Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Biol Rep ; 48(2): 1959-1966, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33590412

RESUMO

Arginine metabolism mediated by arginases plays a critical role in cell and tissue function. The arginine hydrolysis is deeply involved in the urea cycle, which helps the kidney excrete ammonia from blood. Upregulation of arginases affects microenvironment stability due to the presence of excess urea in blood. To regulate the arginase activities properly, a synthetic peptide based on the structure of human arginase I was designed and assessed. Preliminary data shows it inhibits human arginase I and II with an IC50 of 2.4 ± 0.3 and 1.8 ± 0.1 mmol, respectively. Our kinetic analysis indicates the inhibition is not competitive with substrate - suggesting an allosteric mechanism. This result provides a step towards specific inhibitors design.


Assuntos
Arginase/antagonistas & inibidores , Peptídeos/química , Regulação Alostérica , Arginase/química , Arginase/metabolismo , Concentração de Íons de Hidrogênio , Cinética
2.
Clin Sci (Lond) ; 134(11): 1219-1242, 2020 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-32501497

RESUMO

Research using animal models of asthma is currently dominated by mouse models. This has been driven by the comprehensive knowledge on inflammatory and immune reactions in mice, as well as tools to produce genetically modified mice. Many of the identified therapeutic targets influencing airway hyper-responsiveness and inflammation in mouse models, have however been disappointing when tested clinically in asthma. It is therefore a great need for new animal models that more closely resemble human asthma. The guinea pig has for decades been used in asthma research and a comprehensive table of different protocols for asthma models is presented. The studies have primarily been focused on the pharmacological aspects of the disease, where the guinea pig undoubtedly is superior to mice. Further reasons are the anatomical and physiological similarities between human and guinea pig airways compared with that of the mouse, especially with respect to airway branching, neurophysiology, pulmonary circulation and smooth muscle distribution, as well as mast cell localization and mediator secretion. Lack of reagents and specific molecular tools to study inflammatory and immunological reactions in the guinea pig has however greatly diminished its use in asthma research. The aim in this position paper is to review and summarize what we know about different aspects of the use of guinea pig in vivo models for asthma research. The associated aim is to highlight the unmet needs that have to be addressed in the future.


Assuntos
Asma/patologia , Modelos Animais de Doenças , Cobaias/fisiologia , Animais , Desenvolvimento de Medicamentos , Edição de Genes , Cobaias/genética , Pulmão/patologia , Pulmão/fisiopatologia
3.
Am J Physiol Lung Cell Mol Physiol ; 317(6): L768-L777, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31553662

RESUMO

Airway inflammation and remodeling are characteristic features of asthma, with both contributing to airway hyperresponsiveness (AHR) and lung function limitation. Airway smooth muscle (ASM) accumulation and extracellular matrix deposition are characteristic features of airway remodeling, which may contribute to persistent AHR. Laminins containing the α2-chain contribute to characteristics of ASM remodeling in vitro and AHR in animal models of asthma. The role of other laminin chains, including the laminin α4 and α5 chains, which contribute to leukocyte migration in other diseases, is currently unknown. The aim of the current study was to investigate the role of these laminin chains in ASM function and in AHR, remodeling, and inflammation in asthma. Expression of both laminin α4 and α5 was observed in the human and mouse ASM bundle. In vitro, laminin α4 was found to promote a pro-proliferative, pro-contractile, and pro-fibrotic ASM cell phenotype. In line with this, treatment with laminin α4 and α5 function-blocking antibodies reduced allergen-induced increases in ASM mass in a mouse model of allergen-induced asthma. Moreover, eosinophilic inflammation was reduced by the laminin α4 function-blocking antibody as well. Using airway biopsies from healthy subjects and asthmatic patients, we found inverse correlations between ASM α4-chain expression and lung function and AHR, whereas eosinophil numbers correlated positively with expression of laminin α4 in the ASM bundle. This study, for the first time, indicates a prominent role for laminin α4 in ASM function and in inflammation, AHR, and remodeling in asthma, whereas the role of laminin α5 is more subtle.


Assuntos
Remodelação das Vias Aéreas , Asma/complicações , Eosinófilos/patologia , Inflamação/etiologia , Laminina/metabolismo , Adolescente , Adulto , Idoso , Animais , Asma/metabolismo , Asma/patologia , Eosinófilos/metabolismo , Feminino , Humanos , Inflamação/metabolismo , Inflamação/patologia , Laminina/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Contração Muscular , Adulto Jovem
4.
Am J Physiol Lung Cell Mol Physiol ; 316(3): L537-L546, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30628486

RESUMO

The direct relationship between pulmonary structural changes and airway hyperresponsiveness (AHR) in chronic obstructive pulmonary disease (COPD) is unclear. We investigated AHR in relation to airway and parenchymal structural changes in a guinea pig model of COPD and in COPD patients. Precision-cut lung slices (PCLS) were prepared from guinea pigs challenged with lipopolysaccharide or saline two times weekly for 12 wk. Peripheral PCLS were obtained from patients with mild to moderate COPD and non-COPD controls. AHR to methacholine was measured in large and small airways using video-assisted microscopy. Airway smooth muscle mass and alveolar airspace size were determined in the same slices. A mathematical model was used to identify potential changes in biomechanical properties underlying AHR. In guinea pigs, lipopolysaccharide increased the sensitivity of large (>150 µm) airways toward methacholine by 4.4-fold and the maximal constriction of small airways (<150 µm) by 1.5-fold. Similarly increased small airway responsiveness was found in COPD patients. In both lipopolysaccharide-challenged guinea pigs and patients, airway smooth muscle mass was unaltered, whereas increased alveolar airspace correlated with small airway hyperresponsiveness in guinea pigs. Fitting the parameters of the model indicated that COPD weakens matrix mechanical properties and enhances stiffness differences between the airway and the parenchyma, in both species. In conclusion, this study demonstrates small airway hyperresponsiveness in PCLS from COPD patients. These changes may be related to reduced parenchymal retraction forces and biomechanical changes in the airway wall. PCLS from lipopolysaccharide-exposed guinea pigs may be useful to study mechanisms of small airway hyperresponsiveness in COPD.


Assuntos
Remodelação das Vias Aéreas/fisiologia , Músculo Liso/efeitos dos fármacos , Doença Pulmonar Obstrutiva Crônica/patologia , Doença Pulmonar Obstrutiva Crônica/fisiopatologia , Adulto , Idoso , Animais , Asma/patologia , Asma/fisiopatologia , Modelos Animais de Doenças , Feminino , Cobaias , Humanos , Lipopolissacarídeos/farmacologia , Pulmão/patologia , Pulmão/fisiopatologia , Masculino , Pessoa de Meia-Idade , Músculo Liso/fisiopatologia , Doença Pulmonar Obstrutiva Crônica/induzido quimicamente , Hipersensibilidade Respiratória/induzido quimicamente , Hipersensibilidade Respiratória/patologia , Hipersensibilidade Respiratória/fisiopatologia
5.
FASEB J ; 30(5): 1823-35, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26849959

RESUMO

TGF-ß is important in lung injury and remodeling processes. TGF-ß and Wingless/integrase-1 (WNT) signaling are interconnected; however, the WNT ligand-receptor complexes involved are unknown. Thus, we aimed to identify Frizzled (FZD) receptors that mediate TGF-ß-induced profibrotic signaling. MRC-5 and primary human lung fibroblasts were stimulated with TGF-ß1, WNT-5A, or WNT-5B in the presence and absence of specific pathway inhibitors. Specific small interfering RNA was used to knock down FZD8. In vivo studies using bleomycin-induced lung fibrosis were performed in wild-type and FZD8-deficient mice. TGF-ß1 induced FZD8 specifically via Smad3-dependent signaling in MRC-5 and primary human lung fibroblasts. It is noteworthy that FZD8 knockdown reduced TGF-ß1-induced collagen Iα1, fibronectin, versican, α-smooth muscle (sm)-actin, and connective tissue growth factor. Moreover, bleomycin-induced lung fibrosis was attenuated in FZD8-deficient mice in vivo Although inhibition of canonical WNT signaling did not affect TGF-ß1-induced gene expression in vitro, noncanonical WNT-5B mimicked TGF-ß1-induced fibroblast activation. FZD8 knockdown reduced both WNT-5B-induced gene expression of fibronectin and α-sm-actin, as well as WNT-5B-induced changes in cellular impedance. Collectively, our findings demonstrate a role for FZD8 in TGF-ß-induced profibrotic signaling and imply that WNT-5B may be the ligand for FZD8 in these responses.-Spanjer, A. I. R., Baarsma, H. A., Oostenbrink, L. M., Jansen, S. R., Kuipers, C. C., Lindner, M., Postma, D. S., Meurs, H., Heijink, I. H., Gosens, R., Königshoff, M. TGF-ß-induced profibrotic signaling is regulated in part by the WNT receptor Frizzled-8.


Assuntos
Receptores de Superfície Celular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta1/farmacologia , Animais , Linhagem Celular , Matriz Extracelular , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Humanos , Pulmão/citologia , Camundongos , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Superfície Celular/genética , Receptores Acoplados a Proteínas G/genética , Proteína Smad3/genética , Proteína Smad3/metabolismo , Organismos Livres de Patógenos Específicos , Proteínas Wnt/farmacologia , Proteína Wnt-5a/farmacologia
6.
Thorax ; 71(4): 312-22, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26797711

RESUMO

RATIONALE: We have previously shown increased expression of the Frizzled-8 receptor of the Wingless/integrase-1 (WNT) signalling pathway in COPD. Here, we investigated if the Frizzled-8 receptor has a functional role in airway inflammation associated with chronic bronchitis. METHODS: Acute cigarette-smoke-induced airway inflammation was studied in wild-type and Frizzled-8-deficient mice. Genetic association studies and lung expression quantitative trait loci (eQTL) analyses for Frizzled-8 were performed to evaluate polymorphisms in FZD8 and their relationship to tissue expression in chronic bronchitis. Primary human lung fibroblasts and primary human airway epithelial cells were used for in vitro studies. RESULTS: Cigarette-smoke-exposure induced airway inflammation in wild-type mice, which was prevented in Frizzled-8-deficient mice, suggesting a crucial role for Frizzled-8 in airway inflammation. Furthermore, we found a significant genetic association (p=0.009) between single nucleotide polymorphism (SNP) rs663700 in the FZD8 region and chronic mucus hypersecretion, a characteristic of chronic bronchitis, in a large cohort of smoking individuals. We found SNP rs663700 to be a cis-eQTL regulating Frizzled-8 expression in lung tissue. Functional data link mesenchymal Frizzled-8 expression to inflammation as its expression in COPD-derived lung fibroblasts was regulated by pro-inflammatory cytokines in a genotype-dependent manner. Moreover, Frizzled-8 regulates inflammatory cytokine secretion from human lung fibroblasts, which in turn promoted MUC5AC expression by differentiated human airway epithelium. CONCLUSIONS: These findings indicate an important pro-inflammatory role for Frizzled-8 and suggest that its expression is related to chronic bronchitis. Furthermore, our findings indicate an unexpected role for fibroblasts in regulating airway inflammation in COPD.


Assuntos
Bronquite Crônica/genética , Receptores Frizzled/genética , Polimorfismo de Nucleotídeo Único/genética , Animais , Bronquite Crônica/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Fibroblastos/metabolismo , Marcadores Genéticos/genética , Genótipo , Humanos , Técnicas In Vitro , Inflamação/genética , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mucina-5AC/genética , Doença Pulmonar Obstrutiva Crônica/metabolismo , Transdução de Sinais/genética
7.
Respir Res ; 17: 13, 2016 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-26846267

RESUMO

BACKGROUND: The long-acting anticholinergic tiotropium has recently been registered for the treatment of asthma, and its use is associated with a reduction in exacerbation frequency. Anti-inflammatory and anti-remodeling effects of tiotropium have been demonstrated in in vitro and in vivo models. Because tiotropium treatment is used in combination with inhaled corticosteroids, potential additive effects between the two would be clinically relevant. Therefore, the aim of this study was to investigate additive effects between tiotropium and ciclesonide on airway inflammation and remodeling in guinea pig models of asthma. METHODS: Guinea pigs (n = 3-8/group) were sensitized and challenged with ovalbumin in an acute (single challenge) and a chronic model (12 weekly challenges) of allergic asthma. Animals were treated with vehicle, nebulized tiotropium (0.01-0.3 mM) and/or intranasally instilled ciclesonide (0.001-1 mg/kg) before each challenge. Bronchoalveolar lavage fluid and lungs were collected for analysis of airway inflammation and remodeling. RESULTS: Tiotropium and ciclesonide treatment, alone or in combination, did not inhibit airway inflammation in the acute asthma model. In a dose-finding study, low doses of tiotropium and ciclesonide inhibited airway eosinophilia and airway smooth muscle thickening in the chronic asthma model. Threshold doses of 0.01 mM tiotropium (nebulizer concentration) and 0.01 mg/kg ciclesonide were selected to investigate potential additive effects between both drugs. At these doses, tiotropium and ciclesonide did not inhibit airway eosinophilia or airway smooth muscle thickening when administered alone, but significantly inhibited these allergen-induced responses when administered in combination. CONCLUSIONS: Combined treatment with low doses of tiotropium and ciclesonide inhibits airway inflammation and remodeling in a guinea pig model of chronic asthma, suggesting that combined treatment with anticholinergics and corticosteroids may have anti-inflammatory and anti-remodeling activity in allergic airway diseases. Since tiotropium is registered as a therapy for asthma added on to corticosteroid treatment, these beneficial effects of the combination therapy may be clinically relevant.


Assuntos
Remodelação das Vias Aéreas/efeitos dos fármacos , Asma/imunologia , Asma/prevenção & controle , Modelos Animais de Doenças , Pregnenodionas/administração & dosagem , Brometo de Tiotrópio/administração & dosagem , Administração por Inalação , Animais , Antialérgicos/administração & dosagem , Asma/induzido quimicamente , Broncodilatadores/administração & dosagem , Doença Crônica , Relação Dose-Resposta a Droga , Quimioterapia Combinada/métodos , Cobaias , Masculino , Ovalbumina , Resultado do Tratamento
8.
Am J Physiol Lung Cell Mol Physiol ; 308(3): L301-6, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25480330

RESUMO

Hypothyroidism may reduce, whereas hyperthyroidism may aggravate, asthma symptoms. The mechanisms underlying this relationship are largely unknown. Since thyroid hormones have central roles in cell growth and differentiation, we hypothesized that airway remodeling, in particular increased airway smooth muscle (ASM) mass, may be involved. To address this hypothesis, we investigated the effects of triiodothyronine (T3) and l-thyroxine (T4) in the absence and presence of the profibrotic transforming growth factor (TGF)-ß1 on human ASM cell phenotype switching. T3 (1-100 nM) and T4 (1-100 nM) did not affect basal ASM proliferation. However, when combined with TGF-ß1 (2 ng/ml), T4 synergistically increased the proliferative response, whereas only a minor effect was observed for T3. In line with a switch from a contractile to a proliferative ASM phenotype, T4 reduced the TGF-ß1-induced contractile protein expression by ∼50%. Cotreatment with T3 reduced TGF-ß1-induced contractile protein expression by ∼25%. The synergistic increase in proliferation was almost fully inhibited by the integrin αvß3 antagonist tetrac (100 nM), whereas no significant effects of the thyroid receptor antagonist 1-850 (3 µM) were observed. Inhibition of MEK1/2, downstream of the integrin αvß3, also inhibited the T4- and TGF-ß1-induced proliferative responses. Collectively, the results indicate that T4, and to a lesser extent T3, promotes a proliferative ASM phenotype in the presence of TGF-ß1, which is predominantly mediated by the membrane-bound T4 receptor αvß3. These results indicate that thyroid hormones may enhance ASM remodeling in asthma, which could be of relevance for hyperthyroid patients with this disease.


Assuntos
Miócitos de Músculo Liso/fisiologia , Tiroxina/fisiologia , Fator de Crescimento Transformador beta1/fisiologia , Remodelação das Vias Aéreas , Brônquios/patologia , Linhagem Celular , Proliferação de Células , Regulação para Baixo , Expressão Gênica , Humanos , Integrina alfaVbeta3/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Fenótipo , Tri-Iodotironina/fisiologia
9.
Am J Physiol Lung Cell Mol Physiol ; 308(1): L96-103, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25381025

RESUMO

Anticholinergics, blocking the muscarinic M3 receptor, are effective bronchodilators for patients with chronic obstructive pulmonary disease. Recent evidence from M(3) receptor-deficient mice (M(3)R(-/-)) indicates that M3 receptors also regulate neutrophilic inflammation in response to cigarette smoke (CS). M(3) receptors are present on almost all cell types, and in this study we investigated the relative contribution of M(3) receptors on structural cells vs. inflammatory cells to CS-induced inflammation using bone marrow chimeric mice. Bone marrow chimeras (C56Bl/6 mice) were generated, and engraftment was confirmed after 10 wk. Thereafter, irradiated and nonirradiated control animals were exposed to CS or fresh air for four consecutive days. CS induced a significant increase in neutrophil numbers in nonirradiated and irradiated control animals (4- to 35-fold). Interestingly, wild-type animals receiving M(3)R(-/-) bone marrow showed a similar increase in neutrophil number (15-fold). In contrast, no increase in the number of neutrophils was observed in M3R(-/-) animals receiving wild-type bone marrow. The increase in keratinocyte-derived chemokine (KC) levels was similar in all smoke-exposed groups (2.5- to 5.0-fold). Microarray analysis revealed that fibrinogen-α and CD177, both involved in neutrophil migration, were downregulated in CS-exposed M(3)R(-/-) animals receiving wild-type bone marrow compared with CS-exposed wild-type animals, which was confirmed by RT-qPCR (1.6-2.5 fold). These findings indicate that the M(3) receptor on structural cells plays a proinflammatory role in CS-induced neutrophilic inflammation, whereas the M(3) receptor on inflammatory cells does not. This effect is probably not mediated via KC release, but may involve altered adhesion and transmigration of neutrophils via fibrinogen-α and CD177.


Assuntos
Infiltração de Neutrófilos , Neutrófilos/metabolismo , Receptor Muscarínico M3/metabolismo , Transtornos Respiratórios/metabolismo , Fumar/efeitos adversos , Aloenxertos , Animais , Transplante de Medula Óssea , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Regulação para Baixo/genética , Fibrinogênio/genética , Fibrinogênio/metabolismo , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Camundongos Knockout , Neutrófilos/patologia , Receptor Muscarínico M3/genética , Transtornos Respiratórios/etiologia , Transtornos Respiratórios/genética , Transtornos Respiratórios/patologia , Fumar/genética , Fumar/metabolismo , Quimeras de Transplante/genética , Quimeras de Transplante/metabolismo
10.
Thorax ; 70(7): 668-76, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25995156

RESUMO

BACKGROUND: It has been shown that acetylcholine is both a neurotransmitter and acts as a local mediator, produced by airway cells including epithelial cells. In vivo studies have demonstrated an indirect role for acetylcholine in epithelial cell differentiation. Here, we aimed to investigate direct effects of endogenous non-neuronal acetylcholine on epithelial cell differentiation. METHODS: Human airway epithelial cells from healthy donors were cultured at an air-liquid interface (ALI). Cells were exposed to the muscarinic antagonist tiotropium (10 nM), interleukin (IL)-13 (1, 2 and 5 ng/mL), or a combination of IL-13 and tiotropium, during or after differentiation at the ALI. RESULTS: Human airway epithelial cells expressed all components of the non-neuronal cholinergic system, suggesting acetylcholine production. Tiotropium had no effects on epithelial cell differentiation after air exposure. Differentiation into goblet cells was barely induced after air exposure. Therefore, IL-13 (1 ng/mL) was used to induce goblet cell metaplasia. IL-13 induced MUC5AC-positive cells (5-fold) and goblet cells (14-fold), as assessed by histochemistry, and MUC5AC gene expression (105-fold). These effects were partly prevented by tiotropium (47-92%). Goblet cell metaplasia was induced by IL-13 in a dose-dependent manner, which was inhibited by tiotropium. In addition, tiotropium reversed goblet cell metaplasia induced by 2 weeks of IL-13 exposure. IL-13 decreased forkhead box protein A2 (FoxA2) expression (1.6-fold) and increased FoxA3 (3.6-fold) and SAM-pointed domain-containing ETS transcription factor (SPDEF) (5.2-fold) expression. Tiotropium prevented the effects on FoxA2 and FoxA3, but not on SPDEF. CONCLUSIONS: We demonstrate that tiotropium has no effects on epithelial cell differentiation after air exposure, but inhibits and reverses IL-13-induced goblet cell metaplasia, possibly via FoxA2 and FoxA3. This indicates that non-neuronal acetylcholine contributes to goblet cell differentiation by a direct effect on epithelial cells.


Assuntos
Células Caliciformes/efeitos dos fármacos , Interleucina-13/antagonistas & inibidores , Mucosa Respiratória/efeitos dos fármacos , Derivados da Escopolamina/farmacologia , Acetilcolina/fisiologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Células Cultivadas , Antagonistas Colinérgicos/farmacologia , Relação Dose-Resposta a Droga , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células Caliciformes/patologia , Humanos , Interleucina-13/administração & dosagem , Interleucina-13/farmacologia , Metaplasia/induzido quimicamente , Metaplasia/genética , Metaplasia/patologia , Mucina-5AC/biossíntese , Mucina-5AC/genética , Mucosa Respiratória/patologia , Brometo de Tiotrópio , Fatores de Transcrição/metabolismo
12.
FASEB J ; 28(11): 4617-28, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25103224

RESUMO

Cigarette smoke (CS) induces inflammatory responses characterized by increase of immune cells and cytokine release. Remodeling processes, such as mucus hypersecretion and extracellular matrix protein production, are also directly or indirectly induced by CS. Recently, we showed that activation of the exchange protein directly activated by cAMP (Epac) attenuates CS extract-induced interleukin (IL)-8 release from cultured airway smooth muscle cells. Using an acute, short-term model of CS exposure, we now studied the role of Epac1, Epac2, and the Epac effector phospholipase-Cε (PLCε) in airway inflammation and remodeling in vivo. Compared to wild-type mice exposed to CS, the number of total inflammatory cells, macrophages, and neutrophils and total IL-6 release was lower in Epac2(-/-) mice, which was also the case for neutrophils and IL-6 in PLCε(-/-) mice. Taken together, Epac2, acting partly via PLCε, but not Epac1, enhances CS-induced airway inflammation in vivo. In total lung homogenates of Epac1(-/-) mice, MUC5AC and matrix remodeling parameters (transforming growth factor-ß1, collagen I, and fibronectin) were increased at baseline. Our findings suggest that Epac1 primarily is capable of inhibiting remodeling processes, whereas Epac2 primarily increases inflammatory processes in vivo.


Assuntos
Remodelação das Vias Aéreas/fisiologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fumaça/efeitos adversos , Remodelação das Vias Aéreas/genética , Animais , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Citocinas/metabolismo , Feminino , Fatores de Troca do Nucleotídeo Guanina/genética , Inflamação/metabolismo , Pulmão/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout
13.
Am J Respir Cell Mol Biol ; 50(4): 690-8, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24156289

RESUMO

Asthma is a chronic obstructive airway disease, characterized by inflammation and remodeling. Acetylcholine contributes to symptoms by inducing bronchoconstriction via the muscarinic M3 receptor. Recent evidence suggests that bronchoconstriction can regulate airway remodeling, and therefore implies a role for the muscarinic M3 receptor. The objective of this work was to study the contribution of the muscarinic M3 receptor to allergen-induced remodeling using muscarinic M3 receptor subtype-deficient (M3R(-/-)) mice. Wild-type (WT), M1R(-/-), and M2R(-/-) mice were used as controls. C57Bl/6 mice were sensitized and challenged with ovalbumin (twice weekly for 4 wk). Control animals were challenged with saline. Allergen exposure induced goblet cell metaplasia, airway smooth muscle thickening (1.7-fold), pulmonary vascular smooth muscle remodeling (1.5-fold), and deposition of collagen I (1.7-fold) and fibronectin (1.6-fold) in the airway wall of WT mice. These effects were absent or markedly lower in M3R(-/-) mice (30-100%), whereas M1R(-/-) and M2R(-/-) mice responded similarly to WT mice. In addition, airway smooth muscle and pulmonary vascular smooth muscle mass were 35-40% lower in saline-challenged M3R(-/-) mice compared with WT mice. Interestingly, allergen-induced airway inflammation, assessed as infiltrated eosinophils and T helper type 2 cytokine expression, was similar or even enhanced in M3R(-/-) mice. Our data indicate that acetylcholine contributes to allergen-induced remodeling and smooth muscle mass via the muscarinic M3 receptor, and not via M1 or M2 receptors. No stimulatory role for muscarinic M3 receptors in allergic inflammation was observed, suggesting that the role of acetylcholine in remodeling is independent of the allergic inflammatory response, and may involve bronchoconstriction.


Assuntos
Acetilcolina/metabolismo , Remodelação das Vias Aéreas , Alérgenos , Pulmão/metabolismo , Músculo Liso/metabolismo , Ovalbumina , Pneumonia/metabolismo , Receptor Muscarínico M3/metabolismo , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Eosinófilos/metabolismo , Matriz Extracelular/metabolismo , Feminino , Células Caliciformes/metabolismo , Células Caliciformes/patologia , Mediadores da Inflamação/metabolismo , Pulmão/patologia , Pulmão/fisiopatologia , Metaplasia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso/patologia , Músculo Liso/fisiopatologia , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Pneumonia/induzido quimicamente , Pneumonia/patologia , Pneumonia/fisiopatologia , Artéria Pulmonar/metabolismo , Artéria Pulmonar/patologia , Receptor Muscarínico M1/deficiência , Receptor Muscarínico M1/genética , Receptor Muscarínico M2/deficiência , Receptor Muscarínico M2/genética , Receptor Muscarínico M3/deficiência , Receptor Muscarínico M3/genética
14.
Am J Physiol Cell Physiol ; 306(6): C585-97, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24452374

RESUMO

Airway epithelium, which forms the first barrier towards environmental insults, is disturbed by cigarette smoking, a major risk factor for developing chronic obstructive pulmonary disease (COPD). A-kinase anchoring proteins (AKAP) maintain endothelial barrier function and coordinate subcellular localization of protein kinase A (PKA). However, the role of AKAPs in epithelial barrier function is unknown. We studied the role of AKAPs in regulating human bronchial epithelial (Hogg JC, Timens W. Annu Rev Pathol 4: 435-459, 2009; HBE) barrier. Cigarette smoke extract (CSE) reduced barrier function in 16HBE cells and the expression of the adhesion molecule E-cadherin specifically at the cell membrane. In addition, CSE reduced the protein expression of the AKAP family member AKAP9 at the cell membrane. The expression of AKAP5 and AKAP12 was unaffected by CSE. AKAP9 interacted and colocalized with E-cadherin at the cell membrane, suggesting that the reduction of both proteins may be related. Interestingly, disruption of AKAP-PKA interactions by st-Ht31 prevented the CSE-induced reduction of E-cadherin and AKAP9 protein expression and subsequent loss of barrier function. Silencing of AKAP9 reduced the functional epithelial barrier and prevented the ability of st-Ht31 to restore membrane localization of E-cadherin. Our data suggest the possibility of a specific role for AKAP9 in the maintenance of the epithelial barrier. E-cadherin, but not AKAP9, protein expression was reduced in lung tissue from COPD patients compared with controls. However, AKAP9 mRNA expression was decreased in primary bronchial epithelial cells from current smokers compared with non/ex-smokers. In conclusion, our results indicate that AKAP proteins, most likely AKAP9, maintain the bronchial epithelial barrier by regulating the E-cadherin expression at the cell membrane.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Caderinas/metabolismo , Proteínas do Citoesqueleto/metabolismo , Nicotiana/efeitos adversos , Mucosa Respiratória/metabolismo , Fumaça/efeitos adversos , Fumar/metabolismo , Proteínas de Ancoragem à Quinase A/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Brônquios/efeitos dos fármacos , Brônquios/metabolismo , Caderinas/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Proteínas do Citoesqueleto/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Antígenos de Histocompatibilidade Menor , Proteínas Proto-Oncogênicas/metabolismo , Doença Pulmonar Obstrutiva Crônica/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Mucosa Respiratória/efeitos dos fármacos
15.
J Pharmacol Exp Ther ; 348(2): 303-10, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24307202

RESUMO

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.


Assuntos
Agonistas de Receptores Adrenérgicos beta 2/uso terapêutico , Antialérgicos/uso terapêutico , Benzoxazinas/uso terapêutico , Brônquios/efeitos dos fármacos , Modelos Animais de Doenças , Hipersensibilidade Respiratória/tratamento farmacológico , Derivados da Escopolamina/uso terapêutico , Administração por Inalação , Agonistas de Receptores Adrenérgicos beta 2/administração & dosagem , Animais , Animais não Endogâmicos , Antialérgicos/administração & dosagem , Benzoxazinas/administração & dosagem , Brônquios/imunologia , Brônquios/metabolismo , Brônquios/patologia , Broncoconstrição/efeitos dos fármacos , Broncodilatadores/administração & dosagem , Broncodilatadores/uso terapêutico , Antagonistas Colinérgicos/administração & dosagem , Antagonistas Colinérgicos/uso terapêutico , Relação Dose-Resposta a Droga , Esquema de Medicação , Sinergismo Farmacológico , Quimioterapia Combinada , Cobaias , Histamina/administração & dosagem , Histamina/metabolismo , Masculino , Substâncias Protetoras/administração & dosagem , Substâncias Protetoras/uso terapêutico , Hipersensibilidade Respiratória/imunologia , Hipersensibilidade Respiratória/metabolismo , Hipersensibilidade Respiratória/patologia , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia , Derivados da Escopolamina/administração & dosagem , Brometo de Tiotrópio
17.
Am J Respir Cell Mol Biol ; 49(1): 18-27, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23449734

RESUMO

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.


Assuntos
Proliferação de Células , Miócitos de Músculo Liso/efeitos dos fármacos , Receptor Cross-Talk , Receptor Muscarínico M2/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Linhagem Celular Transformada , Meios de Cultura Livres de Soro , Replicação do DNA , Sinergismo Farmacológico , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Trietiodeto de Galamina/farmacologia , Humanos , Integrina alfa5beta1/metabolismo , Cloreto de Metacolina/farmacologia , Mitógenos/farmacologia , Miócitos de Músculo Liso/metabolismo , Oligopeptídeos/farmacologia , Toxina Pertussis/farmacologia , Receptor Muscarínico M2/agonistas , Sistema Respiratório/citologia , Fatores de Tempo
18.
Eur Respir J ; 42(6): 1677-88, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23397297

RESUMO

Cholinergic tone contributes to airflow obstruction in chronic obstructive pulmonary disease. Accordingly, anticholinergics are effective bronchodilators by blocking the muscarinic M3 receptor on airway smooth muscle. Recent evidence indicates that acetylcholine also contributes to airway inflammation. However, which muscarinic receptor subtype(s) regulates this process is unknown. In this study, the contribution of the M1, M2 and M3 receptor subtypes to cigarette smoke-induced airway inflammation was investigated by exposing muscarinic receptor subtype deficient mice to cigarette smoke for 4 days. In wild-type mice, cigarette smoke induced an increase in macrophages, neutrophils and lymphocytes in bronchoalveolar lavage fluid. Neutrophilic inflammation was higher in M1(-/-) and M2(-/-) mice compared to wild-type mice, but lower in M3(-/-) mice. Accordingly, the release of keratinocyte-derived chemokine (KC), monocyte chemotactic protein-1 and interleukin-6 was higher in M1(-/-) and M2(-/-) mice, and reduced in M3(-/-) mice. Markers of remodelling were not increased after cigarette smoke exposure. However, M3(-/-) mice had reduced expression of transforming growth factor-ß1 and matrix proteins. Cigarette smoke-induced inflammatory cell recruitment and KC release were also prevented by the M3-receptor selective antagonist 1-dimethyl-4-diphenylacetoxypiperidinium iodide (4-DAMP) in wild-type mice. Collectively, our data indicate a pro-inflammatory role for the M3 receptor in cigarette smoke-induced neutrophilia and cytokine release, yet an anti-inflammatory role for M1 and M2 receptors.


Assuntos
Inflamação/induzido quimicamente , Inflamação/patologia , Receptor Muscarínico M1/metabolismo , Receptor Muscarínico M2/metabolismo , Receptor Muscarínico M3/metabolismo , Fumar/efeitos adversos , Acetilcolina/química , Animais , Líquido da Lavagem Broncoalveolar , Antagonistas Colinérgicos/química , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
19.
J Pharmacol Exp Ther ; 346(1): 86-95, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23591997

RESUMO

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.


Assuntos
Remodelação das Vias Aéreas , Asma/metabolismo , Colágeno Tipo I/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Sistema de Sinalização das MAP Quinases , Contração Muscular , Músculo Liso/metabolismo , Remodelação das Vias Aéreas/efeitos dos fármacos , Animais , Asma/tratamento farmacológico , Bovinos , Adesão Celular , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Colágeno Tipo I/antagonistas & inibidores , Ativação Enzimática , Proteína-Tirosina Quinases de Adesão Focal/antagonistas & inibidores , Proteína-Tirosina Quinases de Adesão Focal/química , Proteína-Tirosina Quinases de Adesão Focal/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Terapia de Alvo Molecular , Contração Muscular/efeitos dos fármacos , Músculo Liso/citologia , Músculo Liso/efeitos dos fármacos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Fosforilação , Domínios e Motivos de Interação entre Proteínas , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional , Proteínas Tirosina Quinases/biossíntese , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Técnicas de Cultura de Tecidos , Traqueia/citologia , Traqueia/metabolismo
20.
Respir Res ; 14: 113, 2013 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-24152196

RESUMO

BACKGROUND: Glycogen synthase kinase-3 (GSK-3) is a constitutively active kinase that regulates multiple signalling proteins and transcription factors involved in a myriad of cellular processes. The kinase acts as a negative regulator in ß-catenin signalling and is critically involved in the smad pathway. Activation of both pathways may contribute to pulmonary features of chronic obstructive pulmonary disease (COPD). METHODS: In the present study, we investigated the effect of the selective GSK-3 inhibitor SB216763 on pulmonary pathology in a guinea pig model of lipopolysaccharide (LPS)-induced COPD. Guinea pigs were instilled intranasally with LPS or saline twice weekly for 12 weeks and pre-treated with either intranasally instilled SB216763 or corresponding vehicle 30 min prior to each LPS/saline challenge. RESULTS: Repeated LPS exposures activated ß-catenin signalling, primarily in the airway epithelium and submucosa. LPS also induced pulmonary inflammation and tissue remodelling as indicated by inflammatory cell influx, increased pulmonary fibronectin expression and enhanced small airway collagen content. Inhibition of GSK-3 by SB216763 did not affect LPS-induced inflammatory cell influx, but prevented the small airway remodelling and, unexpectedly, inhibited the activation of ß-catenin in vivo. LPS or SB216763 treatment had no effect on the airway smooth muscle content and alveolar airspace size. However, GSK-3 inhibition prevented LPS-induced right ventricle hypertrophy. CONCLUSIONS: Our findings indicate that GSK-3 inhibition prevents LPS-induced pulmonary pathology in guinea pigs, and that locally reduced LPS-induced ß-catenin activation appears in part to underlie this effect.


Assuntos
Inibidores Enzimáticos/uso terapêutico , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Indóis/uso terapêutico , Pulmão/patologia , Maleimidas/uso terapêutico , Doença Pulmonar Obstrutiva Crônica/patologia , Doença Pulmonar Obstrutiva Crônica/prevenção & controle , Animais , Colágeno/metabolismo , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Fibronectinas/metabolismo , Quinase 3 da Glicogênio Sintase/efeitos dos fármacos , Cobaias , Indóis/farmacologia , Lipopolissacarídeos/efeitos adversos , Lipopolissacarídeos/farmacologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Masculino , Maleimidas/farmacologia , Músculo Liso/efeitos dos fármacos , Músculo Liso/metabolismo , Músculo Liso/patologia , Doença Pulmonar Obstrutiva Crônica/induzido quimicamente , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteínas Smad/metabolismo , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA