Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Life Sci Alliance ; 6(8)2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37230801

RESUMEN

Repeated injury of the lung epithelium is proposed to be the main driver of idiopathic pulmonary fibrosis (IPF). However, available therapies do not specifically target the epithelium and human models of fibrotic epithelial damage with suitability for drug discovery are lacking. We developed a model of the aberrant epithelial reprogramming observed in IPF using alveolar organoids derived from human-induced pluripotent stem cells stimulated with a cocktail of pro-fibrotic and inflammatory cytokines. Deconvolution of RNA-seq data of alveolar organoids indicated that the fibrosis cocktail rapidly increased the proportion of transitional cell types including the KRT5 - /KRT17 + aberrant basaloid phenotype recently identified in the lungs of IPF patients. We found that epithelial reprogramming and extracellular matrix (ECM) production persisted after removal of the fibrosis cocktail. We evaluated the effect of the two clinically approved compounds for IPF, nintedanib and pirfenidone, and found that they reduced the expression of ECM and pro-fibrotic mediators but did not completely reverse epithelial reprogramming. Thus, our system recapitulates key aspects of IPF and is a promising system for drug discovery.


Asunto(s)
Fibrosis Pulmonar Idiopática , Células Madre Pluripotentes , Humanos , Células Epiteliales Alveolares/metabolismo , Pulmón/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis , Células Madre Pluripotentes/metabolismo , Organoides/metabolismo
2.
Respir Res ; 24(1): 51, 2023 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-36788603

RESUMEN

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a chronic lung disease that affects 3 million people worldwide. Senescence and small extracellular vesicles (sEVs) have been implicated in the pathogenesis of IPF, although how sEVs promote disease remains unclear. Here, we profile sEVs from bronchial epithelial cells and determine small RNA (smRNA) content. METHODS: Conditioned media was collected and sEVs were isolated from normal human bronchial epithelial cells (NHBEs) and IPF-diseased human bronchial epithelial cells (DHBEs). RESULTS: Increased sEV release from DHBEs compared to NHBEs (n = 4; p < 0.05) was detected by nanoparticle tracking analysis. NHBEs co-cultured with DHBE-derived sEVs for 72 h expressed higher levels of SA-ß-Gal and γH2AX protein, p16 and p21 RNA and increased secretion of IL6 and IL8 proteins (all n = 6-8; p < 0.05). sEVs were also co-cultured with healthy air-liquid interface (ALI) cultures and similar results were observed, with increases in p21 and p16 gene expression and IL6 and IL8 (basal and apical) secretion (n = 6; p < 0.05). Transepithelial electrical resistance (TEER) measurements, a reflection of epithelial barrier integrity, were decreased upon the addition of DHBE-derived sEVs (n = 6; p < 0.05). smRNA-sequencing identified nineteen significantly differentially expressed miRNA in DHBE-derived sEVs compared to NHBE-derived sEVs, with candidate miRNAs validated by qPCR (all n = 5; p < 0.05). Four of these miRNAs were upregulated in NHBEs co-cultured with DHBE-derived sEVs and three in healthy ALI cultures co-cultured with DHBE-derived sEVs (n = 3-4; p < 0.05). CONCLUSIONS: This data demonstrates that DHBE-derived sEVs transfer senescence to neighbouring healthy cells, promoting the disease state in IPF.


Asunto(s)
Vesículas Extracelulares , Fibrosis Pulmonar Idiopática , MicroARNs , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Vesículas Extracelulares/metabolismo
3.
Stem Cell Reports ; 17(12): 2718-2731, 2022 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-36460000

RESUMEN

The respiratory epithelium consists of multiple, functionally distinct cell types and is maintained by regionally specific progenitor populations that repair the epithelium following injury. Several in vitro methods exist for studying lung epithelial repair using primary murine lung cells, but isolation methods are hampered by a lack of surface markers distinguishing epithelial progenitors along the respiratory epithelium. Here, we developed a 3D printed lobe divider (3DLD) to aid in simultaneous isolation of proximal versus distal lung epithelial progenitors from individual mice that give rise to differentiated epithelia in multiple in vitro assays. In contrast to 3DLD-isolated distal progenitor cells, commonly used manual tracheal ligation methods followed by lobe removal resulted in co-isolation of rare proximal cells with distal cells, which altered the transcriptional landscape and size distribution of distal organoids. The 3DLD aids in reproducible isolation of distal versus proximal progenitor populations and minimizes the potential for contaminating populations to confound in vitro assays.


Asunto(s)
Células Epiteliales , Células Madre , Ratones , Animales , Células Epiteliales/metabolismo , Pulmón , Separación Celular , Diferenciación Celular , Impresión Tridimensional
4.
JCI Insight ; 7(16)2022 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-35993367

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a chronic disease of unmet medical need. It is characterized by formation of scar tissue leading to a progressive and irreversible decline in lung function. IPF is associated with repeated injury, which may alter the composition of the extracellular matrix (ECM). Here, we demonstrate that IPF patient-derived pulmonary ECM drives profibrotic response in normal human lung fibroblasts (NHLF) in a 3D spheroid assay. Next, we reveal distinct alterations in composition of the diseased ECM, identifying potentially novel associations with IPF. Growth differentiation factor 15 (GDF15) was identified among the most significantly upregulated proteins in the IPF lung-derived ECM. In vivo, GDF15 neutralization in a bleomycin-induced lung fibrosis model led to significantly less fibrosis. In vitro, recombinant GDF15 (rGDF15) stimulated α smooth muscle actin (αSMA) expression in NHLF, and this was mediated by the activin receptor-like kinase 5 (ALK5) receptor. Furthermore, in the presence of rGDF15, the migration of NHLF in collagen gel was reduced. In addition, we observed a cell type-dependent effect of GDF15 on the expression of cell senescence markers. Our data suggest that GDF15 mediates lung fibrosis through fibroblast activation and differentiation, implicating a potential direct role of this matrix-associated cytokine in promoting aberrant cell responses in disease.


Asunto(s)
Matriz Extracelular , Factor 15 de Diferenciación de Crecimiento , Fibrosis Pulmonar Idiopática , Matriz Extracelular/metabolismo , Fibrosis/genética , Fibrosis/metabolismo , Factor 15 de Diferenciación de Crecimiento/biosíntesis , Factor 15 de Diferenciación de Crecimiento/genética , Humanos , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/metabolismo , Transducción de Señal
5.
Sci Rep ; 11(1): 21584, 2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34732748

RESUMEN

The unfolded protein response (UPR) is a direct consequence of cellular endoplasmic reticulum (ER) stress and a key disease driving mechanism in IPF. The resolution of the UPR is directed by PPP1R15A (GADD34) and leads to the restoration of normal ribosomal activity. While the role of PPP1R15A has been explored in lung epithelial cells, the role of this UPR resolving factor has yet to be explored in lung mesenchymal cells. The objective of the current study was to determine the expression and role of PPP1R15A in IPF fibroblasts and in a bleomycin-induced lung fibrosis model. A survey of IPF lung tissue revealed that PPP1R15A expression was markedly reduced. Targeting PPP1R15A in primary fibroblasts modulated TGF-ß-induced fibroblast to myofibroblast differentiation and exacerbated pulmonary fibrosis in bleomycin-challenged mice. Interestingly, the loss of PPP1R15A appeared to promote lung fibroblast senescence. Taken together, our findings demonstrate the major role of PPP1R15A in the regulation of lung mesenchymal cells, and regulation of PPP1R15A may represent a novel therapeutic strategy in IPF.


Asunto(s)
Senescencia Celular , Fibrosis/metabolismo , Proteína Fosfatasa 1/genética , Respuesta de Proteína Desplegada , Anciano , Animales , Bleomicina , Diferenciación Celular , Proliferación Celular , Estrés del Retículo Endoplásmico , Femenino , Fibroblastos/metabolismo , Genotipo , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Indoles/farmacología , Pulmón/metabolismo , Masculino , Mesodermo/citología , Ratones , Persona de Mediana Edad , Morfolinas/farmacología , Proteína Fosfatasa 1/fisiología , Análisis de Secuencia de ARN , Factor de Crecimiento Transformador beta/metabolismo
6.
Am J Respir Cell Mol Biol ; 65(4): 347-365, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34129811

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a fatal interstitial lung disease with limited therapeutic options. Current evidence suggests that IPF may be initiated by repeated epithelial injuries in the distal lung, which are followed by abnormal wound healing responses that occur because of intrinsic and extrinsic factors. Mechanisms contributing to chronic damage of the alveolar epithelium in IPF include dysregulated cellular processes such as apoptosis, senescence, abnormal activation of the developmental pathways, aging, and genetic mutations. Therefore, targeting the regenerative capacity of the lung epithelium is an attractive approach in the development of novel therapies for IPF. Endogenous lung regeneration is a complex process involving coordinated cross-talk among multiple cell types and reestablishment of a normal extracellular matrix environment. This review will describe the current knowledge of reparative epithelial progenitor cells in the alveolar region of the lung and discuss potential novel therapeutic approaches for IPF, focusing on endogenous alveolar repair.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Enfermedades Pulmonares Intersticiales/metabolismo , Pulmón/metabolismo , Animales , Senescencia Celular/fisiología , Humanos , Células Madre/metabolismo
7.
JCI Insight ; 6(11)2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-33945505

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is characterized by aberrant repair that diminishes lung function via mechanisms that remain poorly understood. CC chemokine receptor (CCR10) and its ligand CCL28 were both elevated in IPF compared with normal donors. CCR10 was highly expressed by various cells from IPF lungs, most notably stage-specific embryonic antigen-4-positive mesenchymal progenitor cells (MPCs). In vitro, CCL28 promoted the proliferation of CCR10+ MPCs while CRISPR/Cas9-mediated targeting of CCR10 resulted in the death of MPCs. Following the intravenous injection of various cells from IPF lungs into immunodeficient (NOD/SCID-γ, NSG) mice, human CCR10+ cells initiated and maintained fibrosis in NSG mice. Eph receptor A3 (EphA3) was among the highest expressed receptor tyrosine kinases detected on IPF CCR10+ cells. Ifabotuzumab-targeted killing of EphA3+ cells significantly reduced the numbers of CCR10+ cells and ameliorated pulmonary fibrosis in humanized NSG mice. Thus, human CCR10+ cells promote pulmonary fibrosis, and EphA3 mAb-directed elimination of these cells inhibits lung fibrosis.


Asunto(s)
Fibrosis Pulmonar Idiopática/metabolismo , Células Madre Mesenquimatosas/metabolismo , Receptor EphA3/metabolismo , Receptores CCR10/metabolismo , Células Epiteliales Alveolares/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Sistemas CRISPR-Cas , Quimiocinas CC/metabolismo , Fibroblastos/metabolismo , Técnicas de Inactivación de Genes , Humanos , Fibrosis Pulmonar Idiopática/patología , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones Endogámicos NOD , Ratones SCID
8.
JCI Insight ; 5(4)2020 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-32102985

RESUMEN

Neutrophils are the most abundant inflammatory cells at the earliest stages of wound healing and play important roles in wound repair and fibrosis. Formyl peptide receptor 1 (FPR-1) is abundantly expressed on neutrophils and has been shown to regulate their function, yet the importance of FPR-1 in fibrosis remains ill defined. FPR-1-deficient (fpr1-/-) mice were protected from bleomycin-induced pulmonary fibrosis but developed renal and hepatic fibrosis normally. Mechanistically, we observed a failure to effectively recruit neutrophils to the lungs of fpr1-/- mice, whereas neutrophil recruitment was unaffected in the liver and kidney. Using an adoptive transfer model we demonstrated that the defect in neutrophil recruitment to the lung was intrinsic to the fpr1-/- neutrophils, as C57BL/6 neutrophils were recruited normally to the damaged lung in fpr1-/- mice. Finally, C57BL/6 mice in which neutrophils had been depleted were protected from pulmonary fibrosis. In conclusion, FPR-1 and FPR-1 ligands are required for effective neutrophil recruitment to the damaged lung. Failure to recruit neutrophils or depletion of neutrophils protects from pulmonary fibrosis.


Asunto(s)
Infiltración Neutrófila/fisiología , Fibrosis Pulmonar/fisiopatología , Receptores de Formil Péptido/fisiología , Animales , Bleomicina/toxicidad , Humanos , Ligandos , Ratones Endogámicos C57BL , Ratones Noqueados , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Receptores de Formil Péptido/genética , Receptores de Formil Péptido/metabolismo
9.
Sci Rep ; 9(1): 6020, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30988425

RESUMEN

Phenotypic changes in lung fibroblasts are believed to contribute to the development of Idiopathic Pulmonary Fibrosis (IPF), a progressive and fatal lung disease. Long intergenic non-coding RNAs (lincRNAs) have been identified as novel regulators of gene expression and protein activity. In non-stimulated cells, we observed reduced proliferation and inflammation but no difference in the fibrotic response of IPF fibroblasts. These functional changes in non-stimulated cells were associated with changes in the expression of the histone marks, H3K4me1, H3K4me3 and H3K27ac indicating a possible involvement of epigenetics. Following activation with TGF-ß1 and IL-1ß, we demonstrated an increased fibrotic but reduced inflammatory response in IPF fibroblasts. There was no significant difference in proliferation following PDGF exposure. The lincRNAs, LINC00960 and LINC01140 were upregulated in IPF fibroblasts. Knockdown studies showed that LINC00960 and LINC01140 were positive regulators of proliferation in both control and IPF fibroblasts but had no effect upon the fibrotic response. Knockdown of LINC01140 but not LINC00960 increased the inflammatory response, which was greater in IPF compared to control fibroblasts. Overall, these studies demonstrate for the first time that lincRNAs are important regulators of proliferation and inflammation in human lung fibroblasts and that these might mediate the reduced inflammatory response observed in IPF-derived fibroblasts.


Asunto(s)
Fibroblastos/patología , Fibrosis Pulmonar Idiopática/genética , Pulmón/patología , ARN Largo no Codificante/genética , Células Cultivadas , Epigénesis Genética , Femenino , Fibroblastos/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/patología , Pulmón/metabolismo , Masculino , Persona de Mediana Edad , Transcriptoma
10.
Sci Rep ; 9(1): 1605, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30733557

RESUMEN

Plasminogen activator inhibitor-1 (PAI-1) is a serine protease inhibitor (serpin) that regulates fibrinolysis, cell adhesion and cell motility via its interactions with plasminogen activators and vitronectin. PAI-1 has been shown to play a role in a number of diverse pathologies including cardiovascular diseases, obesity and cancer and is therefore an attractive therapeutic target. However the multiple patho-physiological roles of PAI-1, and understanding the relative contributions of these in any one disease setting, make the development of therapeutically relevant molecules challenging. Here we describe the identification and characterisation of fully human antibody MEDI-579, which binds with high affinity and specificity to the active form of human PAI-1. MEDI-579 specifically inhibits serine protease interactions with PAI-1 while conserving vitronectin binding. Crystallographic analysis reveals that this specificity is achieved through direct binding of MEDI-579 Fab to the reactive centre loop (RCL) of PAI-1 and at the same exosite used by both tissue and urokinase plasminogen activators (tPA and uPA). We propose that MEDI-579 acts by directly competing with proteases for RCL binding and as such is able to modulate the interaction of PAI-1 with tPA and uPA in a way not previously described for a human PAI-1 inhibitor.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Inhibidor 1 de Activador Plasminogénico/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Neutralizantes/química , Especificidad de Anticuerpos , Humanos , Ratones , Modelos Moleculares , Inhibidor 1 de Activador Plasminogénico/química , Conformación Proteica , Ratas
11.
Am J Respir Crit Care Med ; 197(11): 1443-1456, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29634284

RESUMEN

RATIONALE: Idiopathic pulmonary fibrosis (IPF) is characterized by aberrant lung remodeling, which progressively abolishes lung function in an RTK (receptor tyrosine kinase)-dependent manner. Gas6 (growth arrest-specific 6) ligand, Tyro3 (TYRO3 protein tyrosine kinase 3), and Axl (anexelekto) RTK expression and activity are increased in IPF. OBJECTIVES: To determine if targeting these RTK pathways would inhibit fibroblast activation and the development of pulmonary fibrosis. METHODS: Quantitative genomic, proteomic, and functional analyses were used to determine Gas6/TAM (Tyro3, Axl, and Mertk [MER proto-oncogene, tyrosine kinase]) RTK expression and activation in tissues and fibroblasts from normal and IPF lungs. The profibrotic impact of these RTK pathways were also examined in bleomycin-induced pulmonary fibrosis and in SCID/Bg mice that developed pulmonary fibrosis after the intravenous administration of primary IPF fibroblasts. MEASUREMENTS AND MAIN RESULTS: Gas6, Axl, and Tyro3 were increased in both rapidly and slowly progressive IPF compared with normal lung samples and fibroblasts. Targeting these pathways with either specific antibodies directed at Gas6 or Axl, or with small-molecule TAM inhibitors indicated that the small molecule-mediated targeting approach was more efficacious in both in vitro and in vivo studies. Specifically, the TAM receptor inhibitor R428 (also known as BGB324) significantly inhibited the synthetic, migratory, and proliferative properties of IPF fibroblasts compared with the other Gas6/TAM receptor targeting agents. Finally, loss of Gas6 expression decreased lung fibrotic responses to bleomycin and treatment with R428 inhibited pulmonary fibrosis in humanized SCID/Bg mice. CONCLUSIONS: Gas6/TAM receptor activity contributes to the activation of pulmonary fibroblasts in IPF, suggesting that targeting this RTK pathway might be an effective antifibrotic strategy in this disease.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/efectos de los fármacos , Antibióticos Antineoplásicos/uso terapéutico , Bleomicina/uso terapéutico , Fibroblastos/efectos de los fármacos , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Fibrosis Pulmonar Idiopática/genética , Proteínas de la Membrana/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/genética , Adulto , Anciano , Anciano de 80 o más Años , Proliferación Celular/efectos de los fármacos , Humanos , Fibrosis Pulmonar Idiopática/fisiopatología , Proteínas de la Membrana/genética , Persona de Mediana Edad , Proto-Oncogenes Mas , Transducción de Señal/genética
12.
JCI Insight ; 3(5)2018 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-29515024

RESUMEN

Periplakin is a component of the desmosomes that acts as a cytolinker between intermediate filament scaffolding and the desmosomal plaque. Periplakin is strongly expressed by epithelial cells in the lung and is a target antigen for autoimmunity in idiopathic pulmonary fibrosis. The aim of this study was to determine the role of periplakin during lung injury and remodeling in a mouse model of lung fibrosis induced by bleomycin. We found that periplakin expression was downregulated in the whole lung and in alveolar epithelial cells following bleomycin-induced injury. Deletion of the Ppl gene in mice improved survival and reduced lung fibrosis development after bleomycin-induced injury. Notably, Ppl deletion promoted an antiinflammatory alveolar environment linked to profound changes in type 2 alveolar epithelial cells, including overexpression of antiinflammatory cytokines, decreased expression of profibrotic mediators, and altered cell signaling with a reduced response to TGF-ß1. These results identify periplakin as a previously unidentified regulator of the response to injury in the lung.


Asunto(s)
Células Epiteliales Alveolares/patología , Fibrosis Pulmonar Idiopática/patología , Lesión Pulmonar/patología , Plaquinas/metabolismo , Mucosa Respiratoria/patología , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/inmunología , Animales , Bleomicina/administración & dosificación , Bleomicina/toxicidad , Citocinas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Humanos , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/inmunología , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plaquinas/genética , Mucosa Respiratoria/citología , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/inmunología , Transducción de Señal/inmunología
13.
Front Immunol ; 9: 2906, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30619270

RESUMEN

There is accumulating evidence to indicate that long non-coding RNAs (lncRNAs) are important regulators of the inflammatory response. In this report, we have employed next generation sequencing to identify 14 lncRNAs that are differentially expressed in human lung fibroblasts following the induction of inflammation using interleukin-1ß (IL-1ß). Knockdown of the two most highly expressed lncRNAs, IL7AS, and MIR3142HG, showed that IL7AS negatively regulated IL-6 release whilst MIR3142HG was a positive regulator of IL-8 and CCL2 release. Parallel studies in fibroblasts derived from patients with idiopathic pulmonary fibrosis showed similar increases in IL7AS levels, that also negatively regulate IL-6 release. In contrast, IL-1ß-induced MIR3142HG expression, and its metabolism to miR-146a, was reduced by 4- and 9-fold in IPF fibroblasts, respectively. This correlated with a reduced expression of inflammatory mediators whilst MIR3142HG knockdown showed no effect upon IL-8 and CCL2 release. Pharmacological studies showed that IL-1ß-induced IL7AS and MIR3142HG production and release of IL-6, IL-8, and CCL2 in both control and IPF fibroblasts were mediated via an NF-κB-mediated pathway. In summary, we have cataloged those lncRNAs that are differentially expressed following IL-1ß-activation of human lung fibroblasts, shown that IL7AS and MIR3142HG regulate the inflammatory response and demonstrated that the reduced inflammatory response in IPF fibroblast is correlated with attenuated expression of MIR3142HG/miR-146a.


Asunto(s)
Fibroblastos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Fibrosis Pulmonar Idiopática/genética , Inflamación/genética , Interleucina-1beta/farmacología , ARN Largo no Codificante/genética , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Fibrosis Pulmonar Idiopática/patología , Inflamación/metabolismo , Inflamación/patología , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Masculino , Persona de Mediana Edad
14.
Sci Rep ; 7(1): 15444, 2017 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-29133960

RESUMEN

Lung fibrosis is an unabated wound healing response characterized by the loss and aberrant function of lung epithelial cells. Herein, we report that extracellular Clusterin promoted epithelial cell apoptosis whereas intracellular Clusterin maintained epithelium viability during lung repair. Unlike normal and COPD lungs, IPF lungs were characterized by significantly increased extracellular Clusterin whereas the inverse was evident for intracellular Clusterin. In vitro and in vivo studies demonstrated that extracellular Clusterin promoted epithelial cell apoptosis while intercellular Clusterin modulated the expression of the DNA repair proteins, MSH2, MSH6, OGG1 and BRCA1. The fibrotic response in Clusterin deficient (CLU-/-) mice persisted after bleomycin and it was associated with increased DNA damage, reduced DNA repair responses, and elevated cellular senescence. Remarkably, this pattern mirrored that observed in IPF lung tissues. Together, our results show that cellular localization of Clusterin leads to divergent effects on epithelial cell regeneration and lung repair during fibrosis.


Asunto(s)
Clusterina/metabolismo , Fibrosis Pulmonar Idiopática/patología , Pulmón/patología , Enfermedad Pulmonar Obstructiva Crónica/patología , Anciano , Animales , Apoptosis , Bleomicina/efectos adversos , Estudios de Casos y Controles , Línea Celular , Clusterina/sangre , Clusterina/genética , Citoplasma/metabolismo , Roturas del ADN de Doble Cadena , Reparación de la Incompatibilidad de ADN , Conjuntos de Datos como Asunto , Modelos Animales de Enfermedad , Células Epiteliales/patología , Espacio Extracelular/metabolismo , Femenino , Fibrosis , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Fibrosis Pulmonar Idiopática/sangre , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/genética , Pulmón/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/sangre , ARN Interferente Pequeño/metabolismo , Mucosa Respiratoria/citología , Mucosa Respiratoria/patología
15.
JCI Insight ; 2(16)2017 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-28814671

RESUMEN

The chronic progressive decline in lung function observed in idiopathic pulmonary fibrosis (IPF) appears to result from persistent nonresolving injury to the epithelium, impaired restitution of the epithelial barrier in the lung, and enhanced fibroblast activation. Thus, understanding these key mechanisms and pathways modulating both is essential to greater understanding of IPF pathogenesis. We examined the association of VEGF with the IPF disease state and preclinical models in vivo and in vitro. Tissue and circulating levels of VEGF were significantly reduced in patients with IPF, particularly in those with a rapidly progressive phenotype, compared with healthy controls. Lung-specific overexpression of VEGF significantly protected mice following intratracheal bleomycin challenge, with a decrease in fibrosis and bleomycin-induced cell death observed in the VEGF transgenic mice. In vitro, apoptotic endothelial cell-derived mediators enhanced epithelial cell injury and reduced epithelial wound closure. This process was rescued by VEGF pretreatment of the endothelial cells via a mechanism involving thrombospondin-1 (TSP1). Taken together, these data indicate beneficial roles for VEGF during lung fibrosis via modulating epithelial homeostasis through a previously unrecognized mechanism involving the endothelium.

16.
Respir Res ; 18(1): 82, 2017 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-28468623

RESUMEN

BACKGROUND: Smoking and aberrant epithelial responses are risk factors for lung cancer as well as chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis. In these conditions, disease progression is associated with epithelial damage and fragility, airway remodelling and sub-epithelial fibrosis. The aim of this study was to assess the acute effects of cigarette smoke on epithelial cell phenotype and pro-fibrotic responses in vitro and in vivo. RESULTS: Apoptosis was significantly greater in unstimulated cells from COPD patients compared to control, but proliferation and CXCL8 release were not different. Cigarette smoke dose-dependently induced apoptosis, proliferation and CXCL8 release with normal epithelial cells being more responsive than COPD patient derived cells. Cigarette smoke did not induce epithelial-mesenchymal transition. In vivo, cigarette smoke exposure promoted epithelial apoptosis and proliferation. Moreover, mimicking a virus-induced exacerbation by exposing to mice to poly I:C, exaggerated the inflammatory responses, whereas expression of remodelling genes was similar in both. CONCLUSIONS: Collectively, these data indicate that cigarette smoke promotes epithelial cell activation and hyperplasia, but a secondary stimulus is required for the remodelling phenotype associated with COPD.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Fibrosis Pulmonar/inducido químicamente , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/fisiopatología , Humo/efectos adversos , Productos de Tabaco/envenenamiento , Animales , Apoptosis/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Enfermedad Pulmonar Obstructiva Crónica/inducido químicamente , Enfermedad Pulmonar Obstructiva Crónica/patología , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/fisiopatología , Mucosa Respiratoria/patología
17.
MAbs ; 9(1): 104-113, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27834568

RESUMEN

Excessive transforming growth factor (TGF)-ß is associated with pro-fibrotic responses in lung disease, yet it also plays essential roles in tissue homeostasis and autoimmunity. Therefore, selective inhibition of excessive and aberrant integrin-mediated TGF-ß activation via targeting the α-v family of integrins is being pursued as a therapeutic strategy for chronic lung diseases, to mitigate any potential safety concerns with global TGF-ß inhibition. In this work, we reveal a novel mechanism of inhibiting TGF-ß activation utilized by an αvß8 targeting antibody, 37E1B5. This antibody blocks TGF-ß activation while not inhibiting cell adhesion. We show that an N-linked complex-type Fab glycan in H-CDR2 of 37E1B5 is directly involved in the inhibition of latent TGF-ß activation. Removal of the Fab N-glycosylation site by single amino acid substitution, or removal of N-linked glycans by enzymatic digestion, drastically reduced the antibody's ability to inhibit latency-associated peptide (LAP) and αvß8 association, and TGF-ß activation in an αvß8-mediated TGF-ß signaling reporter assay. Our results indicate a non-competitive, allosteric inhibition of 37E1B5 on αvß8-mediated TGF-ß activation. This unique, H-CDR2 glycan-mediated mechanism may account for the potent but tolerable TGF-b activation inhibition and lack of an effect on cellular adhesion by the antibody.


Asunto(s)
Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Regiones Determinantes de Complementariedad/química , Integrinas/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/farmacocinética , Regiones Determinantes de Complementariedad/inmunología , Glicosilación , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/inmunología , Ratones , Polisacáridos/química , Procesamiento Proteico-Postraduccional
18.
Pharmacol Ther ; 169: 1-12, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27889330

RESUMEN

A common feature of chronic respiratory disease is the progressive decline in lung function. The decline can be indolent, or it can be accelerated by acute exacerbations, whereby the patient experiences a pronounced worsening of disease symptoms. Moreover, acute exacerbations may also be a marker of insufficient disease management. The underlying cause of an acute exacerbation can be due to insults such as pathogens or environmental pollutants, or the cause can be unknown. For each acute exacerbation, the patient may require medical intervention such as rescue medication, or in more severe cases, hospitalization and ventilation and have an increased risk of death. Biologics, such as monoclonal antibodies, are being developed for chronic respiratory diseases including asthma, COPD and IPF. This therapeutic approach is particularly well suited for chronic use based on the route and frequency of delivery and importantly, the potential for disease modification. In recent clinical trials, the frequency of acute exacerbation has often been included as an endpoint, to help determine whether the investigational agent is impacting disease. Therefore the significance of acute exacerbations in driving disease, and their potential as a marker of disease activity and progression, has recently received much attention. There is also now a need to standardize the definition of an acute exacerbation in specific disease settings, particularly as this endpoint is increasingly used in clinical trials to also assess therapeutic efficacy. Moreover, specifically targeting exacerbations may offer a new therapeutic approach for several chronic respiratory diseases.


Asunto(s)
Asma/tratamiento farmacológico , Productos Biológicos/uso terapéutico , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Enfermedad Aguda , Asma/fisiopatología , Productos Biológicos/administración & dosificación , Enfermedad Crónica , Progresión de la Enfermedad , Humanos , Fibrosis Pulmonar Idiopática/fisiopatología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Pruebas de Función Respiratoria
19.
Pharmacol Ther ; 169: 35-46, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27612548

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a chronic fibrosing interstitial pneumonia of unknown cause, characterised by progressive worsening in lung function and dyspnoea with an associated prognosis similar to or worse than many cancers. As a better understanding emerges around the pathogenesis and mechanisms driving disease pathology, a host of novel agents are being tested both pre-clinically and clinically. However even with this deeper understanding and positive pre-clinical supportive data, negative trial outcomes are frequently reported, highlighting the problems faced in treating such a heterogeneous disease with a varied clinical course. Recently, two therapies that slow disease progression, nintedanib and pirfenidone, have been approved for the treatment of IPF, yet the clinical unmet need is still high for IPF patients given their failure to stop disease progression and their potential side-effect profiles. Efforts are being made to not only understand the underlying pathways and genetics that might influence the clinical course of the disease, but also the non-invasive biomarkers that reflect the activity of specific pathways which in turn may highlight progressive treatment plans for individual patients. The cumulative data may be based on the identification of subgroups of patients via biomarker analysis of ongoing clinical trials, or investigation of cohorts of patients over time to understand the relative role of these mediators in their disease progression. Below we review the ongoing quest for novel therapeutic approaches and highlight, where appropriate attempts have been made to identify patients for which a specific pathway or mediator may be driving disease progression.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Medicina de Precisión/métodos , Animales , Antiinflamatorios no Esteroideos/efectos adversos , Biomarcadores/metabolismo , Progresión de la Enfermedad , Humanos , Fibrosis Pulmonar Idiopática/fisiopatología , Indoles/efectos adversos , Indoles/uso terapéutico , Pronóstico , Piridonas/efectos adversos , Piridonas/uso terapéutico
20.
J Immunol ; 195(3): 1182-90, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26109638

RESUMEN

Small airway chronic inflammation is a major pathologic feature of chronic obstructive pulmonary disease (COPD) and is refractory to current treatments. Dendritic cells (DCs) accumulate around small airways in COPD. DCs are critical mediators of Ag surveillance and Ag presentation and amplify adaptive immune responses. How DCs accumulate around airways remains largely unknown. We use 2-photon DC imaging of living murine lung sections to directly visualize the dynamic movement of living DCs around airways in response to either soluble mediators (IL-1ß) or environmental stimuli (cigarette smoke or TLR3 ligands) implicated in COPD pathogenesis. We find that DCs accumulate around murine airways primarily by increasing velocity (chemokinesis) rather than directional migration (chemotaxis) in response to all three stimuli. DC accumulation maximally occurs in a specific zone located 26-50 µm from small airways, which overlaps with zones of maximal DC velocity. Our data suggest that increased accumulation of DCs around airways results from increased numbers of highly chemokinetic DCs entering the lung from the circulation with balanced rates of immigration and emigration. Increases in DC accumulation and chemokinesis are partially dependent on ccr6, a crucial DC chemokine receptor, and fibroblast expression of the integrin αvß8, a critical activator of TGF-ß. αvß8-Mediated TGF-ß activation is known to enhance IL-1ß-dependent fibroblast expression of the only known endogenous ccr6 chemokine ligand, ccl20. Taken together, these data suggest a mechanism by which αvß8, ccl20, and ccr6 interact to lead to DC accumulation around airways in response to COPD-relevant stimuli.


Asunto(s)
Células Dendríticas/inmunología , Integrinas/inmunología , Interleucina-1beta/inmunología , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Factor de Crecimiento Transformador beta/inmunología , Inmunidad Adaptativa/inmunología , Animales , Movimiento Celular/inmunología , Quimiocina CCL20/biosíntesis , Quimiocina CCL20/inmunología , Modelos Animales de Enfermedad , Activación Enzimática/inmunología , Fibroblastos/inmunología , Integrinas/biosíntesis , Interleucina-1beta/biosíntesis , Pulmón/diagnóstico por imagen , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Poli I-C/farmacología , Enfermedad Pulmonar Obstructiva Crónica/patología , Radiografía , Receptores CCR6/genética , Receptores CCR6/inmunología , Humo/efectos adversos , Receptor Toll-Like 3 , Factor de Crecimiento Transformador beta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...