Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
FASEB J ; 35(3): e21422, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33638895

RESUMEN

Idiopathic pulmonary fibrosis is a lethal lung fibrotic disease, associated with aging with a mean survival of 2-5 years and no curative treatment. The GSE4 peptide is able to rescue cells from senescence, DNA and oxidative damage, inflammation, and induces telomerase activity. Here, we investigated the protective effect of GSE4 expression in vitro in rat alveolar epithelial cells (AECs), and in vivo in a bleomycin model of lung fibrosis. Bleomycin-injured rat AECs, expressing GSE4 or treated with GSE4-PLGA/PEI nanoparticles showed an increase of telomerase activity, decreased DNA damage, and decreased expression of IL6 and cleaved-caspase 3. In addition, these cells showed an inhibition in expression of fibrotic markers induced by TGF-ß such as collagen-I and III among others. Furthermore, treatment with GSE4-PLGA/PEI nanoparticles in a rat model of bleomycin-induced fibrosis, increased telomerase activity and decreased DNA damage in proSP-C cells. Both in preventive and therapeutic protocols GSE4-PLGA/PEI nanoparticles prevented and attenuated lung damage monitored by SPECT-CT and inhibited collagen deposition. Lungs of rats treated with bleomycin and GSE4-PLGA/PEI nanoparticles showed reduced expression of α-SMA and pro-inflammatory cytokines, increased number of pro-SPC-multicellular structures and increased DNA synthesis in proSP-C cells, indicating therapeutic efficacy of GSE4-nanoparticles in experimental lung fibrosis and a possible curative treatment for lung fibrotic patients.


Asunto(s)
Apoptosis/efectos de los fármacos , Bleomicina/farmacología , Daño del ADN/efectos de los fármacos , Pulmón/efectos de los fármacos , Nanopartículas/uso terapéutico , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Colágeno/efectos de los fármacos , Colágeno/metabolismo , Humanos , Pulmón/metabolismo , Estrés Oxidativo/efectos de los fármacos , Péptidos/farmacología
2.
J Allergy Clin Immunol ; 135(2): 470-6, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25159466

RESUMEN

BACKGROUND: A number of patients with chronic rhinosinusitis with nasal polyps (CRSwNP) are resistant to oral corticosteroids. Mucin 1 (MUC1) shows anti-inflammatory properties, and its cytoplasmic tail (CT) interacts with transcription factors, facilitating their nuclear translocation. Because glucocorticoid receptor (GR) nuclear translocation is key to the anti-inflammatory effect of corticosteroids, we hypothesized that MUC1 is involved in the effectiveness of corticosteroids. OBJECTIVE: To analyze the role of MUC1 in corticosteroid effectiveness in different cohorts of patients with CRSwNP and elucidate the possible mechanisms involved. METHODS: Seventy-three patients with CRSwNP took oral corticosteroids for 15 days. Corticosteroid resistance was evaluated by nasal endoscopy. The expression of MUC1 and MUC1 CT was evaluated by real-time PCR, Western blotting, and immunohistochemistry. Beas-2B knockdown with RNA interference for MUC1 (siRNA-MUC1) was used to analyze the role of MUC1 in the anti-inflammatory effects of dexamethasone. RESULTS: Nineteen patients had nasal polyps that were resistant to oral corticosteroids (NP-CR). MUC1 expression was downregulated in these patients. Primary epithelial cells from patients with NP-CR were insensitive to the anti-inflammatory effects of dexamethasone. In siRNA-MUC1 Beas-2B, dexamethasone showed weaker anti-inflammatory effects, a reduced inhibition of phospho-extracellular-signal-regulated kinases 1/2, a less severe mitogen-activated protein kinase phosphatase 1 increase, and a reduced GR nuclear translocation. Immunoprecipitation experiments revealed that MUC1-CT and GRα form protein complexes and translocate to the nucleus in response to dexamethasone. MUC1-CT-GRα complex was downregulated in NP-CR tissue. CONCLUSION: MUC1-CT participates in the corticosteroid response that mediates GRα nuclear translocation. The low expression of MUC1 in patients with CRSwNP may participate in corticosteroid resistance.


Asunto(s)
Corticoesteroides/uso terapéutico , Resistencia a Medicamentos/genética , Regulación de la Expresión Génica , Mucina-1/genética , Rinitis/tratamiento farmacológico , Rinitis/genética , Sinusitis/tratamiento farmacológico , Sinusitis/genética , Antiinflamatorios/uso terapéutico , Enfermedad Crónica , Regulación hacia Abajo , Humanos , Mucosa Nasal/efectos de los fármacos , Mucosa Nasal/metabolismo , Pólipos Nasales/complicaciones , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Rinitis/complicaciones , Rinitis/metabolismo , Transducción de Señal , Sinusitis/complicaciones , Sinusitis/metabolismo , Receptores Toll-Like/genética
3.
COPD ; 12(3): 320-31, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25207459

RESUMEN

BACKGROUND: Cigarette smoking contributes to epithelial-mesenchymal transition (EMT) in COPD small bronchi as part of the lung remodeling process. We recently observed that roflumilast N-oxide (RNO), the active metabolite of the PDE4 inhibitor roflumilast, prevents cigarette smoke-induced EMT in differentiated human bronchial epithelial cells. Further, statins were shown to protect renal and alveolar epithelial cells from EMT. OBJECTIVES: To analyze how RNO and simvastatin (SIM) interact on CSE-induced EMT in well-differentiated human bronchial epithelial cells (WD-HBEC) from small bronchi in vitro. METHODS: WD-HBEC were stimulated with CSE (2.5%). The mesenchymal markers vimentin, collagen type I and α-SMA, the epithelial markers E-cadherin and ZO-1, as well as ß-catenin were quantified by real time quantitative PCR or Western blotting. Intracellular reactive oxygen species (ROS) were measured using the H2DCF-DA probe. GTP-Rac1 and pAkt were evaluated by Western blotting. RESULTS: The combination of RNO at 2 nM and SIM at 100 nM was (over) additive to reverse CSE-induced EMT. CSE-induced EMT was partially mediated by the generation of ROS and the activation of the PI3K/Akt/ß-catenin pathway. Both RNO at 2 nM and SIM at 100 nM partially abrogated this pathway, and its combination almost abolished ROS/ PI3K/Akt/ß-catenin signaling and therefore EMT. CONCLUSIONS: The PDE4 inhibitor roflumilast N-oxide acts (over)additively with simvastatin to prevent CSE-induced EMT in WD-HBEC in vitro.


Asunto(s)
Aminopiridinas/farmacología , Benzamidas/farmacología , Bronquios/citología , Células Epiteliales/patología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inhibidores de Fosfodiesterasa 4/farmacología , Simvastatina/farmacología , Células Cultivadas , Ciclopropanos/farmacología , Humanos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Fumar/efectos adversos , beta Catenina/metabolismo
4.
Pulm Pharmacol Ther ; 28(2): 138-48, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24525294

RESUMEN

BACKGROUND: Epithelial to mesenchymal transition (EMT) is under discussion as a potential mechanism of small airway remodelling in COPD. In bronchial epithelium of COPD and smokers markers of EMT were described. In vitro, EMT may be reproduced by exposing well-differentiated human bronchial epithelial cells (WD-HBEC) to cigarette smoke extract (CSE). EMT may be mitigated by an increase in cellular cAMP. OBJECTIVE: This study explored the effects of roflumilast N-oxide, a PDE4 inhibitor on CSE-induced EMT in WD-HBEC and in primary bronchial epithelial cells from smokers and COPD in vitro. METHODS: WD-HBEC from normal donors were stimulated with CSE (2.5%) for 72 h in presence of roflumilast N-oxide (2 nM or 1 µM) or vehicle. mRNA and protein of EMT markers αSMA, vimentin, collagen-1, E-cadherin, ZO-1, KRT5 as well as NOX4 were quantified by real-time quantitative PCR or protein array, respectively. Phosphorylated and total ERK1/2 and Smad3 were assessed by protein array. cAMP and TGFß1 were measured by ELISA. Reactive oxygen species (ROS) were determined by DCF fluorescence, after 30 min CSE (2.5%). Apoptosis was measured with Annexin V/PI labelling. In some experiments, EMT markers were determined in monolayers of bronchial epithelial cells from smokers, COPD versus controls. RESULTS: Roflumilast N-oxide protected from CSE-induced EMT in WD-HBEC. The PDE4 inhibitor reversed both the increase in mesenchymal and the loss in epithelial EMT markers. Roflumilast N-oxide restored the loss in cellular cAMP following CSE, reduced ROS, NOX4 expression, the increase in TGFß1 release, phospho ERK1/2 and Smad3. The PDE4 inhibitor partly protected from the increment in apoptosis with CSE. Finally the PDE4 inhibitor decreased mesenchymal yet increased epithelial phenotype markers in HBEC of COPD and smokers. CONCLUSIONS: Roflumilast N-oxide may mitigate epithelial-mesenchymal transition in bronchial epithelial cells in vitro.


Asunto(s)
Aminopiridinas/farmacología , Benzamidas/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Inhibidores de Fosfodiesterasa 4/farmacología , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Anciano , Apoptosis/efectos de los fármacos , Bronquios/citología , Bronquios/efectos de los fármacos , AMP Cíclico/metabolismo , Ciclopropanos/farmacología , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Humanos , Técnicas In Vitro , Masculino , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/metabolismo , Humo/efectos adversos , Fumar/efectos adversos , Factor de Crecimiento Transformador beta1/metabolismo
5.
Thorax ; 68(5): 410-20, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23299965

RESUMEN

BACKGROUND: Cigarette smoking contributes to lung remodelling in chronic obstructive pulmonary disease (COPD). As part of remodelling, peribronchiolar fibrosis is observed in the small airways of patients with COPD and contributes to airway obstruction. Epithelial to mesenchymal transition (EMT) appears to be involved in the formation of peribronchiolar fibrosis. This study examines the EMT process in human bronchial epithelial cells (HBECs) from non-smokers, smokers and patients with COPD as well as the in vitro effect of cigarette smoke extract (CSE) on EMT. METHODS: HBECs from non-smokers (n=5), smokers (n=12) and patients with COPD (n=15) were collected to measure the mesenchymal markers α-smooth muscle actin, vimentin and collagen type I and the epithelial markers E-cadherin, ZO-1 and cytokeratin 5 and 18 by real time-PCR and protein array. In vitro differentiated bronchial epithelial cells were stimulated with CSE. RESULTS: Mesenchymal markers were upregulated in HBECs of smokers and patients with COPD compared with non-smokers. In contrast, epithelial cell markers were downregulated. In vitro differentiated HBECs underwent EMT after 72 h of CSE exposure through the activation of intracellular reactive oxygen species, the release and autocrine action of transforming growth factor ß1, the phosphorylation of ERK1/2 and Smad3 and by the downregulation of cyclic monophosphate. CONCLUSIONS: The EMT process is present in bronchial epithelial cells of the small bronchi of smokers and patients with COPD and is activated by cigarette smoke in vitro.


Asunto(s)
Bronquios/metabolismo , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Humo/efectos adversos , Fumar/efectos adversos , Anciano , Western Blotting , Bronquios/patología , Diferenciación Celular , Células Cultivadas , AMP Cíclico/biosíntesis , AMP Cíclico/genética , Regulación hacia Abajo/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/efectos de los fármacos , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfermedad Pulmonar Obstructiva Crónica/patología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Factor de Crecimiento Transformador beta1/biosíntesis , Factor de Crecimiento Transformador beta1/genética , Vimentina/biosíntesis , Vimentina/genética
6.
Eur Respir J ; 41(6): 1264-74, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23018909

RESUMEN

Cigarette smoking contributes to lung remodelling in chronic obstructive pulmonary disease (COPD). As part of this remodelling, peribronchiolar fibrosis is observed in the small airways of COPD patients and contributes to airway obstruction. Fibroblast-to-myofibroblast transition is a key step in peribronchiolar fibrosis formation. This in vitro study examined the effect of cigarette smoke on bronchial fibroblast-to-myofibroblast transition, and whether aclidinium bromide inhibits this process. Human bronchial fibroblasts were incubated with aclidinium bromide (10(-9)-10(-7) M) and exposed to cigarette smoke extract. Collagen type I and α-smooth muscle actin (α-SMA) expression were measured by real-time PCR and Western blotting, as myofibroblast markers. Intracellular reactive oxygen species, cyclic AMP (cAMP), extracellular signal-regulated kinase (ERK)1/2 and choline acetyltransferase were measured as intracellular signalling mediators. Cigarette smoke-induced collagen type I and α-SMA was mediated by the production of reactive oxygen species, the depletion of intracellular cAMP and the increase of ERK1/2 phosphorylation and choline acetyltransferase. These effects could be reversed by treatment with the anticholinergic aclidinium bromide, by silencing the mRNA of muscarinic receptors M1, M2 or M3, or by the depletion of extracellular acetylcholine by treatment with acetylcholinesterase. A non-neuronal cholinergic system is implicated in cigarette smoke-induced bronchial fibroblast-to-myofibroblast transition, which is inhibited by aclidinium bromide.


Asunto(s)
Fibroblastos/citología , Regulación de la Expresión Génica , Miofibroblastos/citología , Fumar/efectos adversos , Tropanos/farmacología , Actinas/metabolismo , Bronquios/efectos de los fármacos , Células Cultivadas , Antagonistas Colinérgicos/farmacología , Colágeno Tipo I/metabolismo , AMP Cíclico/metabolismo , Fibroblastos/efectos de los fármacos , Fibrosis , Fluoresceínas/farmacología , Humanos , Inflamación , Pulmón/citología , Pulmón/efectos de los fármacos , Microscopía Fluorescente , Miofibroblastos/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Humo , Factores de Tiempo
7.
Thorax ; 67(3): 229-37, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21957094

RESUMEN

BACKGROUND: Fibroblast to myofibroblast transition is believed to contribute to airway remodelling in lung diseases such as asthma and chronic obstructive pulmonary disease. This study examines the role of aclidinium, a new long-acting muscarinic antagonist, on human fibroblast to myofibroblast transition. METHODS: Human bronchial fibroblasts were stimulated with carbachol (10(-8) to 10(-5) M) or transforming growth factor-ß1 (TGF-ß1; 2 ng/ml) in the presence or absence of aclidinium (10(-9) to 10(-7) M) or different drug modulators for 48 h. Characterisation of myofibroblasts was performed by analysis of collagen type I and α-smooth muscle actin (α-SMA) mRNA and protein expression as well as α-SMA microfilament immunofluorescence. ERK1/2 phosphorylation, RhoA-GTP and muscarinic receptors (M) 1, 2 and 3 protein expression were determined by western blot analysis and adenosine 3'-5' cyclic monophosphate levels were determined by ELISA. Proliferation and migration of fibroblasts were also assessed. RESULTS: Collagen type I and α-SMA mRNA and protein expression, as well as percentage α-SMA microfilament-positive cells, were upregulated in a similar way by carbachol and TGF-ß1, and aclidinium reversed these effects. Carbachol-induced myofibroblast transition was mediated by an increase in ERK1/2 phosphorylation, RhoA-GTP activation and cyclic monophosphate downregulation as well as by the autocrine TGF-ß1 release, which were effectively reduced by aclidinium. TGF-ß1 activated the non-neuronal cholinergic system. Suppression of M1, M2 or M3 partially prevented carbachol- and TGF-ß1-induced myofibroblast transition. Aclidinium dose-dependently reduced fibroblast proliferation and migration. CONCLUSION: Aclidinium inhibits human lung fibroblast to myofibrobast transition.


Asunto(s)
Bronquios/citología , Fibroblastos/efectos de los fármacos , Antagonistas Muscarínicos/farmacología , Miofibroblastos/efectos de los fármacos , Tropanos/farmacología , Actinas/biosíntesis , Actinas/genética , Carbacol/antagonistas & inhibidores , Carbacol/farmacología , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Agonistas Colinérgicos/farmacología , Colágeno Tipo I/biosíntesis , Colágeno Tipo I/genética , Relación Dosis-Respuesta a Droga , Fibroblastos/metabolismo , Humanos , ARN Mensajero/genética , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/farmacología , Regulación hacia Arriba/efectos de los fármacos
8.
Thorax ; 67(2): 147-56, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22106015

RESUMEN

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is characterised by the aberrant epithelial to mesenchymal transition (EMT) and myofibroblast accumulation. Sphingosine-1-phosphate (S1P) and sphingosine kinase 1 (SPHK1) have been implicated in lung myofibroblast transition, but their role in EMT and their expression in patients with IPF is unknown. METHODS AND RESULTS: S1P levels were measured in serum (n=27) and bronchoalveolar lavage (BAL; n=15) from patients with IPF and controls (n=30 for serum and n=15 for BAL studies). SPHK1 expression was measured in lung tissue from patients with IPF (n=12) and controls (n=15). Alveolar type II transformation into mesenchymal cells was studied in response to S1P (10(-9)-10(-5) M). The median (IQR) of S1P serum levels was increased in patients with IPF (1.4 (0.4) µM) versus controls (1 (0.26) µM; p<0.0001). BAL S1P levels were increased in patients with IPF (1.12 (0.53) µM) versus controls (0.2 (0.5); p<0.0001) and correlated with diffusion capacity of the lung for carbon monoxide, forced expiratory volume in 1 s and forced vital capacity (Spearman's r=-0.87, -0.72 and -0.68, respectively) in patients with IPF. SPHK1 was upregulated in lung tissue from patients with IPF and correlated with α-smooth muscle actin, vimentin and collagen type I (Spearman's r=0.82, 0.85 and 0.72, respectively). S1P induced EMT in alveolar type II cells by interacting with S1P(2) and S1P(3), as well as by the activation of p-Smad3, RhoA-GTP, oxidative stress and transforming growth factor-ß1 (TGF-ß1) release. Furthermore, TGF-ß1-induced EMT was partially conducted by the S1P/SPHK1 activation, suggesting crosstalk between TGF-ß1 and the S1P/SPHK1 axis. CONCLUSIONS: S1P is elevated in patients with IPF, correlates with the lung function and mediates EMT.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Fibrosis Pulmonar Idiopática/patología , Lisofosfolípidos/fisiología , Esfingosina/análogos & derivados , Adulto , Anciano , Anciano de 80 o más Años , Líquido del Lavado Bronquioalveolar/citología , Estudios de Casos y Controles , Células Cultivadas , Relación Dosis-Respuesta a Droga , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/metabolismo , Lisofosfolípidos/metabolismo , Masculino , Persona de Mediana Edad , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Alveolos Pulmonares/patología , Alveolos Pulmonares/fisiopatología , Especies Reactivas de Oxígeno/metabolismo , Proteína smad3/fisiología , Esfingosina/metabolismo , Esfingosina/fisiología , Factor de Crecimiento Transformador beta1/farmacología , Factor de Crecimiento Transformador beta1/fisiología , Proteína de Unión al GTP rhoA/fisiología
9.
Respiration ; 83(2): 147-58, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22094549

RESUMEN

BACKGROUND: Neutrophils and oxidative stress have been implicated in the pathogenesis of COPD. Severe, early-onset COPD is characterized by a rapid decline in the lung function at an early age; however, nothing is known about neutrophil activation in COPD patients. OBJECTIVES: The aim of this study was to evaluate peripheral blood neutrophil activation in severe, early-onset COPD patients versus healthy non-smokers and the effect of N-acetyl-L-cysteine (NAC) on neutrophil activation in vitro. METHODS: Neutrophils were isolated from 15 severe, early-onset COPD patients and 15 age-matched healthy subjects and stimulated with N-formyl-Met-Leu-Phe (fMLP) in the presence or absence of NAC (10 µM to 10 mM). Neutrophil chemotaxis, elastase release, reactive oxygen species (ROS), intracellular thiols and apoptosis were measured by Boyden chamber, spectrofluorometry, CMFDA and H2DCF-DA dyes and by annexin V-FITC, respectively. RESULTS: Chemotaxis of peripheral blood neutrophils from COPD patients in response to fMLP was 30% more increased than that observed in healthy subjects. Elastase release in response to fMLP was 2-fold higher in neutrophils from COPD patients versus healthy subjects. Intracellular thiol levels were 30% lower in COPD and ROS was approximately 30% higher in COPD versus healthy neutrophils. Spontaneous apoptosis showed no differences in both groups of patients and fMLP-induced apoptosis was higher in COPD. Pre-treatment with the antioxidant NAC effectively attenuated neutrophil chemotaxis, elastase release and ROS as well as effectively increased thiol levels in COPD. CONCLUSIONS: Neutrophils in severe, early-onset COPD patients are highly activated and this is alleviated by NAC in vitro.


Asunto(s)
Acetilcisteína/farmacología , Depuradores de Radicales Libres/farmacología , N-Formilmetionina Leucil-Fenilalanina/farmacología , Activación Neutrófila/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Apoptosis/efectos de los fármacos , Estudios de Casos y Controles , Quimiotaxis/efectos de los fármacos , Femenino , Humanos , Interleucina-8/metabolismo , Elastasa de Leucocito/efectos de los fármacos , Masculino , Persona de Mediana Edad , Especies Reactivas de Oxígeno/metabolismo , Compuestos de Sulfhidrilo/metabolismo
11.
Curr Eye Res ; 41(7): 890-6, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26554729

RESUMEN

PURPOSE: Tacrolimus ocular preparations are commonly employed in autoimmune or inflammatory ocular disorders. However, currently there are not yet approved ocular formulations. Tacrolimus ocular side effects have been reported in clinical use, so the evaluation of different pharmaceutical preparations is mandatory. In this study, the local corneal tolerance and safety profile of three common tacrolimus 0.03% pharmaceutical preparations were evaluated. MATERIAL AND METHODS: Corneal irritation and permeability of tacrolimus preparations were evaluated with the bovine corneal opacity and permeability (BCOP) test. Complementary corneal hematoxylin/eosin and immunohistochemistry staining for tight junctions and adherent junctions E-cadherin, VE-cadherin and zonula occludens-1 were examined and scored to evaluate and to confirm corneal disruption and irritation scores obtained with the BCOP method. RESULTS: Commercial brand ointment (Protopic®), topical compounded eye ointment (pharmacy elaboration) and tacrolimus suspension eye drops (elaborated from parenteral prograf®) were tested as potential ocular preparations to be used in clinics. Tacrolimus preparations hereby studied do not alter the opacity and permeability of the bovine cornea by more than three units, measured by the In Vitro Irritancy Score, neither affected the immunohistochemical parameters, composite score or transepithelial electrical resistance. CONCLUSIONS: Tacrolimus preparations studied can be safely applied as a topical ocular treatment.


Asunto(s)
Córnea/metabolismo , Opacidad de la Córnea/tratamiento farmacológico , Tacrolimus/administración & dosificación , Animales , Bovinos , Córnea/efectos de los fármacos , Opacidad de la Córnea/inducido químicamente , Opacidad de la Córnea/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inmunohistoquímica , Inmunosupresores/administración & dosificación , Irritantes/toxicidad , Soluciones Oftálmicas/administración & dosificación , Soluciones Oftálmicas/farmacocinética , Permeabilidad/efectos de los fármacos , Tacrolimus/farmacocinética
12.
Eur J Pharmacol ; 715(1-3): 172-80, 2013 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-23747655

RESUMEN

Eosinophils play a prominent role in the process of allergic inflammation. Non-receptor associated Lyn tyrosine kinases generate key initial signals in eosinophils. Bafetinib, a specific Abl/Lyn tyrosine kinase inhibitor has shown a potent antiproliferative activity in leukemic cells, but its effects on eosinophils have not been reported. Therefore, we studied the effects of bafetinib on functional and mechanistic responses of isolated human eosinophils. Bafetinib was more potent than non-specific tyrosin kinase comparators genistein and tyrphostin inhibiting superoxide anion triggered by N-formyl-Met-Leu-Phe (fMLF; 100 nM) (-log IC50=7.25 ± 0.04 M; 6.1 ± 0.04 M; and 6.55 ± 0.03 M, respectively). Bafetinib, genistein and tyrphostin did not modify the [Ca(2+)]i responses to fMLF. Bafetinib inhibited the release of EPO induced by fMLF with higher potency than genistein and tyrphostin (-log IC50=7.24 ± 0.09 M; 5.36 ± 0.28 M; and 5.37 ± 0.19 M, respectively), and nearly suppressed LTC4, ECP and chemotaxis. Bafetinib, genistein and tyrphostin did not change constitutive apoptosis. However bafetinib inhibited the ability of granulocyte-monocyte colony-stimulating factor to prevent apoptosis. The activation of Lyn tyrosine kinase, p-ERK1/2 and p-38 induced by fMLF was suppressed by bafetinib and attenuated by genistein and tyrphostin. In conclusion, bafetinib inhibits oxidative burst and generation of inflammatory mediators, and reverses the eosinophil survival. Therefore, future anti-allergic therapies based on bafetinib, could help to suppress excessive inflammatory response of eosinophils at inflammatory sites.


Asunto(s)
Eosinófilos/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Movimiento Celular/efectos de los fármacos , Proteína Catiónica del Eosinófilo/biosíntesis , Eosinófilos/citología , Eosinófilos/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Interleucina-5/farmacología , Leucotrieno C4/biosíntesis , N-Formilmetionina Leucil-Fenilalanina/farmacología , Peroxidasa/metabolismo , Superóxidos/metabolismo
13.
Biochem Pharmacol ; 80(2): 236-46, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20381461

RESUMEN

The airway epithelium routinely undergoes damage that requires repair to restore epithelial barrier integrity. Cell migration followed by proliferation are necessary steps to achieve epithelial repair. Calcium-sensing receptor (CaSR) is implicated in cell migration and proliferation processes. Thus we hypothesized that CaSR mediates lung epithelial wound repair. We detected CaSR expression in human lung and in well-differentiated human bronchial epithelial cells (HBEC). To test the CaSR functionality, HBEC loaded with fura-2 were stimulated with extracellular Ca(2+) ([Ca(2+)](out)) which resulted in a concentration-dependent intracellular Ca(2+) ([Ca(2+)](i)) increase (potency approximately 5.6mM [Ca(2+)](out)). Furthermore, increasing [Ca(2+)](out) induced phosphorylation of the extracellular signal-regulated kinase (ERK1/2) which was blocked by siRNA-CaSR and the specific inhibitor of CaSR, NPS2390. Epithelial repair after mechanical injury of differentiated HBEC was a process dependent of [Ca(2+)](out) since it accelerated wound repair and HBEC proliferation being highest at 5mM [Ca(2+)](out). Furthermore, U73122 (an inhibitor of phospholipase C (PLC)) and PD 98059 (an inhibitor of ERK1/2) as well as siRNA-CaSR and NPS2390 partially inhibited wound repair and HBEC proliferation. On the other hand, mechanical injury produced an [Ca(2+)](i) wave propagation that was partially inhibited by siRNA-CaSR, NPS2390 and the extracellular Ca(2+) chelator EGTA, which suggest a link of CaSR between cell-cell communication and wound repair in differentiated HBEC. Our data, for the first time, shows that CaSR plays an important role in airway epithelial repair, which may help to develop novel regenerative therapeutics allowing the rapid repair of lung damaged epithelium.


Asunto(s)
Bronquios/citología , Señalización del Calcio , Calcio/metabolismo , Células Epiteliales/citología , Receptores Sensibles al Calcio/metabolismo , Cicatrización de Heridas/fisiología , Bronquios/efectos de los fármacos , Bronquios/fisiología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ácido Egtácico/farmacología , Inhibidores Enzimáticos/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/fisiología , Estrenos/farmacología , Fura-2/farmacología , Humanos , Pirrolidinonas/farmacología , Receptores Sensibles al Calcio/antagonistas & inhibidores , Receptores Sensibles al Calcio/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos
14.
Eur J Pharm Sci ; 39(5): 363-72, 2010 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-20093183

RESUMEN

Cigarette smoke mediated oxidative stress and endothelial dysfunction are important processes in the pathogenesis of several lung disorders. In this study we evaluated the effect of PDE5 inhibition on pulmonary artery endothelial dysfunction induced by cigarette smoke in vitro. Human pulmonary artery endothelial cells (HPAEC) were incubated in the absence or presence of PDE5 inhibitor sildenafil (10 nM-1 microM), PKG agonist 8-Br-cGMP (1mM), or the antioxidants dyphenyleneiodonium (DPI 1 microM) and N-acetylcysteine (NAC 1mM) for 30 min. Then, cigarette smoke extract (CSE) was added for 24h. CSE (2.5-10%)-induced ROS generation was suppressed by DPI, and partially reversed by sildenafil and 8-Br-cGMP. Decreases in intracellular levels of cGMP and extracellular NO induced by CSE were reversed by sildenafil and DPI. Furthermore, CSE-induced pg91(phox) and PDE5 mRNA overexpression were suppressed by both sildenafil and DPI. CSE (2.5-10%) induced upregulation of IL-6, IL-8 and Ang-2, and decreased Ang-1 expression in parallel to apoptosis which were partially suppressed by sildenafil, 8-Br-cGMP, DPI and NAC. This study demonstrates that PDE5 inhibition attenuates the oxidant burden and the inflammatory and remodeling effects of CSE in human HPAEC which may contribute to the therapeutic value of PDE5 inhibitors for pulmonary disorders coursing with endothelial dysfunction.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Pulmón/efectos de los fármacos , Nicotiana , Piperazinas/farmacología , Humo/efectos adversos , Sulfonas/farmacología , Vasodilatadores/farmacología , Secuencia de Bases , Células Cultivadas , Cartilla de ADN , Endotelio Vascular/fisiopatología , Ensayo de Inmunoadsorción Enzimática , Humanos , Pulmón/irrigación sanguínea , Pulmón/fisiopatología , Óxido Nítrico/biosíntesis , Reacción en Cadena de la Polimerasa , Purinas/farmacología , Citrato de Sildenafil
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA