Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
3.
J Pharmacol Exp Ther ; 348(2): 303-10, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24307202

RESUMEN

The novel once-daily ß2-agonist bronchodilator drug olodaterol has recently been shown to be effective in patients with allergic asthma for >24 hours. An increased cholinergic tone common to these patients may decrease the effectiveness of ß2-agonists. This could provide a rationale for combination therapy with olodaterol and the long-acting anticholinergic tiotropium to aim for a once-daily treatment regimen. In guinea pigs, we evaluated the protective effects of olodaterol, alone and in combination with tiotropium, on airway responsiveness to histamine, which is partially mediated by a cholinergic reflex mechanism. In addition, using a guinea pig model of acute allergic asthma, we examined the cooperative effects of these bronchodilators on allergen-induced early (EAR) and late (LAR) asthmatic reactions, airway hyper-responsiveness (AHR) to histamine, and airway inflammation. It was demonstrated that the protective effect of olodaterol against histamine-induced bronchoconstriction was synergistically enhanced and prolonged in the presence of tiotropium. In addition, tiotropium synergistically augmented both the reversal of and the protection against the allergen-induced AHR after the EAR by olodaterol. Olodaterol and tiotropium were highly effective in inhibiting the magnitude of the allergen-induced EAR and LAR, and both reactions were fully inhibited by the combination of these drugs. It is remarkable that these effects were not associated with an effect on inflammatory cell infiltration in the airways. In conclusion, the results indicate that combination therapy with olodaterol and tiotropium may be highly effective in the treatment of allergen-induced asthmatic reactions and AHR.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 2/uso terapéutico , Antialérgicos/uso terapéutico , Benzoxazinas/uso terapéutico , Bronquios/efectos de los fármacos , Modelos Animales de Enfermedad , Hipersensibilidad Respiratoria/tratamiento farmacológico , Derivados de Escopolamina/uso terapéutico , Administración por Inhalación , Agonistas de Receptores Adrenérgicos beta 2/administración & dosificación , Animales , Animales no Consanguíneos , Antialérgicos/administración & dosificación , Benzoxazinas/administración & dosificación , Bronquios/inmunología , Bronquios/metabolismo , Bronquios/patología , Broncoconstricción/efectos de los fármacos , Broncodilatadores/administración & dosificación , Broncodilatadores/uso terapéutico , Antagonistas Colinérgicos/administración & dosificación , Antagonistas Colinérgicos/uso terapéutico , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Sinergismo Farmacológico , Quimioterapia Combinada , Cobayas , Histamina/administración & dosificación , Histamina/metabolismo , Masculino , Sustancias Protectoras/administración & dosificación , Sustancias Protectoras/uso terapéutico , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/metabolismo , Hipersensibilidad Respiratoria/patología , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Derivados de Escopolamina/administración & dosificación , Bromuro de Tiotropio
4.
J Pharmacol Exp Ther ; 346(1): 86-95, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23591997

RESUMEN

Increased extracellular matrix (ECM) deposition and airway smooth muscle (ASM) mass are major contributors to airway remodeling in asthma. Recently, we demonstrated that the ECM protein collagen I, which is increased surrounding asthmatic ASM, induces a proliferative, hypocontractile ASM phenotype. Little is known, however, about the signaling pathways involved. Using bovine tracheal smooth muscle, we investigated the role of focal adhesion kinase (FAK) and downstream signaling pathways in collagen I-induced ASM phenotype modulation. Phosphorylation of FAK was increased during adhesion to both uncoated and collagen I-coated culture dishes, without differences between these matrices. Nor were any differences found in cellular adhesion. Inhibition of FAK activity by overexpression of the FAK deletion mutants FAT (focal adhesion targeting domain) and FRNK (FAK-related nonkinase) attenuated adhesion. After attachment, FAK phosphorylation increased in a time-dependent manner in cells cultured on collagen I, whereas no activation was found on an uncoated plastic matrix. In addition, collagen I increased in a time- and concentration-dependent manner the cell proliferation, which was fully inhibited by FAT and FRNK. Similarly, the specific pharmacologic FAK inhibitor PF-573228 [6-((4-((3-(methanesulfonyl)benzyl)amino)-5-trifluoromethylpyrimidin-2-yl) amino)-3,4-dihydro-1H-quinolin-2-one] as well as specific inhibitors of p38 mitogen-activated protein kinase (MAPK) and Src also fully inhibited collagen I-induced proliferation, whereas partial inhibition was observed by inhibition of phosphatidylinositol-3-kinase (PI3-kinase) and mitogen-activated protein kinase kinase (MEK). The inhibition of cell proliferation by these inhibitors was associated with attenuation of the collagen I-induced hypocontractility. Collectively, the results indicate that induction of a proliferative, hypocontractile ASM phenotype by collagen I is mediated by FAK and downstream signaling pathways.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias) , Asma/metabolismo , Colágeno Tipo I/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Sistema de Señalización de MAP Quinasas , Contracción Muscular , Músculo Liso/metabolismo , Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Animales , Asma/tratamiento farmacológico , Bovinos , Adhesión Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Colágeno Tipo I/antagonistas & inhibidores , Activación Enzimática , Proteína-Tirosina Quinasas de Adhesión Focal/antagonistas & inhibidores , Proteína-Tirosina Quinasas de Adhesión Focal/química , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Terapia Molecular Dirigida , Contracción Muscular/efectos de los fármacos , Músculo Liso/citología , Músculo Liso/efectos de los fármacos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Fosforilación , Dominios y Motivos de Interacción de Proteínas , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional , Proteínas Tirosina Quinasas/biosíntesis , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Técnicas de Cultivo de Tejidos , Tráquea/citología , Tráquea/metabolismo
5.
Am J Respir Cell Mol Biol ; 49(1): 18-27, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23449734

RESUMEN

Transforming growth factor-ß1 (TGF-ß1) is a central mediator in tissue remodeling processes, including fibrosis and airway smooth muscle (ASM) hyperplasia, as observed in asthma. The mechanisms underlying this response, however, remain unclear because TGF-ß1 exerts only weak mitogenic effects on ASM cells. In this study, we hypothesized that the mitogenic effect of TGF-ß1 on ASM is indirect and requires prolonged exposure to allow for extracellular matrix (ECM) deposition. To address this hypothesis, we investigated the effects of acute and prolonged treatment with TGF-ß1, alone and in combination with the muscarinic receptor agonist methacholine, on human ASM cell proliferation. Acutely, TGF-ß1 exerted no mitogenic effect. However, prolonged treatment (for 7 d) with TGF-ß1 increased ASM cell proliferation and potentiated the platelet-derived growth factor-induced mitogenic response. Muscarinic receptor stimulation with methacholine synergistically enhanced the effect of TGF-ß1. Interestingly, the integrin-blocking peptide Arg-Gly-Asp-Ser, as well as integrin α5ß1 function-blocking antibodies, inhibited the effects of TGF-ß1 and its combination with methacholine on cell proliferation. Accordingly, prolonged treatment with TGF-ß1 increased fibronectin expression, which was also synergistically enhanced by methacholine. The synergistic effects of methacholine on TGF-ß1-induced proliferation were reduced by the long-acting muscarinic receptor antagonist tiotropium and the M2 receptor subtype-selective antagonist gallamine, but not the M3-selective antagonist DAU5884. In line with these findings, the irreversible Gi protein inhibitor pertussis toxin also prevented the potentiation of TGF-ß1-induced proliferation by methacholine. We conclude that prolonged exposure to TGF-ß1 enhances ASM cell proliferation, which is mediated by extracellular matrix-integrin interactions, and which can be enhanced by muscarinic M2 receptor stimulation.


Asunto(s)
Proliferación Celular , Miocitos del Músculo Liso/efectos de los fármacos , Receptor Cross-Talk , Receptor Muscarínico M2/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Línea Celular Transformada , Medio de Cultivo Libre de Suero , Replicación del ADN , Sinergismo Farmacológico , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Trietyoduro de Galamina/farmacología , Humanos , Integrina alfa5beta1/metabolismo , Cloruro de Metacolina/farmacología , Mitógenos/farmacología , Miocitos del Músculo Liso/metabolismo , Oligopéptidos/farmacología , Toxina del Pertussis/farmacología , Receptor Muscarínico M2/agonistas , Sistema Respiratorio/citología , Factores de Tiempo
6.
Pulm Pharmacol Ther ; 26(1): 145-55, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22842340

RESUMEN

Since ancient times, anticholinergics have been used as a bronchodilator therapy for obstructive lung diseases. Targets of these drugs are G-protein-coupled muscarinic M(1), M(2) and M(3) receptors in the airways, which have long been recognized to regulate vagally-induced airway smooth muscle contraction and mucus secretion. However, recent studies have revealed that acetylcholine also exerts pro-inflammatory, pro-proliferative and pro-fibrotic actions in the airways, which may involve muscarinic receptor stimulation on mesenchymal, epithelial and inflammatory cells. Moreover, acetylcholine in the airways may not only be derived from vagal nerves, but also from non-neuronal cells, including epithelial and inflammatory cells. Airway smooth muscle cells seem to play a major role in the effects of acetylcholine on airway function. It has become apparent that these cells are multipotent cells that may reversibly adopt (hyper)contractile, proliferative and synthetic phenotypes, which are all under control of muscarinic receptors and differentially involved in bronchoconstriction, airway remodeling and inflammation. Cholinergic contractile tone is increased by airway inflammation associated with asthma and COPD, resulting from exaggerated acetylcholine release as well as increased expression of contraction related proteins in airway smooth muscle. Moreover, muscarinic receptor stimulation promotes proliferation of airway smooth muscle cells as well as fibroblasts, and regulates cytokine, chemokine and extracellular matrix production by these cells, which may contribute to airway smooth muscle growth, airway fibrosis and inflammation. In line, animal models of chronic allergic asthma and COPD have recently demonstrated that tiotropium may potently inhibit airway inflammation and remodeling. These observations indicate that muscarinic receptors have a much larger role in the pathophysiology of obstructive airway diseases than previously thought, which may have important therapeutic implications.


Asunto(s)
Asma/tratamiento farmacológico , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Receptores Muscarínicos/metabolismo , Acetilcolina/metabolismo , Remodelación de las Vías Aéreas (Respiratorias) , Animales , Asma/fisiopatología , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Humanos , Inflamación/tratamiento farmacológico , Inflamación/patología , Terapia Molecular Dirigida , Contracción Muscular , Miocitos del Músculo Liso/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología
7.
J Pharmacol Exp Ther ; 342(3): 780-7, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22685341

RESUMEN

Airway remodeling, including increased airway smooth muscle (ASM) mass and contractility, contributes to increased airway narrowing in asthma. Increased ASM mass may be caused by exposure to mitogens, including platelet-derived growth factor (PDGF) and collagen type I, which induce a proliferative, hypocontractile ASM phenotype. In contrast, prolonged exposure to insulin induces a hypercontractile phenotype. Glucocorticosteroids and ß2-adrenoceptor agonists synergize to increase glucocorticosteroid receptor translocation in ASM cells; however, the impact of this synergism on phenotype modulation is unknown. Using bovine tracheal smooth muscle, we investigated the effects of the glucocorticosteroids fluticasone (10 nM), budesonide (30 nM), and dexamethasone (0.1-1 µM) and the combination of low concentrations of fluticasone (3-100 pM) and fenoterol (10 nM) on ASM phenotype switching in response to PDGF (10 ng/ml), collagen type I (50 µg/ml), and insulin (1 µM). All glucocorticosteroids inhibited PDGF- and collagen I-induced proliferation and hypocontractility, with the effects of collagen I being less susceptible to glucocorticosteroid action. At 100-fold lower concentrations, fluticasone (100 pM) synergized with fenoterol to prevent PDGF- and collagen I-induced phenotype switching. This inhibition of ASM phenotype switching was associated with a normalization of the PDGF-induced decrease in the cell cycle inhibitors p21(WAF1/CIP1) and p57(KIP2). At this concentration, fluticasone also prevented the insulin-induced hypercontractile phenotype. At even lower concentrations, fluticasone (3 pM) synergized with fenoterol to inhibit this phenotype switch. Collectively, these findings indicate that glucocorticosteroids and ß2-agonists synergistically inhibit ASM phenotype switching, which may contribute to the increased effectiveness of combined treatment with glucocorticosteroids and ß2-agonists in asthma.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 2/farmacología , Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Glucocorticoides/farmacología , Músculo Liso/efectos de los fármacos , Remodelación de las Vías Aéreas (Respiratorias)/fisiología , Androstadienos/farmacología , Animales , Budesonida/farmacología , Bovinos , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Colágeno Tipo I/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Dexametasona/farmacología , Sinergismo Farmacológico , Fenoterol/farmacología , Fluticasona , Insulina/metabolismo , Mitógenos/farmacología , Contracción Muscular/efectos de los fármacos , Músculo Liso/metabolismo , Músculo Liso/fisiología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/fisiología , Fenotipo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Sistema Respiratorio/efectos de los fármacos , Sistema Respiratorio/metabolismo , Tráquea/efectos de los fármacos , Tráquea/metabolismo , Tráquea/fisiología
8.
Am J Physiol Lung Cell Mol Physiol ; 303(3): L272-8, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22523282

RESUMEN

Chronic obstructive pulmonary disease (COPD) is an inflammatory disease, characterized by a progressive decline in lung function. Airway smooth muscle (ASM) mass may be increased in COPD, contributing to airflow limitation and proinflammatory cytokine production. Cigarette smoke (CS), the major risk factor of COPD, causes ASM cell proliferation, as well as interleukin-8 (IL-8)-induced neutrophilia. In various cell types, transforming growth factor-ß-activated kinase 1 (TAK1) plays a crucial role in MAP kinase and NF-κB activation, as well as IL-8 release induced by IL-1ß, TNF-α, and lipopolysaccharide. The role of TAK1 in CS-induced IL-8 release is not known. The aim of this study was to investigate the role of TAK1 in CS-induced NF-κB and MAP kinase signaling and IL-8 release by human ASM cells. Stimulation of these cells with CS extract (CSE) increased IL-8 release and ERK-1/2 phosphorylation, as well as Iκ-Bα degradation and p65 NF-κB subunit phosphorylation. CSE-induced ERK-1/2 phosphorylation and Iκ-Bα degradation were both inhibited by pretreatment with the specific TAK1 inhibitor LL-Z-1640-2 (5Z-7-oxozeaenol; 100 nM). Similarly, expression of dominant-negative TAK1 inhibited CSE-induced ERK-1/2 phosphorylation. In addition, inhibitors of TAK1 and the NF-κB (SC-514; 50 µM) and ERK-1/2 (U-0126; 3 µM) signaling inhibited the CSE-induced IL-8 release by ASM cells. These data indicate that TAK1 plays a major role in CSE-induced ERK-1/2 and NF-κB signaling and in IL-8 release by human ASM cells. Furthermore, they identify TAK1 as a novel target for the inhibition of CS-induced inflammatory responses involved in the development and progression of COPD.


Asunto(s)
Bronquios/efectos de los fármacos , Interleucina-8/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Fumar/efectos adversos , Western Blotting , Bronquios/citología , Bronquios/metabolismo , Células Cultivadas , Humanos , Quinasa I-kappa B/antagonistas & inhibidores , Quinasa I-kappa B/metabolismo , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , FN-kappa B/metabolismo
9.
Br J Pharmacol ; 166(1): 359-67, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22053853

RESUMEN

BACKGROUND AND PURPOSE: Airway smooth muscle (ASM) phenotype plasticity, characterized by reversible switching between contractile and proliferative phenotypes, is considered to contribute to increased ASM mass and airway hyper-responsiveness in asthma. Further, increased expression of collagen I has been observed within the ASM bundle of asthmatics. Previously, we showed that exposure of intact bovine tracheal smooth muscle (BTSM) to collagen I induces a switch from a contractile to a hypocontractile, proliferative phenotype. However, the functional relevance of this finding for intact human ASM has not been established. EXPERIMENTAL APPROACH: We investigated the effects of exposure of human tracheal smooth muscle (HTSM) strips to monomeric collagen I and PDGF on contractile responses to methacholine and KCl. Expression of contractile proteins sm-α-actin and sm-MHC was assessed by Western blot analysis. The proliferation of HTSM cells was assessed by cell counting, measuring mitochondrial activity (Alamarblue conversion) and [(3) H]-thymidine incorporation. Proliferation of intact tissue slices was assessed by [(3) H]-thymidine incorporation. KEY RESULTS: Culturing HTSM strips in the presence of collagen I or PDGF for 4 days reduced maximal contractile responses to methacholine or KCl and the expression of contractile proteins. Conversely, collagen I and PDGF increased proliferation of HTSM cells and proliferative responses in tissue slices. PDGF additively increased the proliferation of HTSM cells cultured on collagen I; this additive effect was not observed on contractility, contractile protein expression or proliferation of intact tissue. CONCLUSION AND IMPLICATIONS: These findings indicate that collagen I and PDGF induce a functionally hypocontractile, proliferative phenotype of human ASM, which may contribute to airway remodelling in asthma.


Asunto(s)
Colágeno Tipo I/farmacología , Miocitos del Músculo Liso/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Tráquea/efectos de los fármacos , Actinas/metabolismo , Western Blotting , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Humanos , Cloruro de Metacolina/farmacología , Miocitos del Músculo Liso/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Fenotipo , Cloruro de Potasio/farmacología , Tráquea/citología , Tráquea/metabolismo
10.
Am J Physiol Lung Cell Mol Physiol ; 301(5): L822-8, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21873447

RESUMEN

Increased airway smooth muscle (ASM) mass is a major feature of airway remodeling in asthma and chronic obstructive pulmonary disease. Growth factors induce a proliferative ASM phenotype, characterized by an increased proliferative state and a decreased contractile protein expression, reducing contractility of the muscle. Transforming growth factor-ß-activated kinase 1 (TAK1), a mitogen-activated protein kinase kinase kinase, is a key enzyme in proinflammatory signaling in various cell types; however, its function in ASM is unknown. The aim of this study was to investigate the role of TAK1 in growth factor-induced phenotypic modulation of ASM. Using bovine tracheal smooth muscle (BTSM) strips and cells, as well as human tracheal smooth muscle cells, we investigated the role of TAK1 in growth factor-induced proliferation and hypocontractility. Platelet-derived growth factor- (PDGF; 10 ng/ml) and fetal bovine serum (5%)-induced increases in DNA synthesis and cell number in bovine and human cells were significantly inhibited by pretreatment with the specific TAK1 inhibitor LL-Z-1640-2 (5Z-7-oxozeaenol; 100 nM). PDGF-induced DNA synthesis and extracellular signal-regulated kinase-1/2 phosphorylation in BTSM cells were strongly inhibited by both LL-Z-1640-2 pretreatment and transfection of dominant-negative TAK1. In addition, LL-Z-1640-2 inhibited PDGF-induced reduction of BTSM contractility and smooth muscle α-actin expression. The data indicate that TAK1 plays a major role in growth factor-induced phenotypic modulation of ASM.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Asma/metabolismo , Contracción Muscular/efectos de los fármacos , Músculo Liso/metabolismo , Miocitos del Músculo Liso/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Tráquea/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Asma/patología , Asma/fisiopatología , Western Blotting , Bovinos , Proliferación Celular/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Contracción Muscular/fisiología , Músculo Liso/citología , Miocitos del Músculo Liso/citología , Fosforilación/efectos de los fármacos , Plásmidos , Enfermedad Pulmonar Obstructiva Crónica/patología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Transducción de Señal/efectos de los fármacos , Tráquea/citología , Transfección , Zearalenona/análogos & derivados , Zearalenona/farmacología
11.
J Cell Mol Med ; 15(11): 2430-42, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21199324

RESUMEN

Airway smooth muscle cells exhibit phenotype plasticity that underpins their ability to contribute both to acute bronchospasm and to the features of airway remodelling in chronic asthma. A feature of mature, contractile smooth muscle cells is the presence of abundant caveolae, plasma membrane invaginations that develop from the association of lipid rafts with caveolin-1, but the functional role of caveolae and caveolin-1 in smooth muscle phenotype plasticity is unknown. Here, we report a key role for caveolin-1 in promoting phenotype maturation of differentiated airway smooth muscle induced by transforming growth factor (TGF)-ß(1). As assessed by Western analysis and laser scanning cytometry, caveolin-1 protein expression was selectively enriched in contractile phenotype airway myocytes. Treatment with TGF-ß(1) induced profound increases in the contractile phenotype markers sm-α-actin and calponin in cells that also accumulated abundant caveolin-1; however, siRNA or shRNAi inhibition of caveolin-1 expression largely prevented the induction of these contractile phenotype marker proteins by TGF-ß(1). The failure by TGF-ß(1) to adequately induce the expression of these smooth muscle specific proteins was accompanied by a strongly impaired induction of eukaryotic initiation factor-4E binding protein(4E-BP)1 phosphorylation with caveolin-1 knockdown, indicating that caveolin-1 expression promotes TGF-ß(1) signalling associated with myocyte maturation and hypertrophy. Furthermore, we observed increased expression of caveolin-1 within the airway smooth muscle bundle of guinea pigs repeatedly challenged with allergen, which was associated with increased contractile protein expression, thus providing in vivo evidence linking caveolin-1 expression with accumulation of contractile phenotype myocytes. Collectively, we identify a new function for caveolin-1 in controlling smooth muscle phenotype; this mechanism could contribute to allergic asthma.


Asunto(s)
Caveolina 1/metabolismo , Contracción Muscular , Miocitos del Músculo Liso/metabolismo , Sistema Respiratorio/metabolismo , Actinas/biosíntesis , Remodelación de las Vías Aéreas (Respiratorias) , Animales , Asma/fisiopatología , Proteínas de Unión al Calcio , Caveolas/metabolismo , Caveolas/fisiología , Caveolina 1/genética , Células Cultivadas , Perros , Factor 4E Eucariótico de Iniciación/metabolismo , Cobayas , Humanos , Proteínas de Microfilamentos , Células Musculares , Miocitos del Músculo Liso/fisiología , Fenotipo , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Calponinas
12.
Respir Res ; 11: 170, 2010 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-21129174

RESUMEN

BACKGROUND: Fibroproliferative airway remodelling, including increased airway smooth muscle (ASM) mass and contractility, contributes to airway hyperresponsiveness in asthma. In vitro studies have shown that maturation of ASM cells to a (hyper)contractile phenotype is dependent on laminin, which can be inhibited by the laminin-competing peptide Tyr-Ile-Gly-Ser-Arg (YIGSR). The role of laminins in ASM remodelling in chronic asthma in vivo, however, has not yet been established. METHODS: Using an established guinea pig model of allergic asthma, we investigated the effects of topical treatment of the airways with YIGSR on features of airway remodelling induced by repeated allergen challenge, including ASM hyperplasia and hypercontractility, inflammation and fibrosis. Human ASM cells were used to investigate the direct effects of YIGSR on ASM proliferation in vitro. RESULTS: Topical administration of YIGSR attenuated allergen-induced ASM hyperplasia and pulmonary expression of the proliferative marker proliferating cell nuclear antigen (PCNA). Treatment with YIGSR also increased both the expression of sm-MHC and ASM contractility in saline- and allergen-challenged animals; this suggests that treatment with the laminin-competing peptide YIGSR mimics rather than inhibits laminin function in vivo. In addition, treatment with YIGSR increased allergen-induced fibrosis and submucosal eosinophilia. Immobilized YIGSR concentration-dependently reduced PDGF-induced proliferation of cultured ASM to a similar extent as laminin-coated culture plates. Notably, the effects of both immobilized YIGSR and laminin were antagonized by soluble YIGSR. CONCLUSION: These results indicate that the laminin-competing peptide YIGSR promotes a contractile, hypoproliferative ASM phenotype in vivo, an effect that appears to be linked to the microenvironment in which the cells are exposed to the peptide.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Asma/metabolismo , Laminina/metabolismo , Pulmón/fisiopatología , Contracción Muscular/efectos de los fármacos , Músculo Liso/fisiopatología , Oligopéptidos/administración & dosificación , Administración Tópica , Animales , Modelos Animales de Enfermedad , Cobayas , Humanos , Laminina/antagonistas & inhibidores , Pulmón/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Fenotipo
13.
Respir Res ; 11: 48, 2010 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-20429916

RESUMEN

BACKGROUND: A major feature of chronic obstructive pulmonary disease (COPD) is airway remodelling, which includes an increased airway smooth muscle (ASM) mass. The mechanisms underlying ASM remodelling in COPD are currently unknown. We hypothesized that cigarette smoke (CS) and/or lipopolysaccharide (LPS), a major constituent of CS, organic dust and gram-negative bacteria, that may be involved in recurrent airway infections and exacerbations in COPD patients, would induce phenotype changes of ASM. METHODS: To this aim, using cultured bovine tracheal smooth muscle (BTSM) cells and tissue, we investigated the direct effects of CS extract (CSE) and LPS on ASM proliferation and contractility. RESULTS: Both CSE and LPS induced a profound and concentration-dependent increase in DNA synthesis in BTSM cells. CSE and LPS also induced a significant increase in BTSM cell number, which was associated with increased cyclin D1 expression and dependent on activation of ERK 1/2 and p38 MAP kinase. Consistent with a shift to a more proliferative phenotype, prolonged treatment of BTSM strips with CSE or LPS significantly decreased maximal methacholine- and KCl-induced contraction. CONCLUSIONS: Direct exposure of ASM to CSE or LPS causes the induction of a proliferative, hypocontractile ASM phenotype, which may be involved in airway remodelling in COPD.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Lipopolisacáridos/farmacología , Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Humo/efectos adversos , Fumar/efectos adversos , Tráquea/efectos de los fármacos , Animales , Bovinos , Células Cultivadas , Ciclina D1/metabolismo , Replicación del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Flavonoides/farmacología , Imidazoles/farmacología , Contracción Isométrica/efectos de los fármacos , Cloruro de Metacolina/farmacología , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Músculo Liso/patología , Miocitos del Músculo Liso/patología , Fenotipo , Fosforilación , Cloruro de Potasio/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Enfermedad Pulmonar Obstructiva Crónica/patología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Pirimidinas/farmacología , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Tráquea/patología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Am J Respir Crit Care Med ; 181(6): 556-65, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20019343

RESUMEN

RATIONALE: Airway remodeling, including increased airway smooth muscle (ASM) mass and contractility, contributes to airway hyperresponsiveness in asthma. The mechanisms driving these changes are, however, incompletely understood. Recently, an important role for extracellular matrix proteins in regulating ASM proliferation and contractility has been found, suggesting that matrix proteins and their integrins actively modulate airway remodeling. OBJECTIVES: To investigate the role of RGD (Arg-Gly-Asp)-binding integrins in airway remodeling in an animal model of allergic asthma. METHODS: Using a guinea pig model of allergic asthma, the effects of topical application of the integrin-blocking peptide RGDS (Arg-Gly-Asp-Ser) and its negative control GRADSP (Gly-Arg-Ala-Asp-Ser-Pro) were assessed on markers of ASM remodeling, fibrosis, and inflammation induced by repeated allergen challenge. In addition, effects of these peptides on human ASM proliferation and maturation were investigated in vitro. MEASUREMENTS AND MAIN RESULTS: RGDS attenuated allergen-induced ASM hyperplasia and hypercontractility as well as increased pulmonary expression of smooth muscle myosin heavy chain and the proliferative marker proliferating cell nuclear antigen (PCNA). No effects were observed for GRADSP. The RGDS effects were ASM selective, as allergen-induced eosinophil and neutrophil infiltration as well as fibrosis were unaffected. In cultured human ASM cells, we demonstrated that proliferation induced by collagen I, fibronectin, serum, and platelet-derived growth factor requires signaling via RGD-binding integrins, particularly of the alpha(5)beta(1) subtype. In addition, RGDS inhibited smooth muscle alpha-actin accumulation in serum-deprived ASM cells. CONCLUSIONS: This is the first study indicating that integrins modulate ASM remodeling in an animal model of allergic asthma, which can be inhibited by a small peptide containing the RGD motif.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Asma/fisiopatología , Músculo Liso/efectos de los fármacos , Oligopéptidos/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Administración Tópica , Remodelación de las Vías Aéreas (Respiratorias)/inmunología , Animales , Anticuerpos/inmunología , Asma/inmunología , Biomarcadores , Western Blotting , Técnicas de Cultivo de Célula , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Fibrosis/inmunología , Cobayas , Humanos , Inflamación/inmunología , Masculino , Contracción Muscular/efectos de los fármacos , Músculo Liso/inmunología , Oligopéptidos/inmunología , Inhibidores de Agregación Plaquetaria/inmunología , Antígeno Nuclear de Célula en Proliferación/efectos de los fármacos , Antígeno Nuclear de Célula en Proliferación/inmunología
15.
Hepatology ; 50(6): 1924-35, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19842096

RESUMEN

UNLABELLED: Increased intrahepatic resistance and splanchnic blood flow cause portal hypertension in liver cirrhosis. Nonselective beta-adrenoceptor (beta-AR) antagonists have beneficial effects on hyperdynamic circulation and are in clinical use. In this context, the role of the beta(3)-AR is undefined. Here we investigated their expression and role in portal hypertension in patients and rats with liver cirrhosis. We analyzed cirrhotic human and rat tissues (liver, splanchnic vessels) and primary rat cells. Protein expression of beta(3)-AR was determined by western blot and messenger RNA (mRNA) levels by reverse-transcription polymerase chain reaction (RT-PCR). Activities of Rho-kinase and the nitric oxide (NO) effector protein kinase G (PKG) were assessed by way of substrate phosphorylation (moesin, vasodilator-stimulated phosphoprotein [VASP]). Cyclic 3',5' adenosine monophosphate (cAMP) accumulation was determined by an enzyme-immunoassay kit. The effects of selective beta(3)-AR agonists (CGP12177A, BRL37344) and antagonist (SR59230A) were investigated by collagen matrix contraction of hepatic stellate cells (HSCs), in situ liver perfusions, and in vivo hemodynamic parameters in bile duct ligation and carbon tetrachloride intoxication in cirrhotic rats. In cirrhosis of humans and rats, beta(3)-AR expression is markedly increased in hepatic and in splanchnic tissues. Stimulation of beta(3)-AR leads to relaxation of HSCs by way of cAMP accumulation, and by inhibition of Rho-kinase activity; any role of NO and its effector PKG was not observed. beta(3)-AR agonists decrease intrahepatic resistance and portal pressure in cirrhotic rats. CONCLUSION: There is a marked hepatic and mesenteric up-regulation of beta(3)-ARs in human cirrhosis and in two different animal models of cirrhosis. The beta(3)-AR-agonists should be further evaluated for therapy of portal hypertension.


Asunto(s)
Hipertensión Portal/etiología , Cirrosis Hepática/metabolismo , Receptores Adrenérgicos beta 3/fisiología , Resistencia Vascular , Animales , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Células Estrelladas Hepáticas/fisiología , Humanos , Propanolaminas/farmacología , Propranolol/farmacología , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 3/genética
16.
Br J Pharmacol ; 158(3): 652-64, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19703164

RESUMEN

Allergic asthma is a chronic inflammatory airways' disease, characterized by allergen-induced early and late bronchial obstructive reactions, airway hyperresponsiveness (AHR), airway inflammation and airway remodelling. Recent ex vivo and in vivo studies in animal models and asthmatic patients have indicated that arginase may play a central role in all these features. Thus, increased arginase activity in the airways induces reduced bioavailability of L-arginine to constitutive (cNOS) and inducible (iNOS) nitric oxide synthases, causing a deficiency of bronchodilating and anti-inflammatory NO, as well as increased formation of peroxynitrite, which may be involved in allergen-induced airways obstruction, AHR and inflammation. In addition, both via reduced NO production and enhanced synthesis of L-ornithine, increased arginase activity may be involved in airway remodelling by promoting cell proliferation and collagen deposition in the airway wall. Therefore, arginase inhibitors may have therapeutic potential in the treatment of acute and chronic asthma. This review focuses on the pathophysiological role of arginase in allergic asthma and the emerging effectiveness of arginase inhibitors in the treatment of this disease.


Asunto(s)
Arginasa/fisiología , Asma/tratamiento farmacológico , Asma/enzimología , Remodelación de las Vías Aéreas (Respiratorias) , Animales , Antialérgicos/uso terapéutico , Antiinflamatorios/uso terapéutico , Arginasa/antagonistas & inhibidores , Asma/fisiopatología , Hiperreactividad Bronquial/enzimología , Hiperreactividad Bronquial/fisiopatología , Humanos , Inflamación/enzimología , Inflamación/fisiopatología , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa/fisiología , Fenómenos Fisiológicos Respiratorios
17.
Naunyn Schmiedebergs Arch Pharmacol ; 380(1): 67-77, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19255745

RESUMEN

In human bronchial epithelial (16HBE14o(-)) cells, CB(1) and CB(2) cannabinoid receptors are present, and their activation by the endocannabinoid virodhamine and the synthetic non-selective receptor agonist CP55,940 inhibits adenylyl cyclase and cellular interleukin-8 release. Here, we analyzed changes in intracellular calcium ([Ca2+](i)) evoked by Delta(9)-tetrahydrocannabinol (Delta(9)-THC), CP55,940, and virodhamine in 16HBE14o(-) cells. Delta(9)-THC induced [Ca2+](i) increase and a large transient [Ca2+](i) mobilization, the latter probably reflecting store-depletion-driven capacitative Ca2+ entry (CCE). In contrast, CP55,940 induced a rather moderate Ca2+ influx and a sustained [Ca2+](i) mobilization. CP55,940-induced Ca2+ influx was inhibited by Ni2+, indicating CCE, possibly mediated by transient receptor potential channel TRPC1, the mRNA of which is expressed in 16HBE14o(-) cells. CP55,940-induced calcium alterations were mimicked by virodhamine concentrations below 30 microM. Interestingly, higher virodhamine induced an additional Ca2+ entry, insensitive to Ni2+, but sensitive to the TRPV1 antagonist capsazepine, the TRPV1-TRPV4 inhibitor ruthenium red, and the non-CCE (NCCE) inhibitors La3+ and Gd3+. Such pharmacological profile is supported by the presence of TRPV1, TRPV4, and TRPC6 mRNAs as well as TRPV1 and TRPC6 proteins in 16HBE14o(-) cells. Cannabinoid receptor antagonists increased virodhamine-induced Ca2+ entry. Virodhamine also enhanced arachidonic acid release, which was insensitive to cannabinoid receptor antagonism, but sensitive to the phospholipase A(2) inhibitor quinacrine, and to capsazepine. Arachidonic acid induced [Ca2+](i) increase similar to virodhamine. Collectively, these observations suggest that [Ca2+](i) alterations induced by Delta(9)-THC, CP55,940 and by low concentrations of virodhamine involve mobilization and subsequent CCE mechanisms, whereas such responses by high virodhamine concentrations involve NCCE pathways.


Asunto(s)
Calcio/metabolismo , Moduladores de Receptores de Cannabinoides/farmacología , Cannabinoides/farmacología , Ácido Araquidónico/metabolismo , Ácidos Araquidónicos/administración & dosificación , Ácidos Araquidónicos/farmacología , Bronquios/citología , Bronquios/efectos de los fármacos , Bronquios/metabolismo , Moduladores de Receptores de Cannabinoides/administración & dosificación , Cannabinoides/administración & dosificación , Línea Celular , Ciclohexanoles/administración & dosificación , Ciclohexanoles/farmacología , Relación Dosis-Respuesta a Droga , Dronabinol/administración & dosificación , Dronabinol/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , ARN Mensajero/metabolismo , Canales Catiónicos TRPC/metabolismo
18.
Am J Respir Cell Mol Biol ; 41(4): 494-504, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19213874

RESUMEN

Airway smooth muscle (ASM) plays a key role in the development of airway hyperresponsiveness and remodeling in asthma, which may involve maturation of ASM cells to a hypercontractile phenotype. In vitro studies have indicated that long-term exposure of bovine tracheal smooth muscle (BTSM) to insulin induces a functional hypercontractile, hypoproliferative phenotype. Similarly, the extracellular matrix protein laminin has been found to be involved in both the induction and maintenance of a contractile ASM phenotype. Using BTSM, we now investigated the role of laminins in the insulin-induced hypercontractile, hypoproliferative ASM phenotype. The results demonstrate that insulin-induced hypercontractility after 8 days of tissue culture was fully prevented by combined treatment of BTSM-strips with the laminin competing peptides Tyr-Ile-Gly-Ser-Arg (YIGSR) and Arg-Gly-Asp-Ser (RGDS). YIGSR also prevented insulin-induced increases in sm-myosin expression and abrogated the suppressive effects of prolonged insulin treatment on platelet-derived growth factor-induced DNA synthesis in cultured cells. In addition, insulin time-dependently increased laminin alpha2, beta1, and gamma1 chain protein, but not mRNA abundance in BTSM strips. Moreover, as previously found for contractile protein accumulation, signaling through PI3-kinase- and Rho kinase-dependent pathways was required for the insulin-induced increase in laminin abundance and contractility. Collectively, our results indicate a critical role for beta1-containing laminins, likely laminin-211, in the induction of a hypercontractile, hypoproliferative ASM phenotype by prolonged insulin exposure. Increased laminin production by ASM could be involved in the increased ASM contractility and contractile protein expression in asthma. Moreover, the results may be of interest for the use of inhaled insulin administrations by diabetics.


Asunto(s)
Insulina/farmacología , Laminina/biosíntesis , Contracción Muscular/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Tráquea/efectos de los fármacos , Administración por Inhalación , Animales , Bovinos , Cromonas/farmacología , Replicación del ADN/efectos de los fármacos , Técnicas In Vitro , Insulina/toxicidad , Contracción Isométrica/efectos de los fármacos , Cloruro de Metacolina/farmacología , Ratones , Morfolinas/farmacología , Músculo Liso/metabolismo , Oligopéptidos/farmacología , Fenotipo , Fosfatidilinositol 3-Quinasas/fisiología , Inhibidores de las Quinasa Fosfoinosítidos-3 , ARN Mensajero/análisis , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Tráquea/metabolismo , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/fisiología
19.
Am J Respir Crit Care Med ; 178(6): 565-73, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18583571

RESUMEN

RATIONALE: In a guinea pig model of allergic asthma, using perfused tracheal preparations ex vivo, we demonstrated that L-arginine limitation due to increased arginase activity underlies a deficiency of bronchodilating nitric oxide (NO) and airway hyperresponsiveness (AHR) after the allergen-induced early and late asthmatic reaction. OBJECTIVES: Using the same animal model, we investigated the acute effects of the specific arginase inhibitor 2(S)-amino-6-boronohexanoic acid (ABH) and of L-arginine on AHR after the early and late reaction in vivo. In addition, we investigated the protection of allergen-induced asthmatic reactions, AHR, and airway inflammation by pretreatment with the drug. METHODS: Airway responsiveness to inhaled histamine was measured in permanently instrumented, freely moving guinea pigs sensitized to ovalbumin at 24 hours before allergen challenge and after the allergen-induced early and late asthmatic reactions by assessing histamine PC(100) (provocative concentration causing a 100% increase of pleural pressure) values. MEASUREMENTS AND MAIN RESULTS: Inhaled ABH acutely reversed AHR to histamine after the early reaction from 4.77 +/- 0.56-fold to 2.04 +/- 0.34-fold (P < 0.001), and a tendency to inhibition was observed after the late reaction (from 1.95 +/- 0.56-fold to 1.56 +/- 0.47-fold, P < 0.10). Quantitatively similar results were obtained with inhaled l-arginine. Remarkably, after pretreatment with ABH a 33-fold higher dose of allergen was needed to induce airway obstruction (P < 0.01). Consequently, ABH inhalation 0.5 hour before and 8 hours after allergen challenge protected against the allergen-induced early and late asthmatic reactions, AHR and inflammatory cell infiltration. CONCLUSIONS: Inhalation of ABH or l-arginine acutely reverses allergen-induced AHR after the early and late asthmatic reaction, presumably by attenuating arginase-induced substrate deficiency to NO synthase in the airways. Moreover, ABH considerably reduces the airway sensitivity to inhaled allergen and protects against allergen-induced bronchial obstructive reactions, AHR, and airway inflammation. This is the first in vivo study indicating that arginase inhibitors may have therapeutic potential in allergic asthma.


Asunto(s)
Obstrucción de las Vías Aéreas/prevención & control , Arginasa/antagonistas & inhibidores , Asma/fisiopatología , Hiperreactividad Bronquial/fisiopatología , Obstrucción de las Vías Aéreas/inmunología , Aminocaproatos/farmacología , Animales , Arginasa/metabolismo , Asma/tratamiento farmacológico , Compuestos de Boro/farmacología , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/metabolismo , Pruebas de Provocación Bronquial , Lavado Broncoalveolar , Cobayas , Cloruro de Metacolina/farmacología , Modelos Animales , Óxido Nítrico
20.
Am J Physiol Lung Cell Mol Physiol ; 295(1): L214-9, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18487358

RESUMEN

Recently, we have shown that allergen-induced airway hyperresponsiveness (AHR) after the early (EAR) and late (LAR) asthmatic reaction in guinea pigs could be reversed acutely by inhalation of the Rho kinase inhibitor Y-27632. The present study addresses the effects of pretreatment with inhaled Y-27632 on the severity of the allergen-induced EAR and LAR, the development of AHR after these reactions, and airway inflammation. Using permanently instrumented and unrestrained ovalbumin (OA)-sensitized guinea pigs, single OA challenge-induced EAR and LAR, expressed as area under the lung function (pleural pressure, P(pl)) time-response curve, were measured, and histamine PC(100) (provocation concentration causing a 100% increase of P(pl)) values were assessed 24 h before, and at 6 and 24 h after, the OA challenge (after the EAR and LAR, respectively). Thirty minutes before and 8 h after OA challenge, saline or Y-27632 (5 mM) was nebulized. After the last PC(100) value, bronchoalveolar lavage (BAL) was performed, and the inflammatory cell profile was determined. It was demonstrated that inhalation of Y-27632 before allergen challenge markedly reduced the immediate allergen-induced peak rise in P(pl), without significantly reducing the overall EAR and LAR. Also, pretreatment with Y-27632 considerably protected against the development of AHR after the EAR and fully prevented AHR after the LAR. These effects could not be explained by a direct effect of Y-27632 on the histamine responsiveness, because of the short duration of the acute bronchoprotection of Y-27632 (<90 min). In addition, Y-27632 reduced the number of total inflammatory cells, eosinophils, macrophages, and neutrophils recovered from the BAL. Altogether, inhaled Y-27632 protects against acute allergen-induced bronchoconstriction, development of AHR after the EAR and LAR, and airway inflammation in an established guinea pig model of allergic asthma.


Asunto(s)
Alérgenos/toxicidad , Amidas/farmacología , Asma/prevención & control , Inhibidores Enzimáticos/farmacología , Neutrófilos/enzimología , Piridinas/farmacología , Quinasas Asociadas a rho/antagonistas & inhibidores , Enfermedad Aguda , Animales , Asma/inducido químicamente , Asma/enzimología , Asma/patología , Modelos Animales de Enfermedad , Cobayas , Inflamación/inducido químicamente , Inflamación/enzimología , Inflamación/prevención & control , Masculino , Neutrófilos/patología , Factores de Tiempo , Quinasas Asociadas a rho/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...