Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Respir Crit Care Med ; 199(12): 1550-1560, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-30557518

RESUMEN

Rationale: Remodeling and fibrosis of the right ventricle (RV) may cause RV dysfunction and poor survival in patients with pulmonary hypertension. Objectives: To investigate the consequences of RV fibrosis modulation and the accompanying cellular changes on RV function. Methods: Expression of fibrotic markers was assessed in the RV of patients with pulmonary hypertension, the murine pulmonary artery banding, and rat monocrotaline and Sugen5416/hypoxia models. Invasive hemodynamic and echocardiographic assessment was performed on galectin-3 knockout or inhibitor-treated mice. Measurements and Main Results: Established fibrosis was characterized by marked expression of galectin-3 and an enhanced number of proliferating RV fibroblasts. Galectin-3 genetic and pharmacologic inhibition or antifibrotic treatment with pirfenidone significantly diminished RV fibrosis progression in the pulmonary artery banding model, without improving RV functional parameters. RV fibrotic regions were populated with mesenchymal cells coexpressing vimentin and PDGFRα (platelet-derived growth factor receptor-α), but generally lacked αSMA (α-smooth muscle actin) positivity. Serum levels of galectin-3 were increased in patients with idiopathic pulmonary arterial hypertension but did not correlate with cardiac function. No changes of galectin-3 expression were observed in the lungs. Conclusions: We identified extrapulmonary galectin-3 as an important mediator that drives RV fibrosis in pulmonary hypertension through the expansion of PDGFRα/vimentin-expressing cardiac fibroblasts. However, interventions effectively targeting fibrosis lack significant beneficial effects on RV function.


Asunto(s)
Fibrosis/complicaciones , Fibrosis/fisiopatología , Galectina 3/inmunología , Hipertrofia Ventricular Derecha/etiología , Hipertrofia Ventricular Derecha/fisiopatología , Disfunción Ventricular Derecha/etiología , Disfunción Ventricular Derecha/fisiopatología , Animales , Austria , Baltimore , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratas , Función Ventricular Derecha/efectos de los fármacos
2.
Eur Respir J ; 54(3)2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31320452

RESUMEN

The interleukin (IL)-1 family of cytokines is strongly associated with systemic sclerosis (SSc) and pulmonary involvement, but the molecular mechanisms are poorly understood. The aim of this study was to assess the role of IL-1α and IL-1ß in pulmonary vascular and interstitial remodelling in a mouse model of SSc.IL-1α and IL-1ß were localised in lungs of SSc patients and in the fos-related antigen-2 (Fra-2) transgenic (TG) mouse model of SSc. Lung function, haemodynamic parameters and pulmonary inflammation were measured in Fra-2 TG mice with or without 8 weeks of treatment with the IL-1 receptor antagonist anakinra (25 mg·kg-1·day-1). Direct effects of IL-1 on pulmonary arterial smooth muscle cells (PASMCs) and parenchymal fibroblasts were investigated in vitroFra-2 TG mice exhibited increased collagen deposition in the lung, restrictive lung function and enhanced muscularisation of the vasculature with concomitant pulmonary hypertension reminiscent of the changes in SSc patients. Immunoreactivity of IL-1α and IL-1ß was increased in Fra-2 TG mice and in patients with SSc. IL-1 stimulation reduced collagen expression in PASMCs and parenchymal fibroblasts via distinct signalling pathways. Blocking IL-1 signalling in Fra-2 TG worsened pulmonary fibrosis and restriction, enhanced T-helper cell type 2 (Th2) inflammation, and increased the number of pro-fibrotic, alternatively activated macrophages.Our data suggest that blocking IL-1 signalling as currently investigated in several clinical studies might aggravate pulmonary fibrosis in specific patient subsets due to Th2 skewing of immune responses and formation of alternatively activated pro-fibrogenic macrophages.


Asunto(s)
Inflamación/metabolismo , Receptores Tipo I de Interleucina-1/antagonistas & inhibidores , Esclerodermia Sistémica/metabolismo , Células Th2/metabolismo , Animales , Líquido del Lavado Bronquioalveolar , Modelos Animales de Enfermedad , Femenino , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Transgénicos , Miocitos del Músculo Liso/metabolismo , Fibrosis Pulmonar/patología , Pruebas de Función Respiratoria , Transducción de Señal
3.
Eur Respir J ; 53(6)2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31023847

RESUMEN

Our systematic analysis of anion channels and transporters in idiopathic pulmonary arterial hypertension (IPAH) showed marked upregulation of the Cl- channel TMEM16A gene. We hypothesised that TMEM16A overexpression might represent a novel vicious circle in the molecular pathways causing pulmonary arterial hypertension (PAH).We investigated healthy donor lungs (n=40) and recipient lungs with IPAH (n=38) for the expression of anion channel and transporter genes in small pulmonary arteries and pulmonary artery smooth muscle cells (PASMCs).In IPAH, TMEM16A was strongly upregulated and patch-clamp recordings confirmed an increased Cl- current in PASMCs (n=9-10). These cells were depolarised and could be repolarised by TMEM16A inhibitors or knock-down experiments (n=6-10). Inhibition/knock-down of TMEM16A reduced the proliferation of IPAH-PASMCs (n=6). Conversely, overexpression of TMEM16A in healthy donor PASMCs produced an IPAH-like phenotype. Chronic application of benzbromarone in two independent animal models significantly decreased right ventricular pressure and reversed remodelling of established pulmonary hypertension.Our findings suggest that increased TMEM16A expression and activity comprise an important pathologic mechanism underlying the vasoconstriction and remodelling of pulmonary arteries in PAH. Inhibition of TMEM16A represents a novel therapeutic approach to reverse remodelling in PAH.


Asunto(s)
Anoctamina-1/metabolismo , Hipertensión Pulmonar Primaria Familiar/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas de Neoplasias/metabolismo , Remodelación Vascular , Vasoconstricción , Adulto , Anciano , Animales , Anoctamina-1/genética , Estudios de Casos y Controles , Proliferación Celular , Modelos Animales de Enfermedad , Hipertensión Pulmonar Primaria Familiar/patología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/patología , Proteínas de Neoplasias/genética , Técnicas de Placa-Clamp , Arteria Pulmonar/fisiopatología , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba
4.
J Pathol ; 244(4): 485-498, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29359814

RESUMEN

Pulmonary vascular remodeling is the main pathological hallmark of pulmonary hypertension disease. We undertook a comprehensive and multilevel approach to investigate the origin of smooth muscle actin-expressing cells in remodeled vessels. Transgenic mice that allow for specific, inducible, and permanent labeling of endothelial (Cdh5-tdTomato), smooth muscle (Acta2-, Myh11-tdTomato), pericyte (Cspg4-tdTomato), and fibroblast (Pdgfra-tdTomato) lineages were used to delineate the cellular origins of pulmonary vascular remodeling. Mapping the fate of major lung resident cell types revealed smooth muscle cells (SMCs) as the predominant source of cells that populate remodeled pulmonary vessels in chronic hypoxia and allergen-induced murine models. Combining in vivo cell type-specific, time-controlled labeling of proliferating cells with a pulmonary artery phenotypic explant assay, we identified proliferation of SMCs as an underlying remodeling pathomechanism. Multicolor immunofluorescence analysis showed a preserved pattern of cell type marker localization in murine and human pulmonary arteries, in both donors and idiopathic pulmonary arterial hypertension (IPAH) patients. Whilst neural glial antigen 2 (chondroitin sulfate proteoglycan 4) labeled mostly vascular supportive cells with partial overlap with SMC markers, PDGFRα-expressing cells were observed in the perivascular compartment. The luminal vessel side was lined by a single cell layer expressing endothelial markers followed by an adjacent and distinct layer defined by SMC marker expression and pronounced thickening in remodeled vessels. Quantitative flow cytometric analysis of single cell digests of diverse pulmonary artery layers showed the preserved separation into two discrete cell populations expressing either endothelial cell (EC) or SMC markers in human remodeled vessels. Additionally, we found no evidence of overlap between EC and SMC ultrastructural characteristics using electron microscopy in either donor or IPAH arteries. Lineage-specific marker expression profiles are retained during pulmonary vascular remodeling without any indication of cell type conversion. The expansion of resident SMCs is the major underlying and evolutionarily conserved paradigm of pulmonary vascular disease pathogenesis. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Linaje de la Célula , Genes Reporteros , Hipoxia/patología , Pulmón/irrigación sanguínea , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Hipersensibilidad Respiratoria/patología , Remodelación Vascular , Actinas/genética , Actinas/metabolismo , Animales , Antígenos/genética , Antígenos/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Enfermedad Crónica , Modelos Animales de Enfermedad , Hipertensión Pulmonar Primaria Familiar/metabolismo , Hipertensión Pulmonar Primaria Familiar/patología , Hipertensión Pulmonar Primaria Familiar/fisiopatología , Técnica del Anticuerpo Fluorescente , Humanos , Hipoxia/genética , Hipoxia/metabolismo , Hipoxia/fisiopatología , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones Transgénicos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/metabolismo , Fenotipo , Proteoglicanos/genética , Proteoglicanos/metabolismo , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología , Arteria Pulmonar/fisiopatología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Hipersensibilidad Respiratoria/genética , Hipersensibilidad Respiratoria/metabolismo , Hipersensibilidad Respiratoria/fisiopatología , Proteína Fluorescente Roja
5.
Am J Respir Crit Care Med ; 197(2): 244-260, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29095649

RESUMEN

RATIONALE: Vascular remodeling in pulmonary arterial hypertension (PAH) results from smooth muscle cell hypertrophy and proliferation of vascular cells. Loss of BMPR-II (bone morphogenetic protein receptor 2) signaling and increased signaling via TGF-ß (transforming growth factor ß) and its downstream mediators SMAD (small body size [a C. elegans protein] mothers against decapentaplegic [a Drosophila protein family])-2/3 has been proposed to drive lung vascular remodeling; yet, proteomic analyses indicate a loss of SMAD3 in PAH. OBJECTIVES: We proposed that SMAD3 may be dysregulated in PAH and that loss of SMAD3 may present a pathophysiological master switch by disinhibiting its interaction partner, MRTF (myocardin-related transcription factor), which drives muscle protein expression. METHODS: SMAD3 levels were measured in lungs from PAH patients, rats treated either with Sugen/hypoxia or monocrotaline (MCT), and in mice carrying a BMPR2 mutation. In vitro, effects of SMAD3 or BMPR2 silencing or SMAD3 overexpression on cell proliferation or smooth muscle hypertrophy were assessed. In vivo, the therapeutic and prophylactic potential of CCG1423, an inhibitor of MRTF, was investigated in Sugen/hypoxia rats. MEASUREMENTS AND MAIN RESULTS: SMAD3 was downregulated in lungs of patients with PAH and in pulmonary arteries of three independent PAH animal models. TGF-ß treatment replicated the loss of SMAD3 in human pulmonary artery smooth muscle cells (huPASMCs) and human pulmonary artery endothelial cells. SMAD3 silencing increased proliferation and migration in huPASMCs and human pulmonary artery endothelial cells. Coimmunoprecipitation revealed reduced interaction of MRTF with SMAD3 in TGF-ß-treated huPASMCs and pulmonary arteries of PAH animal models. In huPASMCs, loss of SMAD3 or BMPR-II increased smooth muscle actin expression, which was attenuated by MRTF inhibition. Conversely, SMAD3 overexpression prevented TGF-ß-induced activation of an MRTF reporter and reduced actin stress fibers in BMPR2-silenced huPASMCs. MRTF inhibition attenuated PAH and lung vascular remodeling in Sugen/hypoxia rats. CONCLUSIONS: Loss of SMAD3 presents a novel pathomechanism in PAH that promotes vascular cell proliferation and-via MRTF disinhibition-hypertrophy of huPASMCs, thereby reconciling the parallel induction of a synthetic and contractile huPASMC phenotype.


Asunto(s)
Hipertensión Pulmonar/genética , Proteína smad3/genética , Factores de Transcripción/genética , Factor de Crecimiento Transformador beta/farmacología , Remodelación Vascular/genética , Animales , Movimiento Celular/genética , Proliferación Celular/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo , Humanos , Hipertensión Pulmonar/fisiopatología , Masculino , Células Musculares/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Sensibilidad y Especificidad , Transfección
6.
Eur Respir J ; 51(1)2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29371380

RESUMEN

Increasing evidence points towards an inflammatory component underlying pulmonary hypertension. However, the conclusive characterisation of multiple inflammatory cell populations in the lung is challenging due to the complexity of marker specificity and tissue inaccessibility. We used an unbiased computational flow cytometry approach to delineate the inflammatory landscape of idiopathic pulmonary arterial hypertension (IPAH) and healthy donor lungs.Donor and IPAH samples were discriminated clearly using principal component analysis to reduce the multidimensional data obtained from single-cell flow cytometry analysis. In IPAH lungs, the predominant CD45+ cell type switched from neutrophils to CD3+ T-cells, with increases in CD4+, CD8+ and γδT-cell subsets. Additionally, diversely activated classical myeloid-derived dendritic cells (CD14-HLA-DR+CD11c+CD1a+/-) and nonclassical plasmacytoid dendritic cells (pDCs; CD14-CD11c-CD123+HLA-DR+), together with mast cells and basophils, were more abundant in IPAH samples. We describe, for the first time, the presence and regulation of two cell types in IPAH, γδT-cells and pDCs, which link innate and adaptive immunity.With our high-throughput flow cytometry with multidimensional dataset analysis, we have revealed the interactive interplay between multiple inflammatory cells is a crucial part of their integrative network. The identification of γδT-cells and pDCs in this disease potentially provides a missing link between IPAH, autoimmunity and inflammation.


Asunto(s)
Hipertensión Pulmonar/patología , Inflamación/patología , Pulmón/patología , Inmunidad Adaptativa , Adulto , Biología Computacional , Simulación por Computador , Células Dendríticas/citología , Femenino , Citometría de Flujo , Humanos , Hipertensión Pulmonar/metabolismo , Inmunofenotipificación , Inflamación/metabolismo , Linfocitos Intraepiteliales/citología , Antígenos Comunes de Leucocito/metabolismo , Pulmón/metabolismo , Masculino , Persona de Mediana Edad , Análisis de Componente Principal , Arteria Pulmonar/patología
7.
Am J Physiol Lung Cell Mol Physiol ; 313(5): L741-L751, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28705908

RESUMEN

The tryptophan metabolite kynurenine is significantly increased in pulmonary arterial hypertension (PAH) patients, and it is a potent vasodilator of systemic arteries. Our aim was to investigate the role of kynurenine in the pulmonary circulation. Serum tryptophan, kynurenine, and kynurenic acid levels were measured in 20 idiopathic PAH (IPAH) patients, 20 healthy controls, and 20 patients with chronic lung disease or metabolic syndrome without PH. Laser-dissected pulmonary arteries from IPAH and control lungs were tested for the expression of indoleamine-2, 3-dioxygenase (IDO), the rate-limiting enzyme for the conversion from tryptophan to kynurenine. Acute effects of kynurenine were tested in pulmonary vascular preparations, two different models of chronic pulmonary hypertension (PH), and in human pulmonary arterial smooth muscle cells (hPASMCs). In IPAH vs. control serum, kynurenine was significantly elevated (3.6 ± 0.2 vs. 2.6 ± 0.1 µM, P < 0.0001), and strongly associated with PH (area under the curve = 0.86), but kynurenine levels were not elevated in lung disease and metabolic syndrome. Among all investigated tryptophan metabolites, kynurenine displayed the strongest correlation with mean pulmonary arterial pressure (mPAP) (ρ: 0.770, P < 0.0001). Tryptophan was significantly decreased in IPAH lungs; however, IDO expression was not changed. In hPASMCs, kynurenine increased both cAMP and cGMP; in intrapulmonary arteries, it relaxed the preconstriction via NO/cGMP and cAMP pathways, and in two models of established PH, it acutely decreased the mPAP. Our data suggest that kynurenine elevation might be specifically associated with mPAP; kynurenine acts on hPASMCs in synergy with NO and exerts acute pulmonary vasodilatation in chronic PH models. Kynurenine might provide both a new biomarker and a new therapeutic option for PH.


Asunto(s)
Hipertensión Pulmonar/metabolismo , Quinurenina/metabolismo , Pulmón/metabolismo , Arteria Pulmonar/metabolismo , Adolescente , Adulto , Anciano , Animales , Niño , Enfermedad Crónica , Femenino , Humanos , Hipertensión Pulmonar/patología , Pulmón/fisiopatología , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Miocitos del Músculo Liso/metabolismo , Arteria Pulmonar/patología , Vasodilatadores/farmacología , Adulto Joven
8.
Eur Respir J ; 50(1)2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28729471

RESUMEN

Chronic obstructive pulmonary disease (COPD) is a heterogeneous disease in which the amount of emphysema and airway disease may be very different between individuals, even in end-stage disease. Emphysema formation may be linked to the involvement of the small pulmonary vessels. The NAPDH oxidase (Nox) family is emerging as a key disease-related factor in vascular diseases, but currently its role in hypoxia-induced pulmonary remodelling in COPD remains unclear.Here we investigate the role of p22phox, a regulatory subunit of Nox, in COPD lungs, hypoxic pulmonary vasoconstriction (HPV), hypoxia-induced pulmonary vascular remodelling and pulmonary hypertension.In COPD, compared to control lungs, p22phox expression was significantly reduced. The expression was correlated positively with mean pulmonary arterial pressure and oxygenation index and negatively with the diffusing capacity of the lung for carbon monoxide (p<0.02). This suggests a role of p22phox in ventilation/perfusion ratio matching, vascular remodelling and loss of perfused lung area. In p22phox-/- mice, HPV was significantly impaired. In the chronic hypoxic setting, lack of p22phox was associated with improved right ventricular function and decreased pulmonary vascular remodelling.p22phox-dependent Nox plays an important role in the COPD phenotype, by its action on phase II HPV and chronic vascular remodelling.


Asunto(s)
Grupo Citocromo b/metabolismo , Hipertensión Pulmonar/metabolismo , Pulmón/fisiopatología , NADPH Oxidasas/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfisema Pulmonar/metabolismo , Adulto , Animales , Monóxido de Carbono/análisis , Estudios de Casos y Controles , Grupo Citocromo b/genética , Femenino , Humanos , Hipoxia/fisiopatología , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , NADPH Oxidasas/genética , Enfermedad Pulmonar Obstructiva Crónica/complicaciones , Remodelación Vascular , Vasoconstricción , Función Ventricular Derecha , Adulto Joven
9.
Am J Physiol Lung Cell Mol Physiol ; 311(1): L59-73, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27106290

RESUMEN

Adaptation of the right ventricle (RV) to increased afterload is crucial for survival in pulmonary hypertension (PH), but it is challenging to assess RV function and identify associated molecular mechanisms. The aim of the current study was to analyze the relationship between invasive and noninvasive parameters of RV morphology and function and associated molecular changes. The response of mice to normobaric hypoxia was assessed by hechocardiography, invasive hemodynamics, and histological and molecular analyses. Plasma levels of possibly novel markers of RV remodeling were measured by ELISA in patients with idiopathic pulmonary arterial hypertension (IPAH) and matched healthy controls. Chronic hypoxia-induced PH was accompanied by significantly decreased tricuspid annular plane systolic excursion (TAPSE) and unchanged RV contractility index and tau. RV hypertrophy was present without an increase in fibrosis. There was no change in α- and ß-major histocompatibility class or natriuretic peptides expression. Comparative microarray analysis identified two soluble factors, fibroblast growth factor-5 (FGF5) and interleukin-22 receptor alpha-2 (IL22RA2), as being possibly associated with RV remodeling. We observed significantly higher plasma levels of IL22RA2, but not FGF5, in patients with IPAH. Hypoxic pulmonary hypertension in a stage of RV remodeling with preserved systolic function is associated with decreased pulmonary vascular compliance, mild diastolic RV dysfunction, and significant decrease in TAPSE. Subtle gene expression changes in the RV vs. the left ventricle upon chronic hypoxia suggest that the majority of changes are due to hypoxia and not due to changes in afterload. Increased IL22RA2 levels might represent a novel RV adaptive mechanism.


Asunto(s)
Ventrículos Cardíacos/patología , Hipertensión Pulmonar/fisiopatología , Válvula Tricúspide/patología , Animales , Hipoxia de la Célula , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/fisiopatología , Hipertensión Pulmonar/diagnóstico por imagen , Hipertensión Pulmonar/metabolismo , Hipertrofia Ventricular Derecha/diagnóstico por imagen , Hipertrofia Ventricular Derecha/metabolismo , Hipertrofia Ventricular Derecha/patología , Masculino , Ratones , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miocardio/metabolismo , Receptores de Interleucina/sangre , Transcriptoma , Función Ventricular Derecha , Remodelación Ventricular
10.
Eur Respir J ; 48(2): 459-69, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27103380

RESUMEN

Circulating mononuclear cells may play an important role for the vascular remodelling in pulmonary arterial hypertension (PAH), but studies addressing multiple progenitor populations are rare and inconsistent.We used a comprehensive fluorescence-activated cell sorting analysis of circulating mononuclear cells in 20 PAH patients and 20 age- and sex-matched controls, and additionally analysed CD133(+) cells in the lung tissue of five PAH transplant recipients and five healthy controls (donor lungs).PAH patients were characterised by increased numbers of circulating CD133(+) cells and lymphopenia as compared with control. In PAH, CD133(+) subpopulations positive for CD117 or CD45 were significantly increased, whereas CD133(+)CD309(+), CD133(+)CXCR2(+) and CD133(+)CD31(+) cells were decreased. In CD133(+) cells, SOX2, Nanog, Ki67 and CXCR4 were not detected, but Oct3/4 mRNA was present in both PAH and controls. In the lung tissue, CD133(+) cells included three main populations: type 2 pneumocytes, monocytes and undifferentiated cells without significant differences between PAH and controls.In conclusion, circulating CD133(+) progenitor cells are elevated in PAH and consist of phenotypically different subpopulations that may be up- or downregulated. This may explain the inconsistent results in the literature. CD133(+) type 2 pneumocytes in the lung tissue are not associated with circulating CD133(+) mononuclear cells.


Asunto(s)
Antígeno AC133/metabolismo , Hipertensión Pulmonar/metabolismo , Leucocitos Mononucleares/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Separación Celular , Femenino , Citometría de Flujo , Humanos , Antígenos Comunes de Leucocito/metabolismo , Pulmón/patología , Masculino , Persona de Mediana Edad , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Arteria Pulmonar/patología , Receptores de Interleucina-8B/metabolismo , Factores de Transcripción SOXB1/metabolismo , Células Madre , Remodelación Vascular , Adulto Joven
11.
Eur Respir J ; 48(4): 1127-1136, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27540020

RESUMEN

Cardioprotective benefits of ω-3 fatty acids such as docosahexaenoic acid (DHA) are well established, but the regulatory effect of DHA on vascular tone and pressure in pulmonary hypertension is largely unknown.As DHA is a potent regulator of K+ channels, we hypothesised that DHA modulates the membrane potential of pulmonary artery smooth muscle cells (PASMCs) through K+ channels and thus exerts its effects on pulmonary vascular tone and pressure.We show that DHA caused dose-dependent activation of the calcium-activated K+ (KCa) current in primary human PASMCs and endothelium-dependent relaxation of pulmonary arteries. This vasodilation was significantly diminished in KCa-/- (Kcnma1-/-) mice. In vivo, acute DHA returned the right ventricular systolic pressure in the chronic hypoxia-induced pulmonary hypertension animal model to the level of normoxic animals. Interestingly, in idiopathic pulmonary arterial hypertension the KCa channels and their subunits were upregulated. DHA activated KCa channels in these human PASMCs and hyperpolarised the membrane potential of the idiopathic pulmonary arterial hypertension PASMCs to that of the PASMCs from healthy donors.Our findings indicate that DHA activates PASMC KCa channels leading to vasorelaxation in pulmonary hypertension. This effect might provide a molecular explanation for the previously undescribed role of DHA as an acute vasodilator in pulmonary hypertension.


Asunto(s)
Ácidos Docosahexaenoicos/farmacología , Hipertensión Pulmonar/tratamiento farmacológico , Miocitos del Músculo Liso/efectos de los fármacos , Arteria Pulmonar/efectos de los fármacos , Adulto , Animales , Hipertensión Pulmonar Primaria Familiar/fisiopatología , Femenino , Humanos , Hipertensión Pulmonar/fisiopatología , Hipoxia/fisiopatología , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Músculo Liso Vascular/citología , Perfusión , Arteria Pulmonar/fisiopatología , Vasodilatación
12.
Clin Transplant ; 30(8): 912-7, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27220108

RESUMEN

BACKGROUND: Incidentally discovered lung cancers in lung transplant (LuTX) recipients are rare but may affect outcome. We aim to report our single center experience with incidence, management, and survival of patients with previously unverified primary lung cancer discovered at the time of LuTX. METHODS: A total of 1262 patients undergoing LuTX between 1989 and 2012 were retrospectively analyzed in our prospective database. RESULTS: Patients identified were six men and five women with a mean age of 54.4±9.9 years. The indication for LuTX was COPD (n=9), pulmonary fibrosis (n=1), and cystic fibrosis (n=1). Histological diagnosis of the explanted lung revealed adenocarcinoma in six, squamous cell carcinoma in three, and lepidic predominant adenocarcinoma in two cases, respectively. Staging revealed stage IA (pT1a/b pN0) in eight and IB (pT2a pN0) in two cases and one patient in stage IIA (pT1b pN1). Subsequent cancer staging after LuTX revealed no metastasis. Immunosuppression was adjusted and no adjuvant chemo- or radiotherapy was administered. The 5-year survival rate was 90.5% with no detection of recurrence. CONCLUSION: In patients with incidentally detected early stage lung cancer at the time of LuTX, rates of recurrence and survival based on this sample appear to be acceptable.


Asunto(s)
Hospitales de Alto Volumen , Hallazgos Incidentales , Neoplasias Pulmonares/cirugía , Trasplante de Pulmón/métodos , Estadificación de Neoplasias , Austria/epidemiología , Biopsia , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/mortalidad , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Tasa de Supervivencia/tendencias
13.
J Pathol ; 237(2): 203-14, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26011651

RESUMEN

Malignant pleural mesothelioma (MPM) is a devastating malignancy characterized by invasive growth and rapid recurrence. The identification and inhibition of molecular components leading to this migratory and invasive phenotype are thus essential. Accordingly, a genome-wide expression array analysis was performed on MPM cell lines and a set of 139 genes was identified as differentially expressed in cells with high versus low migratory activity. Reduced expression of the novel tumour suppressor integrin α7 (ITGA7) was found in highly motile cells. A significant negative correlation was observed between ITGA7 transcript levels and average displacement of cells. Forced overexpression of ITGA7 in MPM cells with low endogenous ITGA7 expression inhibited cell motility, providing direct evidence for the regulatory role of ITGA7 in MPM cell migration. MPM cells showed decreased ITGA7 expressions at both transcription and protein levels when compared to non-malignant mesothelial cells. The majority of MPM cell cultures displayed hypermethylation of the ITGA7 promoter when compared to mesothelial cultures. A statistically significant negative correlation between ITGA7 methylation and ITGA7 expression was also observed in MPM cells. While normal human pleura samples unambiguously expressed ITGA7, a varying level of expression was found in a panel of 200 human MPM samples. In multivariate analysis, ITGA7 expression was found to be an independent prognostic factor. Although there was no correlation between histological subtypes and ITGA7 expression, importantly, patients with high tumour cell ITGA7 expression had an increased median overall survival compared to the low- or no-expression groups (463 versus 278 days). In conclusion, our data suggest that ITGA7 is an epigenetically regulated tumour suppressor gene and a prognostic factor in human MPM.


Asunto(s)
Antígenos CD/metabolismo , Movimiento Celular , Epigénesis Genética , Cadenas alfa de Integrinas/metabolismo , Neoplasias Pulmonares/metabolismo , Mesotelioma/metabolismo , Neoplasias Pleurales/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Antígenos CD/genética , Línea Celular Tumoral , Metilación de ADN , Regulación hacia Abajo , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Cadenas alfa de Integrinas/genética , Estimación de Kaplan-Meier , Laminina/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Mesotelioma/genética , Mesotelioma/mortalidad , Mesotelioma/patología , Mesotelioma Maligno , Análisis Multivariante , Invasividad Neoplásica , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Pleurales/genética , Neoplasias Pleurales/mortalidad , Neoplasias Pleurales/patología , Pronóstico , Regiones Promotoras Genéticas , Modelos de Riesgos Proporcionales , ARN Mensajero/metabolismo , Factores de Riesgo , Transducción de Señal , Factores de Tiempo , Transfección , Proteínas Supresoras de Tumor/genética
14.
Eur Arch Otorhinolaryngol ; 273(5): 1283-92, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-25801951

RESUMEN

Minor salivary gland carcinoma is a rare and heterogeneous type of cancer. Molecular prognostic and predictive markers are sparse. The aim of this study was to identify new prognostic and predictive markers in minor salivary gland carcinoma. 50 tissue samples of carcinomas of the minor salivary glands (adenoid cystic carcinoma n = 23, mucoepidermoid carcinoma n = 12, adenocarcinoma n = 10, carcinoma ex pleomorphic adenoma n = 2, salivary duct carcinoma n = 1, clear cell carcinoma n = 1, basal cell carcinoma n = 1) were immunohistochemically stained for ß-catenin, cyclin D1 and PIN1. Expression patterns were analyzed and correlated to clinical outcome of 37 patients with complete clinical data. High expression of membranous ß-catenin was linked to significantly better overall survival in patients with adenoid cystic carcinoma (log rank test, χ (2) = 13.3, p = .00397, Bonferroni corrected p = .024). PIN1 and cyclin D1 did not show any significant correlation to patients' clinical outcome. Expression of ß-catenin in adenoid cystic carcinoma of the minor salivary glands significantly correlates with better overall survival. Hence, evaluation of ß-catenin might serve as a clinical prognostic marker.


Asunto(s)
Carcinoma Adenoide Quístico , Ciclina D1/metabolismo , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Neoplasias de las Glándulas Salivales , Glándulas Salivales Menores/patología , beta Catenina/metabolismo , Austria , Biomarcadores de Tumor/metabolismo , Carcinoma Adenoide Quístico/metabolismo , Carcinoma Adenoide Quístico/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Evaluación de Resultado en la Atención de Salud , Valor Predictivo de las Pruebas , Pronóstico , Estudios Retrospectivos , Neoplasias de las Glándulas Salivales/metabolismo , Neoplasias de las Glándulas Salivales/patología , Estadística como Asunto , Análisis de Supervivencia
15.
J Cell Mol Med ; 19(5): 1151-61, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25726846

RESUMEN

Extracellular high-mobility group box-1 (HMGB1) acts as a signalling molecule during inflammation, cell differentiation and angiogenesis. Increased abundance of HMGB1 is associated with several pathological disorders such as cancer, asthma and chronic obstructive pulmonary disease (COPD). In this study, we investigated the relevance of HMGB1 in the pathological remodelling present in patients with idiopathic pulmonary arterial hypertension (IPAH) and pulmonary hypertension (PH) associated with COPD. Remodelled vessels present in COPD with PH and IPAH lung samples were often surrounded by HMGB1-positive cells. Increased HMGB1 serum levels were detected in both patient populations compared to control samples. The effects of physiological HMGB1 concentrations were then examined on cellular responses in vitro. HMGB1 enhanced proliferation of pulmonary arterial smooth muscle cells (PASMC) and primary human arterial endothelial cells (PAEC). HMGB1 stimulated p38, extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) phosphorylation. Furthermore, activation of the downstream AP-1 complex proteins c-Fos and c-Jun was observed. Silencing of c-Jun ablated the HMGB1-induced proliferation in PASMC. Thus, an inflammatory component such as HMGB1 can contribute to PASMC and PAEC proliferation and therefore potentially to vascular remodelling and PH pathogenesis.


Asunto(s)
Proteína HMGB1/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Remodelación Vascular/fisiología , Anciano , Western Blotting , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Activación Enzimática/efectos de los fármacos , Hipertensión Pulmonar Primaria Familiar/genética , Hipertensión Pulmonar Primaria Familiar/metabolismo , Hipertensión Pulmonar Primaria Familiar/patología , Femenino , Proteína HMGB1/sangre , Proteína HMGB1/farmacología , Humanos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Inmunohistoquímica , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Masculino , Persona de Mediana Edad , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Arteria Pulmonar/citología , Interferencia de ARN , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Remodelación Vascular/efectos de los fármacos , Remodelación Vascular/genética
16.
Am J Physiol Lung Cell Mol Physiol ; 308(10): L1002-13, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25840998

RESUMEN

Alterations in extracellular matrix (ECM) have been implicated in the pathophysiology of pulmonary hypertension. Here, we have undertaken a compartment-specific study to elucidate the expression profile of collagens and their processing enzymes in donor and idiopathic pulmonary arterial hypertension (IPAH) pulmonary arteries. Predominant intimal, but also medial and perivascular, remodeling and reduced lumen diameter were detected in IPAH pulmonary arteries. Two-photon microscopy demonstrated accumulation of collagen fibers. Quantification of collagen in pulmonary arteries revealed collagen accumulation mainly in the intima of IPAH pulmonary arteries compared with donors. Laser capture-microdissected pulmonary artery profiles (intima+media and perivascular tissue) were analyzed by real-time PCR for ECM gene expression. In the intima+media of IPAH vessels, collagens (COL4A5, COL14A1, and COL18A1), matrix metalloproteinase (MMP) 19, and a disintegrin and metalloprotease (ADAM) 33 were higher expressed, whereas MMP10, ADAM17, TIMP1, and TIMP3 were less abundant. Localization of COLXVIII, its cleavage product endostatin, and MMP10, ADAM33, and TIMP1 was confirmed in pulmonary arteries by immunohistochemistry. ELISA for collagen XVIII/endostatin demonstrated significantly elevated plasma levels in IPAH patients compared with donors, whereas circulating MMP10, ADAM33, and TIMP1 levels were similar between the two groups. Endostatin levels were correlated with pulmonary arterial wedge pressure, and established prognostic markers of IPAH, right atrial pressure, cardiac index, 6-min walking distance, NH2-terminal pro-brain natriuretic peptide, and uric acid. Expression of unstudied collagens, MMPs, ADAMs, and TIMPs were found to be significantly altered in IPAH intima+media. Elevated levels of circulating collagen XVIII/endostatin are associated with markers of a poor prognosis.


Asunto(s)
Colágeno/biosíntesis , Regulación de la Expresión Génica , Hipertensión Pulmonar/metabolismo , Arteria Pulmonar/metabolismo , Túnica Íntima/metabolismo , Biomarcadores/sangre , Endostatinas/sangre , Femenino , Humanos , Hipertensión Pulmonar/diagnóstico , Hipertensión Pulmonar/patología , Masculino , Metaloproteasas , Persona de Mediana Edad , Pronóstico , Arteria Pulmonar/patología , Túnica Íntima/patología
17.
Respir Res ; 16: 127, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26483185

RESUMEN

BACKGROUND: Chronic obstructive lung disease (COPD) is a common cause of death in industrialized countries often induced by exposure to tobacco smoke. A substantial number of patients with COPD also suffer from pulmonary hypertension that may be caused by hypoxia or other hypoxia-independent stimuli - inducing pulmonary vascular remodeling. The Ca(2+) binding protein, S100A4 is known to play a role in non-COPD-driven vascular remodeling of intrapulmonary arteries. Therefore, we have investigated the potential involvement of S100A4 in COPD induced vascular remodeling. METHODS: Lung tissue was obtained from explanted lungs of five COPD patients and five non-transplanted donor lungs. Additionally, mice lungs of a tobacco-smoke-induced lung emphysema model (exposure for 3 and 8 month) and controls were investigated. Real-time RT-PCR analysis of S100A4 and RAGE mRNA was performed from laser-microdissected intrapulmonary arteries. S100A4 immunohistochemistry was semi-quantitatively evaluated. Mobility shift assay and siRNA knock-down were used to prove hypoxia responsive elements (HRE) and HIF binding within the S100A4 promoter. RESULTS: Laser-microdissection in combination with real-time PCR analysis revealed higher expression of S100A4 mRNA in intrapulmonary arteries of COPD patients compared to donors. These findings were mirrored by semi-quantitative analysis of S100A4 immunostaining. Analogous to human lungs, in mice with tobacco-smoke-induced emphysema an up-regulation of S100A4 mRNA and protein was observed in intrapulmonary arteries. Putative HREs could be identified in the promoter region of the human S100A4 gene and their functionality was confirmed by mobility shift assay. Knock-down of HIF1/2 by siRNA attenuated hypoxia-dependent increase in S100A4 mRNA levels in human primary pulmonary artery smooth muscle cells. Interestingly, RAGE mRNA expression was enhanced in pulmonary arteries of tobacco-smoke exposed mice but not in pulmonary arteries of COPD patients. CONCLUSIONS: As enhanced S100A4 expression was observed in remodeled intrapulmonary arteries of COPD patients, targeting S100A4 could serve as potential therapeutic option for prevention of vascular remodeling in COPD patients.


Asunto(s)
Arteria Pulmonar/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfisema Pulmonar/metabolismo , Proteínas S100/metabolismo , Fumar/efectos adversos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Sitios de Unión , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Factor 1 Inducible por Hipoxia/genética , Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Arteria Pulmonar/patología , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfermedad Pulmonar Obstructiva Crónica/patología , Enfisema Pulmonar/etiología , Enfisema Pulmonar/genética , Enfisema Pulmonar/patología , Interferencia de ARN , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Elementos de Respuesta , Proteína de Unión al Calcio S100A4 , Proteínas S100/genética , Transfección , Regulación hacia Arriba , Remodelación Vascular
18.
J Pathol ; 233(1): 7-17, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24258247

RESUMEN

Vascular remodelling is a hallmark of pulmonary hypertension (PH) and is characterized by enhanced proliferation of pulmonary artery smooth muscle cells (PASMCs). Accumulating evidence indicates a crucial role of transcription factors in the vascular remodelling processes. Here, we characterize the involvement of meprin ß, a novel activator protein-1 (AP-1) effector molecule, in PH. Fra-2 transgenic (TG) mice exhibited increased right ventricular systolic pressure (RVSP), accompanied by vascular remodelling and activation of the pro-proliferative and pro-fibrotic AKT pathway. Microarray studies revealed the collagen-processing metalloprotease meprin ß as the most up-regulated gene in Fra-2 TG mice. Its expression, increased at all investigated time points, preceded the decreased expression of MMPs and increased TGFß, followed by collagen deposition. Correspondingly, remodelled pulmonary arteries from explanted idiopathic pulmonary arterial hypertension (IPAH) patients' lungs exhibited pronounced expression of meprin ß. Fra-2 and meprin ß expression in human PASMCs was regulated by PDGF-BB and TGFß in a complementary fashion. Importantly, PDGF-BB-dependent proliferation was attenuated by silencing AP-1 expression or by meprin ß inhibition. This study delineates a novel molecular mechanism underlying PASMCs proliferation and extracellular matrix (ECM) deposition by identifying meprin ß as an important mediator in regulating vascular remodelling processes. Thus, meprin ß may represent a new molecule that can be targeted in pulmonary hypertension.


Asunto(s)
Proliferación Celular , Hipertensión Pulmonar/metabolismo , Metaloendopeptidasas/metabolismo , Arteria Pulmonar/metabolismo , Animales , Becaplermina , Células Cultivadas , Colágeno/metabolismo , Modelos Animales de Enfermedad , Hipertensión Pulmonar Primaria Familiar , Fibrosis , Antígeno 2 Relacionado con Fos/genética , Antígeno 2 Relacionado con Fos/metabolismo , Humanos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/fisiopatología , Metaloproteinasas de la Matriz/metabolismo , Metaloendopeptidasas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-sis/metabolismo , Arteria Pulmonar/patología , Arteria Pulmonar/fisiopatología , Interferencia de ARN , Transducción de Señal , Factores de Tiempo , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Transfección , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba , Función Ventricular Derecha , Presión Ventricular
19.
Am J Respir Crit Care Med ; 190(1): 98-111, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24918967

RESUMEN

RATIONALE: The development of pulmonary hypertension (PH) in patients with idiopathic pulmonary fibrosis (IPF) or chronic obstructive pulmonary disease (COPD) is associated with increased morbidity. OBJECTIVES: To elucidate whether vascular remodeling in a well-characterized PH-COPD and PH-IPF patient cohort results from similar or divergent molecular changes. METHODS: Vascular remodeling of donor, PH-COPD, and PH-IPF pulmonary arteries was assessed. Laser capture microdissected pulmonary artery profiles in combination with whole genome microarrays were performed. MEASUREMENTS AND MAIN RESULTS: Pulmonary arteries from patients with COPD and IPF with PH exhibited remodeling of vascular layers and reduction of lumen area. Pathway analyses comparing normalized gene expression profiles obtained from patients with PH-IPF or PH-COPD revealed the retinol and extracellular matrix (ECM) receptor interaction to be the most perturbed processes. Within the ECM-receptor pathway, differential regulation of 5 out of the top 10 results (collagen, type III, α-1; tenascin C; collagen, type VI, α-3; thrombospondin 2; and von Willebrand factor) were verified by real-time polymerase chain reaction and immunohistochemical staining. CONCLUSIONS: Despite clinical and histologic vascular remodeling in all patients with PH-COPD and PH-IPF, differential gene expression pattern was present in pulmonary artery profiles. Several genes involved in retinol metabolism and ECM receptor interaction enable discrimination of vascular remodeling in PH-IPF or PH-COPD. This suggests that pulmonary arterial remodeling in PH-COPD and PH-IPF is caused by different molecular mechanisms and may require specific therapeutic options.


Asunto(s)
Hipertensión Pulmonar/genética , Fibrosis Pulmonar Idiopática/genética , Arteria Pulmonar/patología , Enfermedad Pulmonar Obstructiva Crónica/genética , Adulto , Anciano , Remodelación de las Vías Aéreas (Respiratorias)/genética , Remodelación de las Vías Aéreas (Respiratorias)/fisiología , Austria , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/patología , Fibrosis Pulmonar Idiopática/complicaciones , Fibrosis Pulmonar Idiopática/patología , Captura por Microdisección con Láser , Masculino , Persona de Mediana Edad , Pronóstico , Arteria Pulmonar/fisiopatología , Enfermedad Pulmonar Obstructiva Crónica/complicaciones , Enfermedad Pulmonar Obstructiva Crónica/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Transcriptoma
20.
Am J Respir Crit Care Med ; 190(7): 763-72, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25188816

RESUMEN

RATIONALE: Malignant pleural mesothelioma is an aggressive malignancy characterized by frequent resistance to chemo- and radiotherapy, poor outcome, and limited therapeutic options. Fibroblast growth factors (FGFs) and their receptors are potential targets for cancer therapy, but their significance in mesothelioma has remained largely undefined. OBJECTIVES: To investigate the antimesothelioma potential of FGF receptor 1 (FGFR1) inhibition. METHODS: Expression of FGFs and their receptors was analyzed in mesothelioma cell lines and tissue specimens. Several cell models were used to investigate FGFR1 inhibition in vitro and in combination with cisplatin and irradiation. Mouse intraperitoneal xenotransplant models were used for in vivo validation. MEASUREMENTS AND MAIN RESULTS: FGFR1, FGF2, and FGF18 were overexpressed in mesothelioma. Stimulation with FGF2 led to increased cell proliferation, migration, and transition to a more sarcomatoid phenotype in subsets of mesothelioma cell lines. In contrast, inhibition of FGFR1 by a specific kinase inhibitor or a dominant-negative FGFR1 construct led to significantly decreased proliferation, clonogenicity, migration, spheroid formation, and G1 cell cycle arrest in several mesothelioma cell lines, accompanied by apoptosis induction and decreased mitogen-activated protein kinase pathway activity. Reduced tumor growth, proliferation, mitogenic signaling, and apoptosis induction were observed in vivo. Inhibition of FGFR1 synergistically enhanced the cytotoxic effects of ionizing radiation and cisplatin. CONCLUSIONS: Our data suggest that the malignant phenotype of mesothelioma cells depends on intact FGF signals, which should be considered as therapeutic targets with a promising chemo- and radiosensitizing potential.


Asunto(s)
Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/radioterapia , Mesotelioma/tratamiento farmacológico , Mesotelioma/radioterapia , Inhibidores de Proteínas Quinasas/farmacología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Cisplatino/farmacología , Terapia Combinada/métodos , Modelos Animales de Enfermedad , Humanos , Neoplasias Pulmonares/genética , Mesotelioma/genética , Mesotelioma Maligno , Ratones , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/efectos de los fármacos , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA