Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mikrochim Acta ; 191(5): 285, 2024 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-38652174

RESUMEN

One significant constraint in the advancement of biosensors is the signal-to-noise ratio, which is adversely affected by the presence of interfering factors such as blood in the sample matrix. In the present investigation, a specific aptamer binding was chosen for its affinity, while exhibiting no binding affinity towards non-target bacterial cells. This selective binding property was leveraged to facilitate the production of magnetic microparticles decorated with aptamers. A novel assay was developed to effectively isolate S. pneumoniae from PBS or directly from blood samples using an aptamer with an affinity constant of 72.8 nM. The capture experiments demonstrated efficiencies up to 87% and 66% are achievable for isolating spiked S. pneumoniae in 1 mL PBS and blood samples, respectively.


Asunto(s)
Aptámeros de Nucleótidos , Dióxido de Silicio , Aptámeros de Nucleótidos/química , Dióxido de Silicio/química , Streptococcus pneumoniae/aislamiento & purificación , Streptococcus pneumoniae/química , Humanos , Técnicas Biosensibles/métodos , Nanopartículas de Magnetita/química
2.
Int J Mol Sci ; 25(11)2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38892358

RESUMEN

Obese patients with asthma present with aggravated symptoms that are also harder to treat. Here, we used a mouse model of allergic asthma sensitised and challenged to house dust mite (HDM) extracts to determine whether high-fat-diet consumption would exacerbate the key features of allergic airway inflammation. C57BL/6 mice were intranasally sensitised and challenged with HDM extracts over a duration of 3 weeks. The impact of high-fat-diet (HFD) vs. normal diet (ND) chow was studied on HDM-induced lung inflammation and inflammatory cell infiltration as well as cytokine production. HFD-fed mice had greater inflammatory cell infiltration around airways and blood vessels, and an overall more severe degree of inflammation than in the ND-fed mice (semiquantitative blinded evaluation). Quantitative assessment of HDM-associated Th2 responses (numbers of lung CD4+ T cells, eosinophils, serum levels of allergen-specific IgE as well as the expression of Th2 cytokines (Il5 and Il13)) did not show significant changes between the HFD and ND groups. Interestingly, the HFD group exhibited a more pronounced neutrophilic infiltration within their lung tissues and an increase in non-Th2 cytokines (Il17, Tnfa, Tgf-b, Il-1b). These findings provide additional evidence that obesity triggered by a high-fat-diet regimen may exacerbate asthma by involving non-Th2 and neutrophilic pathways.


Asunto(s)
Asma , Citocinas , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Obesidad , Células Th2 , Animales , Asma/inmunología , Asma/etiología , Asma/patología , Asma/metabolismo , Obesidad/inmunología , Obesidad/metabolismo , Ratones , Dieta Alta en Grasa/efectos adversos , Células Th2/inmunología , Células Th2/metabolismo , Citocinas/metabolismo , Pyroglyphidae/inmunología , Pulmón/patología , Pulmón/inmunología , Pulmón/metabolismo , Inflamación/patología , Inflamación/inmunología , Inflamación/metabolismo , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Femenino , Alérgenos/inmunología
3.
Adv Exp Med Biol ; 1303: 1-12, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33788184

RESUMEN

The mechanisms driving corticosteroid insensitivity in asthma are still unclear although evidence points toward a potential role of lung mast cells. Indeed, a number of in vitro studies using various cell types showed that different mediators produced by activated mast cells, including cytokines, have the capacity to interfere with the therapeutic action of corticosteroids. In patients with severe allergic refractory asthma, the anti-IgE monoclonal antibody (mAb), Omalizumab, has been shown to be associated with a marked reduction in inhaled and systemic use of corticosteroids, further suggesting a key role of mast cells in the poor response of patients to these drugs. The present chapter will discuss the possible underlying mechanisms by which mast cells could contribute to reducing corticosteroid sensitivity seen in patients with severe asthma.


Asunto(s)
Asma , Mastocitos , Corticoesteroides/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Asma/tratamiento farmacológico , Humanos
4.
Proc Natl Acad Sci U S A ; 113(16): 4524-9, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27071102

RESUMEN

G protein-coupled receptors (GPCRs) are known to initiate a plethora of signaling pathways in vitro. However, it is unclear which of these pathways are engaged to mediate physiological responses. Here, we examine the distinct roles of Gq/11-dependent signaling and receptor phosphorylation-dependent signaling in bronchial airway contraction and lung function regulated through the M3-muscarinic acetylcholine receptor (M3-mAChR). By using a genetically engineered mouse expressing a G protein-biased M3-mAChR mutant, we reveal the first evidence, to our knowledge, of a role for M3-mAChR phosphorylation in bronchial smooth muscle contraction in health and in a disease state with relevance to human asthma. Furthermore, this mouse model can be used to distinguish the physiological responses that are regulated by M3-mAChR phosphorylation (which include control of lung function) from those responses that are downstream of G protein signaling. In this way, we present an approach by which to predict the physiological/therapeutic outcome of M3-mAChR-biased ligands with important implications for drug discovery.


Asunto(s)
Bronquios/metabolismo , Músculo Liso/metabolismo , Receptor Muscarínico M3/metabolismo , Transducción de Señal/fisiología , Animales , Bronquios/citología , Humanos , Ratones , Ratones Noqueados , Músculo Liso/citología , Fosforilación/fisiología , Receptor Muscarínico M3/genética
5.
J Immunol ; 196(1): 55-63, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26608913

RESUMEN

Human lung mast cells (HLMCs) play a central role in asthma pathogenesis through their relocation to the airway smooth muscle (ASM) bundles. ß2 adrenoceptor (ß2-AR)-agonists are used to relieve bronchoconstriction in asthma, but may reduce asthma control, particularly when used as monotherapy. We hypothesized that HLMC and human ASM cell (HASMC) responsiveness to ß2-AR agonists would be attenuated when HLMCs are in contact with HASMCs. Cells were cultured in the presence of the short-acting ß2-agonist albuterol, and the long-acting ß2-agonists formoterol and olodaterol. Constitutive and FcεRI-dependent HLMC histamine release, HASMC contraction, and ß2-AR phosphorylation at Tyr(350) were assessed. Constitutive HLMC histamine release was increased in HLMC-HASMC coculture and this was enhanced by ß2-AR agonists. Inhibition of FcεRI-dependent HLMC mediator release by ß2-agonists was greatly reduced in HLMC-HASMC coculture. These effects were reversed by neutralization of stem cell factor (SCF) or cell adhesion molecule 1 (CADM1). ß2-AR agonists did not prevent HASMC contraction when HLMCs were present, but this was reversed by fluticasone. ß2-AR phosphorylation at Tyr(350) occurred within 5 min in both HLMCs and HASMCs when the cells were cocultured, and was inhibited by neutralizing SCF or CADM1. HLMC interactions with HASMCs via CADM1 and Kit inhibit the potentially beneficial effects of ß2-AR agonists on these cells via phosphorylation of the ß2-AR. These results may explain the potentially adverse effects of ß2-ARs agonists when used for asthma therapy. Targeting SCF and CADM1 may enhance ß2-AR efficacy, particularly in corticosteroid-resistant patients.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 2/uso terapéutico , Asma/inmunología , Pulmón/inmunología , Mastocitos/inmunología , Músculo Liso/inmunología , Receptores Adrenérgicos beta 2/metabolismo , Albuterol/farmacología , Asma/tratamiento farmacológico , Asma/patología , Benzoxazinas/farmacología , Molécula 1 de Adhesión Celular , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Fluticasona/farmacología , Fumarato de Formoterol/farmacología , Histamina/metabolismo , Liberación de Histamina/inmunología , Humanos , Inmunoglobulinas/metabolismo , Pulmón/citología , Miocitos del Músculo Liso/metabolismo , Fosforilación , Receptores de IgE/inmunología , Factor de Células Madre/metabolismo
7.
J Immunol ; 195(6): 2852-60, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26276873

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a common, progressive, and invariably lethal interstitial lung disease with no effective therapy. The key cell driving the development of fibrosis is the myofibroblast. Lipoxin A4 (LXA4) is an anti-inflammatory lipid, important in the resolution of inflammation, and it has potential antifibrotic activity. However, the effects of LXA4 on primary human lung myofibroblasts (HLMFs) have not previously been investigated. Therefore, the aim of this study was to examine the effects of LXA4 on TGF-ß1-dependent responses in IPF- and nonfibrotic control (NFC)-derived HLMFs. HLMFs were isolated from IPF and NFC patients and grown in vitro. The effects of LXA4 on HLMF proliferation, collagen secretion, α-smooth muscle actin (αSMA) expression, and Smad2/3 activation were examined constitutively and following TGF-ß1 stimulation. The LXA4 receptor (ALXR) was expressed in both NFC- and IPF-derived HLMFs. LXA4 (10(-10) and 10(-8) mol) reduced constitutive αSMA expression, actin stress fiber formation, contraction, and nuclear Smad2/3, indicating regression from a myofibroblast to fibroblast phenotype. LXA4 also significantly inhibited FBS-dependent proliferation and TGF-ß1-dependent collagen secretion, αSMA expression, and Smad2/3 nuclear translocation in IPF-derived HLMFs. LXA4 did not inhibit Smad2/3 phosphorylation. In summary, LXA4 attenuated profibrotic HLMF activity and promoted HLMF regression to a quiescent fibroblast phenotype. LXA4 or its stable analogs delivered by aerosol may offer a novel approach to the treatment of IPF.


Asunto(s)
Fibrosis Pulmonar Idiopática/patología , Lipoxinas/farmacología , Miofibroblastos/metabolismo , Receptores de Formil Péptido/biosíntesis , Receptores de Lipoxina/biosíntesis , Factor de Crecimiento Transformador beta1/farmacología , Actinas/biosíntesis , Proliferación Celular , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Activación Enzimática/efectos de los fármacos , Humanos , Fibrosis Pulmonar Idiopática/inmunología , Inflamación/inmunología , Inflamación/patología , Pulmón/citología , Pulmón/patología , Fosforilación/efectos de los fármacos , ARN Mensajero/biosíntesis , Proteína Smad2/metabolismo , Proteína smad3/metabolismo
8.
J Allergy Clin Immunol ; 135(2): 395-406, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25312757

RESUMEN

BACKGROUND: The D prostanoid receptor 2 (DP2; also known as chemoattractant receptor-homologous molecule expressed on TH2 cells) is implicated in the pathogenesis of asthma, but its expression within bronchial biopsy specimens is unknown. OBJECTIVES: We sought to investigate the bronchial submucosal DP2 expression in asthmatic patients and healthy control subjects and to explore its functional role in epithelial cells. METHODS: DP2 protein expression was assessed in bronchial biopsy specimens from asthmatic patients (n = 22) and healthy control subjects (n = 10) by using immunohistochemistry and in primary epithelial cells by using flow cytometry, immunofluorescence, and quantitative RT-PCR. The effects of the selective DP2 agonist 13, 14-dihydro-15-keto prostaglandin D2 on epithelial cell migration and differentiation were determined. RESULTS: Numbers of submucosal DP2(+) cells were increased in asthmatic patients compared with those in healthy control subjects (mean [SEM]: 78 [5] vs 22 [3]/mm(2) submucosa, P < .001). The bronchial epithelium expressed DP2, but its expression was decreased in asthmatic patients compared with that seen in healthy control subjects (mean [SEM]: 21 [3] vs 72 [11]/10 mm(2) epithelial area, P = .001), with similar differences observed in vitro by primary epithelial cells. Squamous metaplasia of the bronchial epithelium was increased in asthmatic patients and related to decreased DP2 expression (rs = 0.69, P < .001). 13, 14-Dihydro-15-keto prostaglandin D2 promoted epithelial cell migration and at air-liquid interface cultures increased the number of MUC5AC(+) and involucrin-positive cells, which were blocked with the DP2-selective antagonist AZD6430. CONCLUSIONS: DP2 is expressed by the bronchial epithelium, and its activation drives epithelial differentiation, suggesting that in addition to its well-characterized role in inflammatory cell migration, DP2 might contribute to airway remodeling in asthmatic patients.


Asunto(s)
Asma/metabolismo , Bronquios/metabolismo , Proteínas de Unión al ADN/metabolismo , Mucosa Respiratoria/metabolismo , Factores de Transcripción/metabolismo , Adulto , Remodelación de las Vías Aéreas (Respiratorias)/genética , Asma/diagnóstico , Asma/genética , Asma/inmunología , Biopsia , Bronquios/inmunología , Bronquios/patología , Calcio/metabolismo , Estudios de Casos y Controles , Diferenciación Celular/genética , Movimiento Celular , Células Cultivadas , Proteínas de Unión al ADN/genética , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Pruebas de Función Respiratoria , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/patología , Factores de Riesgo , Índice de Severidad de la Enfermedad , Factores de Transcripción/genética
9.
Am J Respir Cell Mol Biol ; 53(5): 728-37, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25897650

RESUMEN

Preclinical models of human conditions including asthma showed the therapeutic potential of Compound A (CpdA), a dissociated glucocorticoid (GC) receptor (GRα) ligand. Whether CpdA inhibits GC resistance, a central feature of severe asthma, has not been addressed. We investigated whether CpdA modulates cytokine-induced GC resistance in human airway smooth muscle (ASM) cells. Healthy and asthmatic ASM cells were treated with TNF-α/IFN-γ for 24 hours in the presence or absence of CpdA. ELISA and quantitative PCR assays were used to assess the effect of CpdA on chemokine expression. Activation of GRα by CpdA was assessed by quantitative PCR, immunostaining, and receptor antagonism using RU486. An effect of CpdA on the transcription factor interferon regulatory factor 1 (IRF-1) was investigated using immunoblot, immunostaining, and small interfering RNA (siRNA) knockdown. CpdA inhibited production of fluticasone-resistant chemokines CCL5, CX3CL1, and CXCL10 at protein and mRNA levels in both asthmatic and healthy cells. CpdA failed to induce expression of GC-induced Leucine Zipper while transiently inducing mitogen-activated protein kinase phosphatase 1 (MKP-1) at both mRNA and protein levels. CpdA inhibitory action was not associated with GRα nuclear translocation, nor was it prevented by RU486 antagonism. Activation of IRF-1 by TNF-α/IFN-γ was inhibited by CpdA. IRF-1 siRNA knockdown reduced cytokine-induced CCL5 and CX3CL1 production. siRNA MKP-1 prevented the inhibitory effect of CpdA on cytokine-induced CXCL10 production. For the first time, we show that CpdA inhibits the production of GC-resistant chemokines via GRα-independent mechanisms involving the inhibition of IRF-1 and up-regulation of MKP-1. Thus, targeting CpdA-sensitive pathways in ASM cells represents an alternative therapeutic approach to treat GC resistance in asthma.


Asunto(s)
Acetatos/farmacología , Resistencia a Medicamentos/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Mucosa Respiratoria/efectos de los fármacos , Tiramina/análogos & derivados , Adulto , Antiasmáticos/farmacología , Asma/tratamiento farmacológico , Asma/genética , Asma/inmunología , Asma/patología , Estudios de Casos y Controles , Quimiocina CCL5/genética , Quimiocina CCL5/inmunología , Quimiocina CX3CL1/genética , Quimiocina CX3CL1/inmunología , Quimiocina CXCL10/genética , Quimiocina CXCL10/inmunología , Fosfatasa 1 de Especificidad Dual/genética , Fosfatasa 1 de Especificidad Dual/inmunología , Células Epiteliales/inmunología , Células Epiteliales/patología , Femenino , Fluticasona/farmacología , Expresión Génica/inmunología , Humanos , Factor 1 Regulador del Interferón/antagonistas & inhibidores , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/inmunología , Interferón gamma/farmacología , Masculino , Persona de Mediana Edad , Mifepristona/farmacología , Cultivo Primario de Células , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/inmunología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/patología , Factor de Necrosis Tumoral alfa/farmacología , Tiramina/farmacología
10.
J Cell Physiol ; 230(6): 1189-98, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25204635

RESUMEN

Asthma in the pediatric population remains a significant contributor to morbidity and increasing healthcare costs. Vitamin D3 insufficiency and deficiency have been associated with development of asthma. Recent studies in models of adult airway diseases suggest that the bioactive Vitamin D3 metabolite, calcitriol (1,25-dihydroxyvitamin D3 ; 1,25(OH)2 D3 ), modulates responses to inflammation; however, this concept has not been explored in developing airways in the context of pediatric asthma. We used human fetal airway smooth muscle (ASM) cells as a model of the early postnatal airway to explore how calcitriol modulates remodeling induced by pro-inflammatory cytokines. Cells were pre-treated with calcitriol and then exposed to TNFα or TGFß for up to 72 h. Matrix metalloproteinase (MMP) activity, production of extracellular matrix (ECM), and cell proliferation were assessed. Calcitriol attenuated TNFα enhancement of MMP-9 expression and activity. Additionally, calcitriol attenuated TNFα and TGFß-induced collagen III expression and deposition, and separately, inhibited proliferation of fetal ASM cells induced by either inflammatory mediator. Analysis of signaling pathways suggested that calcitriol effects in fetal ASM involve ERK signaling, but not other major inflammatory pathways. Overall, our data demonstrate that calcitriol can blunt multiple effects of TNFα and TGFß in developing airway, and point to a potentially novel approach to alleviating structural changes in inflammatory airway diseases of childhood.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Asma/metabolismo , Calcitriol/deficiencia , Citocinas/metabolismo , Miocitos del Músculo Liso/metabolismo , Deficiencia de Vitamina D/metabolismo , Proliferación Celular/efectos de los fármacos , Matriz Extracelular/metabolismo , Humanos , Músculo Liso/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
11.
Am J Physiol Lung Cell Mol Physiol ; 309(4): L348-59, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26092996

RESUMEN

Although the majority of patients with asthma are well controlled by inhaled glucocorticoids (GCs), patients with severe asthma are poorly responsive to GCs. This latter group is responsible for a disproportionate share of health care costs associated with asthma. Recent studies in immune cells have incriminated interferon-γ (IFN-γ) as a possible trigger of GC insensitivity in severe asthma; however, little is known about the role of IFN-γ in modulating GC effects in other clinically relevant nonimmune cells, such as airway epithelial cells. We hypothesized that IFN-γ-induced JAK/STAT-associated signaling pathways in airway epithelial cells are insensitive to GCs and that strategies aimed at inhibiting JAK/STAT pathways can restore steroid responsiveness. Using Western blot analysis we found that all steps of the IFN-γ-induced JAK/STAT signaling pathway were indeed GC insensitive. Transfection of cells with reporter plasmid showed IFN-γ-induced STAT1-dependent gene transcription to be also GC insensitive. Interestingly, real-time PCR analysis showed that IFN-γ-inducible genes (IIGs) were differentially affected by GC, with CXCL10 being GC sensitive and CXCL11 and IFIT2 being GC insensitive. Further investigation showed that the differential sensitivity of IIGs to GC was due to their variable dependency to JAK/STAT vs. NF-κB signaling pathways with GC-sensitive IIGs being more NF-κB dependent and GC-insensitive IIGs being more JAK/STAT dependent. Importantly, transfection of cells with siRNA-STAT1 was able to restore steroid responsiveness of GC-insensitive IIGs. Taken together, our results show the insensitivity of IFN-γ-induced JAK/STAT signaling pathways to GC effects in epithelial cells and also suggest that targeting STAT1 could restore GC responsiveness in patients with severe asthma.


Asunto(s)
Androstadienos/farmacología , Células Epiteliales/metabolismo , Glucocorticoides/farmacología , Interferón gamma/fisiología , Factor de Transcripción STAT1/metabolismo , Transporte Activo de Núcleo Celular , Anciano , Asma/tratamiento farmacológico , Asma/metabolismo , Asma/patología , Línea Celular Tumoral , Células Epiteliales/efectos de los fármacos , Femenino , Fluticasona , Humanos , Quinasas Janus/metabolismo , Masculino , Persona de Mediana Edad , FN-kappa B/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Mucosa Respiratoria/patología , Transducción de Señal , Transcripción Genética
12.
Pediatr Res ; 78(6): 650-6, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26331770

RESUMEN

BACKGROUND: Adult human airway smooth muscle (ASM) produce cytokines involved in recruitment and survival of leukocytes within airway walls. Cytokine generation by adult ASM is glucocorticoid-sensitive. Whether developing lung ASM produces cytokines in a glucocorticoid-sensitive fashion is unknown. METHODS: Cultured fetal human ASM cells stimulated with TNF-α (0-20 ng/ml) were incubated with TNF-α receptor-blocking antibodies, fluticasone (1 and 100 nm), or vehicle. Supernatants and cells were assayed for the production of CCL5, CXCL10, and CXCL8 mRNA and protein and glucocorticoid receptor phosphorylation. RESULTS: CCL5, CXCL10, and CXCL8 mRNA and protein production by fetal ASM cell was significantly and dose-dependently following TNF-α treatment. Cytokine mRNA and protein production were effectively blocked by TNF-α R1 and R2 receptor neutralizing antibodies but variably inhibited by fluticasone. TNF-α-induced TNF-R1 and R2 receptor mRNA expression was only partially attenuated by fluticasone. Glucocorticoid receptor phosphorylation at serine (Ser) 211 but not at Ser 226 was enhanced by fluticasone. CONCLUSION: Production of CCL5, CXCL10, and CXCL8 by fetal ASM appears to involve pathways that are both qualitatively and mechanistically distinct to those described for adult ASM. The findings imply developing ASM has potential to recruit leukocyte into airways and, therefore, of relevance to childhood airway diseases.


Asunto(s)
Quimiotaxis de Leucocito/efectos de los fármacos , Citocinas/metabolismo , Fluticasona/farmacología , Glucocorticoides/farmacología , Pulmón/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Anticuerpos/farmacología , Células Cultivadas , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Citocinas/inmunología , Relación Dosis-Respuesta a Droga , Edad Gestacional , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Pulmón/embriología , Pulmón/inmunología , Pulmón/metabolismo , Miocitos del Músculo Liso/inmunología , Miocitos del Músculo Liso/metabolismo , Fosforilación , Receptores de Glucocorticoides/agonistas , Receptores de Glucocorticoides/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/efectos de los fármacos , Receptores Tipo I de Factores de Necrosis Tumoral/inmunología , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/efectos de los fármacos , Receptores Tipo II del Factor de Necrosis Tumoral/inmunología , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Serina , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología
13.
J Immunol ; 191(5): 2624-2636, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23904164

RESUMEN

Identifying the factors responsible for relative glucocorticosteroid (GC) resistance present in patients with severe asthma and finding tools to reverse it are of paramount importance. In asthma we see in vivo evidence of GC-resistant pathways in airway smooth muscle (ASM) bundles that can be modeled in vitro by exposing cultured ASM cells to TNF-α/IFN-γ. This action drives GC insensitivity via protein phosphatase 5-dependent impairment of GC receptor phosphorylation. In this study, we investigated whether KCa3.1 ion channels modulate the activity of GC-resistant pathways using our ASM model of GC insensitivity. Immunohistochemical staining of endobronchial biopsies revealed that KCa3.1 channels are localized to the plasma membrane and nucleus of ASM in both healthy controls and asthmatic patients, irrespective of disease severity. Western blot assays and immunofluorescence staining confirmed the nuclear localization of KCa3.1 channels in ASM cells. The functional importance of KCa3.1 channels in the regulation of GC-resistant chemokines induced by TNF-α/IFN-γ was assessed using complementary inhibitory strategies, including KCa3.1 blockers (TRAM-34 and ICA-17043) or KCa3.1-specific small hairpin RNA delivered by adenoviruses. KCa3.1 channel blockade led to a significant reduction of fluticasone-resistant CX3CL1, CCL5, and CCL11 gene and protein expression. KCa3.1 channel blockade also restored fluticasone-induced GC receptor-α phosphorylation at Ser(211) and transactivation properties via the suppression of cytokine-induced protein phosphatase 5 expression. The effect of KCa3.1 blockade was evident in ASM cells from both healthy controls and asthmatic subjects. In summary, KCa3.1 channels contribute to the regulation of GC-resistant inflammatory pathways in ASM cells: blocking KCa3.1 channels may enhance corticosteroid activity in severe asthma.


Asunto(s)
Asma/metabolismo , Bronquios/metabolismo , Glucocorticoides , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Miocitos del Músculo Liso/metabolismo , Adulto , Asma/tratamiento farmacológico , Western Blotting , Células Cultivadas , Resistencia a Medicamentos/fisiología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Masculino , Técnicas de Placa-Clamp , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Am J Respir Cell Mol Biol ; 50(2): 301-15, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24024586

RESUMEN

Like many steroid receptors, the glucocorticoid (GC) receptor (GR) is a phosphoprotein. Although there are multiple phosphorylation sites critical for GR transcriptional activity (i.e., serine [S]203, S211, and S226), their respective role in driving GR functions is highly cell specific. We have recently identified protein phosphatase 5 as an essential Ser/Thr phosphatase responsible for impairing GR function via S211 dephosphorylation in airway smooth muscle (ASM) cells. Because p38 mitogen-activated protein kinase (MAPK) directly phosphorylates GR in different cell types in a stimulus- and cell-dependent manner, we investigated the role of p38 MAPK on GR phosphorylation and function in ASM cells. Cells were transfected with 100 nM p38 MAPK small interfering RNA or 2 µg MAPK kinase 3 expression vector (a specific kinase that directly activates p38 MAPK) in the presence or absence of fluticasone (100 nM) and/or p38 MAPK pharmacological inhibitor SB203580. We found that p38 MAPK blockade positively regulates GR nuclear translocation and GR-dependent induction of the steroid-target gene GC-induced leucine zipper in a hormone-independent manner. We also found that p38 MAPK-dependent regulation of GR functions was associated with a differential action on GR phosphorylation at S203 and S211 residues. This study demonstrated that the inactive state of GR in resting conditions is not only ensured by the absence of the GC ligand but also by p38 MAPK-dependent phosphorylation of unliganded GR at specific residues, which appears to be important in determining the overall GC responsiveness of ASM cells.


Asunto(s)
Glucocorticoides/farmacología , Miocitos del Músculo Liso/metabolismo , Receptores de Glucocorticoides/metabolismo , Sistema Respiratorio/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Células Cultivadas , Humanos , Imidazoles/farmacología , Leucina Zippers/fisiología , Miocitos del Músculo Liso/efectos de los fármacos , Fosforilación/efectos de los fármacos , Piridinas/farmacología , Sistema Respiratorio/efectos de los fármacos , Transducción de Señal/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
15.
Respir Res ; 15: 155, 2014 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-25476248

RESUMEN

BACKGROUND: Idiopathic pulmonary fibrosis is a common and invariably fatal disease with limited therapeutic options. Ca2+-activated KCa3.1 potassium channels play a key role in promoting TGFß1 and bFGF-dependent profibrotic responses in human lung myofibroblasts (HLMFs). We hypothesised that KCa3.1 channel-dependent cell processes regulate HLMF αSMA expression via Smad2/3 signalling pathways. METHODS: In this study we have compared the phenotype of HLMFs derived from non-fibrotic healthy control lungs (NFC) with cells derived from IPF lungs. HLMFs grown in vitro were examined for αSMA expression by immunofluorescence (IF), RT-PCR and flow cytommetry. Basal Smad2/3 signalling was examined by RT-PCR, western blot and immunofluorescence. Two specific and distinct KCa3.1 blockers (TRAM-34 200 nM and ICA-17043 [Senicapoc] 100 nM) were used to determine their effects on HLMF differentiation and the Smad2/3 signalling pathways. RESULTS: IPF-derived HLMFs demonstrated increased constitutive expression of both α-smooth muscle actin (αSMA) and actin stress fibres, indicative of greater myofibroblast differentiation. This was associated with increased constitutive Smad2/3 mRNA and protein expression, and increased Smad2/3 nuclear localisation. The increased Smad2/3 nuclear localisation was inhibited by removing extracellular Ca2+ or blocking KCa3.1 ion channels with selective KCa3.1 blockers (TRAM-34, ICA-17043). This was accompanied by de-differentiation of IPF-derived HLMFs towards a quiescent fibroblast phenotype as demonstrated by reduced αSMA expression and reduced actin stress fibre formation. CONCLUSIONS: Taken together, these data suggest that Ca2+- and KCa3.1-dependent processes facilitate "constitutive" Smad2/3 signalling in IPF-derived fibroblasts, and thus promote fibroblast to myofibroblast differentiation. Importantly, inhibiting KCa3.1 channels reverses this process. Targeting KCa3.1 may therefore provide a novel and effective approach for the treatment of IPF and there is the potential for the rapid translation of KCa3.1-directed therapy to the clinic.


Asunto(s)
Actinas/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Pulmón/metabolismo , Miofibroblastos/metabolismo , Proteína Smad2/metabolismo , Proteína Smad4/metabolismo , Actinas/genética , Estudios de Casos y Controles , Diferenciación Celular , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/patología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/antagonistas & inhibidores , Pulmón/efectos de los fármacos , Pulmón/patología , Miofibroblastos/efectos de los fármacos , Miofibroblastos/patología , Fenotipo , Bloqueadores de los Canales de Potasio/farmacología , Transducción de Señal , Proteína Smad2/genética , Proteína Smad4/genética
16.
Cells ; 13(10)2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38786103

RESUMEN

Cigarette smoke is one of the main factors in Chronic Obstructive Pulmonary Disease (COPD), a respiratory syndrome marked by persistent respiratory symptoms and increasing airway obstruction. Perturbed NAD+/NADH levels may play a role in various diseases, including lung disorders like COPD. In our study, we investigated the preventive effect of NADH supplementation in an experimental model of COPD induced by cigarette smoke extract (CSE). N = 64 mice randomly distributed in eight groups were injected with NADH (two doses of 100 mg/kg or 200 mg/kg) or dexamethasone (2 mg/kg) before being exposed to CSE for up to 9 weeks. Additionally, NADH supplementation preserved lung antioxidant defenses by preventing the functional loss of key enzymes such as superoxide dismutase (SOD), glutathione peroxidase (GPX), catalase, and the expression levels of glutathione (GSH) (n = 4, p < 0.001). It also reduced oxidative damage markers, such as malondialdehyde (MDA) and nitrites (n = 4, p < 0.001). A marked increase in tissue myeloperoxidase activity was assessed (MPO), confirming neutrophils implication in the inflammatory process. The latter was significantly ameliorated in the NADH-treated groups (p < 0.001). Finally, NADH prevented the CSE-induced secretion of cytokines such as Tumor Necrosis Factor alpha (TNF-α), IL-17, and IFN-y (n = 4, p < 0.001). Our study shows, for the first time, the clinical potential of NADH supplementation in preventing key features of COPD via its unique anti-inflammatory and antioxidant properties.


Asunto(s)
Modelos Animales de Enfermedad , Ratones Endogámicos BALB C , NAD , Neumonía , Enfermedad Pulmonar Obstructiva Crónica , Animales , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/patología , Enfermedad Pulmonar Obstructiva Crónica/prevención & control , Enfermedad Pulmonar Obstructiva Crónica/etiología , NAD/metabolismo , Ratones , Neumonía/prevención & control , Neumonía/metabolismo , Neumonía/patología , Inyecciones Intraperitoneales , Humo/efectos adversos , Estrés Oxidativo/efectos de los fármacos , Masculino , Antioxidantes/metabolismo , Antioxidantes/farmacología , Citocinas/metabolismo , Pulmón/patología , Pulmón/metabolismo , Pulmón/efectos de los fármacos , Peroxidasa/metabolismo
17.
Rheumatology (Oxford) ; 52(7): 1190-201, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23436580

RESUMEN

OBJECTIVES: Activation of TLR7 and TLR9 and high serum levels of BAFF have been implicated in the pathogenesis of SLE. However, little is known about the effects of TLR9 activation on BAFF expression by human B cells. We investigated the effect of the TLR9 agonist, CpG-ODN 2006, on the expression of BAFF and its receptors BAFF-R, TACI and BCMA, in isolated B cells from healthy donors. METHODS: We used RT-PCR, flow cytometry and ELISA to investigate the expression of BAFF, and flow cytometry for BAFF-R, TACI and BCMA. Functional assays assessed the responses of resting and CpG-ODN-activated B cells to exogenous soluble and membrane-bound BAFF. RESULTS: CpG-ODN did not induce BAFF secretion, but increased expression of membrane-bound BAFF on B cells. CpG-ODN also induced the expression of TACI and BCMA, but did not up-regulate BAFF-R expression. In functional studies, CpG-ODN sensitized human B cells to proliferate in response to exogenous BAFF. This effect was inhibited by a blocking antibody against BAFF-R, but was not inhibited by anti-TACI or anti-BCMA antibodies. Membrane-bound BAFF, induced by CpG-ODN, co-stimulated the proliferation of B cells stimulated with anti-IgM in a manner that was dependent on the expression of surface BAFF on the CpG-ODN-treated B cells. CONCLUSION: TLR9 activation induces expression of membrane-bound BAFF on human B cells and leads to increased proliferation in response to both soluble and membrane-bound BAFF. These data extend our understanding of the role of TLR9 activation on human B cells and provide insights into the mechanisms by which TLR9 may participate in the pathogenesis of SLE.


Asunto(s)
Factor Activador de Células B/metabolismo , Receptor del Factor Activador de Células B/metabolismo , Linfocitos B/citología , Oligodesoxirribonucleótidos/farmacología , Receptor Toll-Like 9/metabolismo , Adulto , Factor Activador de Células B/genética , Receptor del Factor Activador de Células B/genética , Antígeno de Maduración de Linfocitos B/genética , Antígeno de Maduración de Linfocitos B/metabolismo , Linfocitos B/metabolismo , Proliferación Celular , Células Cultivadas , Cartilla de ADN/química , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Expresión Génica , Humanos , Activación de Linfocitos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptor Toll-Like 9/agonistas , Proteína Activadora Transmembrana y Interactiva del CAML/genética , Proteína Activadora Transmembrana y Interactiva del CAML/metabolismo
18.
Am J Respir Cell Mol Biol ; 47(4): 464-73, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22592921

RESUMEN

Corticosteroid insensitivity (CSI) represents a profound challenge in managing patients with asthma. We recently demonstrated that short exposure of airway smooth muscle cells (ASMCs) to proasthmatic cytokines drastically reduced their responsiveness to glucocorticoids (GCs), an effect that was partially mediated via interferon regulatory factor-1, suggesting the involvement of additional mechanisms (Am J Respir Cell Mol Biol 2008;38:463-472). Although GC receptor (GR) can be phosphorylated at multiple serines in the N-terminal region, the major phosphorylation sites critical for GR transcriptional activity are serines 211 (Ser211) and 226 (Ser226). We tested the novel hypothesis that cytokine-induced CSI in ASMCs is due to an impaired GR phosphorylation. Cells were treated with TNF-α (10 ng/ml) and IFN-γ (500 UI/ml) for 6 hours and/or fluticasone (100 nm) added 2 hours before. GR was constitutively phosphorylated at Ser226 but not at Ser211 residues. Cytokines dramatically suppressed fluticasone-induced phosphorylation of GR on Ser211 but not on Ser226 residues while increasing the expression of Ser/Thr protein phosphatase (PP)5 but not that of PP1 or PP2A. Transfection studies using a reporter construct containing GC responsive elements showed that the specific small interfering RNA-induced mRNA knockdown of PP5, but not that of PP1 or PP2A, partially prevented the cytokine suppressive effects on GR-meditated transactivation activity. Similarly, cytokines failed to inhibit GC-induced GR-Ser211 phosphorylation when expression of PP5 was suppressed. We propose that the novel mechanism that proasthmatic cytokine-induced CSI in ASMCs is due, in part, to PP5-mediated impairment of GR-Ser211 phosphorylation.


Asunto(s)
Citocinas/fisiología , Miocitos del Músculo Liso/enzimología , Proteínas Nucleares/fisiología , Fosfoproteínas Fosfatasas/fisiología , Procesamiento Proteico-Postraduccional , Receptores de Glucocorticoides/metabolismo , Sistema Respiratorio/citología , Androstadienos/farmacología , Células Cultivadas , Fluticasona , Técnicas de Silenciamiento del Gen , Glucocorticoides/farmacología , Glucocorticoides/fisiología , Humanos , Mutación Missense , Miocitos del Músculo Liso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación , Interferencia de ARN , Receptores de Glucocorticoides/genética
19.
Am J Physiol Lung Cell Mol Physiol ; 303(8): L711-9, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22923637

RESUMEN

Maintenance of blood oxygen saturation dictates supplemental oxygen administration to premature infants, but hyperoxia predisposes survivors to respiratory diseases such as asthma. Although much research has focused on oxygen effects on alveoli in the setting of bronchopulmonary dysplasia, the mechanisms by which oxygen affects airway structure or function relevant to asthma are still under investigation. We used isolated human fetal airway smooth muscle (fASM) cells from 18-20 postconceptual age lungs (canalicular stage) to examine oxygen effects on intracellular Ca(2+) ([Ca(2+)](i)) and cellular proliferation. fASM cells expressed substantial smooth muscle actin and myosin and several Ca(2+) regulatory proteins but not fibroblast or epithelial markers, profiles qualitatively comparable to adult human ASM. Fluorescence Ca(2+) imaging showed robust [Ca(2+)](i) responses to 1 µM acetylcholine (ACh) and 10 µM histamine (albeit smaller and slower than adult ASM), partly sensitive to zero extracellular Ca(2+). Compared with adult, fASM showed greater baseline proliferation. Based on this validation, we assessed fASM responses to 10% hypoxia through 90% hyperoxia and found enhanced proliferation at <60% oxygen but increased apoptosis at >60%, effects accompanied by appropriate changes in proliferative vs. apoptotic markers and enhanced mitochondrial fission at >60% oxygen. [Ca(2+)](i) responses to ACh were enhanced for <60% but blunted at >60% oxygen. These results suggest that hyperoxia has dose-dependent effects on structure and function of developing ASM, which could have consequences for airway diseases of childhood. Thus detrimental effects on ASM should be an additional consideration in assessing risks of supplemental oxygen in prematurity.


Asunto(s)
Hiperoxia/metabolismo , Hipoxia/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Oxígeno/efectos adversos , Tráquea/metabolismo , Adulto , Asma/epidemiología , Asma/metabolismo , Asma/patología , Calcio/metabolismo , Proliferación Celular , Células Cultivadas , Feto/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Hiperoxia/epidemiología , Hiperoxia/patología , Hipoxia/epidemiología , Hipoxia/patología , Recién Nacido , Recien Nacido Prematuro , Mitocondrias/metabolismo , Miocitos del Músculo Liso/citología , Oxígeno/administración & dosificación , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Factores de Riesgo , Tráquea/citología , Tráquea/embriología
20.
BMC Complement Altern Med ; 12: 47, 2012 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-22520446

RESUMEN

BACKGROUND: Flaxseed (FS), a nutritional supplement consisting mainly of omega-3 fatty acids and lignan phenolics has potent anti-inflammatory, anti-fibrotic and antioxidant properties. The usefulness of flaxseed as an alternative and complimentary treatment option has been known since ancient times. We have shown that dietary FS supplementation ameliorates oxidative stress and inflammation in experimental models of acute and chronic lung injury in mice resulting from diverse toxicants. The development of lung tissue damage in response to direct or indirect oxidant stress is a complex process, associated with changes in expression levels of a number of genes. We therefore postulated that flaxseed might modulate gene expression of vital signaling pathways, thus interfering with the development of tissue injury. METHODS: We evaluated gene expression in lungs of flaxseed-fed (10%FS) mice under unchallenged, control conditions. We reasoned that array technology would provide a powerful tool for studying the mechanisms behind this response and aid the evaluation of dietary flaxseed intervention with a focus on toxicologically relevant molecular gene targets. Gene expression levels in lung tissues were analyzed using a large-scale array whereby 28,800 genes were evaluated. RESULTS: 3,713 genes (12.8%) were significantly (p < 0.05) differentially expressed, of which 2,088 had a >1.5-fold change. Genes affected by FS include those in protective pathways such as Phase I and Phase II. CONCLUSIONS: The array studies have provided information on how FS modulates gene expression in lung and how they might be related to protective mechanisms. In addition, our study has confirmed that flaxseed is a nutritional supplement with potentially useful therapeutic applications in complementary and alternative (CAM) medicine especially in relation to treatment of lung disease.


Asunto(s)
Antiinflamatorios/farmacología , Antioxidantes/farmacología , Lino/química , Enfermedades Pulmonares , Pulmón/efectos de los fármacos , Preparaciones de Plantas/farmacología , Transcriptoma , Animales , Antiinflamatorios/uso terapéutico , Antioxidantes/uso terapéutico , Suplementos Dietéticos , Modelos Animales de Enfermedad , Ácidos Grasos Omega-3/farmacología , Ácidos Grasos Omega-3/uso terapéutico , Perfilación de la Expresión Génica/métodos , Lignanos/farmacología , Lignanos/uso terapéutico , Pulmón/metabolismo , Enfermedades Pulmonares/tratamiento farmacológico , Enfermedades Pulmonares/genética , Enfermedades Pulmonares/metabolismo , Ratones , Fenoles/farmacología , Fenoles/uso terapéutico , Fitoterapia , Preparaciones de Plantas/uso terapéutico , Semillas/química , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA