17A. Inhibition of p38 ablated T(H1), and IL-10 and IL-17A responses, whilst modestly enhancing IL-5 secretion. In contrast, inhibition of MEK1/2 abolished IL-17A production, whilst leaving other responses unaffected, whereas inhibition of JNK or PI3K had no discernable effect. In summary, we describe the expression of IL-12-family cytokines from DC and propose a modified model for their regulation. This study further clarifies the potential for therapeutic modulation through these mediators.


Assuntos
Células Dendríticas/metabolismo , Regulação da Expressão Gênica , Interleucina-12/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Células Cultivadas , Células Dendríticas/enzimologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-12/genética , Interleucina-23/genética , Interleucina-23/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA