Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Clin Sci (Lond) ; 126(8): 567-80, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24152048

RESUMEN

Viral respiratory tract infections are known triggers of asthma exacerbations in both adults and children. The current standard of care, inhaled CS (corticosteroids) and LABAs (long-acting ß2-adrenoceptor agonists), fails to prevent the loss of control that manifests as an exacerbation. In order to better understand the mechanisms underlying viral asthma exacerbations we established an in vivo model using the clinically relevant aeroallergen HDM (house dust mite) and the viral mimetic/TLR3 (Toll-like receptor 3) agonist poly(I:C). Poly(I:C) alone induced a similar neutrophilic inflammatory profile in the BAL (bronchoalveolar lavage) to that of HRV1b (human rhinovirus 1b) alone, accompanied by both elevated BAL KC (keratinocyte-derived chemokine) and IL-1ß (interleukin-1ß). When mice allergic to HDM were also challenged with poly(I:C) the neutrophilic inflammatory profile was exacerbated. Increased CD8(+) T-cell numbers, increased CD4(+) and CD8(+) cell activation and elevated KC and IL-1ß were observed. No increases in Th2 cytokines or the eosinophil chemoattractant CCL11 [chemokine (C-C motif) ligand 11], above those induced by HDM alone, were observed. The poly(I:C)-exacerbated neutrophilia did not translate into changes in AHR (airways hyper-responsiveness), indicating that in this model inflammation and AHR are two mechanistically independent events. To test the clinical relevance of this model CS sensitivity was assessed using prednisone, a synthetic oral CS used to manage exacerbations in asthmatic patients already on maximal doses of inhaled CS. The increased neutrophils, and accompanying cytokines/chemokines KC and IL-1ß induced by poly(I:C) challenge of HDM-sensitized and challenged mice were insensitive to oral prednisone therapy. In summary we have described a CS-resistant mouse model mimicking the key aspects of viral asthma exacerbation using the clinically relevant aeroallergen HDM and the viral mimic poly(I:C). This model may provide better understanding of disease mechanisms underlying viral exacerbations and could be used to build early confidence in novel therapeutic axes targeting viral asthma exacerbations in Th2 asthmatics.


Asunto(s)
Asma/inmunología , Modelos Animales de Enfermedad , Infecciones por Picornaviridae/inmunología , Rhinovirus/inmunología , Animales , Asma/tratamiento farmacológico , Asma/virología , Hiperreactividad Bronquial/inmunología , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Quimiocinas/inmunología , Femenino , Glucocorticoides/uso terapéutico , Células HeLa , Interacciones Huésped-Patógeno/inmunología , Humanos , Interleucina-1beta/inmunología , Ratones , Ratones Endogámicos BALB C , Neutrófilos/inmunología , Infecciones por Picornaviridae/virología , Neumonía/inmunología , Poli I-C/inmunología , Prednisona/uso terapéutico , Pyroglyphidae/inmunología , Rhinovirus/fisiología , Receptor Toll-Like 3/inmunología
2.
PLoS Pathog ; 6(11): e1001178, 2010 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-21079690

RESUMEN

The relative roles of the endosomal TLR3/7/8 versus the intracellular RNA helicases RIG-I and MDA5 in viral infection is much debated. We investigated the roles of each pattern recognition receptor in rhinovirus infection using primary bronchial epithelial cells. TLR3 was constitutively expressed; however, RIG-I and MDA5 were inducible by 8-12 h following rhinovirus infection. Bronchial epithelial tissue from normal volunteers challenged with rhinovirus in vivo exhibited low levels of RIG-I and MDA5 that were increased at day 4 post infection. Inhibition of TLR3, RIG-I and MDA5 by siRNA reduced innate cytokine mRNA, and increased rhinovirus replication. Inhibition of TLR3 and TRIF using siRNA reduced rhinovirus induced RNA helicases. Furthermore, IFNAR1 deficient mice exhibited RIG-I and MDA5 induction early during RV1B infection in an interferon independent manner. Hence anti-viral defense within bronchial epithelium requires co-ordinated recognition of rhinovirus infection, initially via TLR3/TRIF and later via inducible RNA helicases.


Asunto(s)
Bronquios/metabolismo , ARN Helicasas DEAD-box/metabolismo , Epitelio/metabolismo , Infecciones por Picornaviridae/metabolismo , Rhinovirus/patogenicidad , Receptor Toll-Like 3/metabolismo , Animales , Western Blotting , Bronquios/inmunología , Bronquios/virología , Células Cultivadas , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/antagonistas & inhibidores , ARN Helicasas DEAD-box/genética , Epitelio/inmunología , Epitelio/virología , Femenino , Técnica del Anticuerpo Fluorescente , Células HeLa , Humanos , Inmunidad Innata , Helicasa Inducida por Interferón IFIH1 , Ratones , Ratones Noqueados , Infecciones por Picornaviridae/inmunología , Infecciones por Picornaviridae/virología , ARN Bicatenario , ARN Mensajero/genética , ARN Interferente Pequeño/genética , ARN Viral/genética , Receptor de Interferón alfa y beta/fisiología , Receptores Inmunológicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptor Toll-Like 3/antagonistas & inhibidores , Receptor Toll-Like 3/genética
3.
J Biol Chem ; 285(38): 29101-10, 2010 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-20833730

RESUMEN

Asthmatic airway smooth muscle (ASM) expresses interferon-γ-inducible protein-10 (CXCL10), a chemokine known to mediate mast cell migration into ASM bundles that has been reported in the airways of asthmatic patients. CXCL10 is elevated in patients suffering from viral exacerbations of asthma and in patients with chronic obstructive pulmonary disease (COPD), diseases in which corticosteroids are largely ineffective. IFNγ and TNFα synergistically induce CXCL10 release from human ASM cells in a steroid-insensitive manner, via an as yet undefined mechanism. We report that TNFα activates the classical NF-κB (nuclear factor κB) pathway, whereas IFNγ activates JAK2/STAT-1α and that inhibition of the JAK/STAT pathway is more effective in abrogating CXCL10 release than the steroid fluticasone. The synergy observed with TNFα and IFNγ together, however, did not lie at the level of NF-κB activation, STAT-1α phosphorylation, or in vivo binding of these transcription factors to the CXCL10 promoter. Stimulation of human ASM cells with TNFα and IFNγ induced histone H4 but not histone H3 acetylation at the CXCL10 promoter, although no synergism was observed when both cytokines were combined. We show, however, that TNFα and IFNγ exert a synergistic effect on the recruitment of CREB-binding protein (CBP) to the CXCL10, which is accompanied by increased RNA polymerase II. Our results provide evidence that synergism between TNFα and IFNγ lies at the level of coactivator recruitment in human ASM and suggest that inhibition of JAK/STAT signaling may be of therapeutic benefit in steroid-resistant airway disease.


Asunto(s)
Proteína de Unión a CREB/metabolismo , Quimiocina CXCL10/metabolismo , Interferón gamma/farmacología , Miocitos del Músculo Liso/metabolismo , FN-kappa B/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Western Blotting , Proteína de Unión a CREB/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Cultivadas , Quimiocina CXCL10/genética , Inmunoprecipitación de Cromatina , Humanos , Miocitos del Músculo Liso/efectos de los fármacos , FN-kappa B/genética , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , ARN Polimerasa II/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT1/genética
4.
Thorax ; 66(12): 1029-35, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21606476

RESUMEN

BACKGROUND: Asthma and chronic obstructive pulmonary disease are airway inflammatory diseases characterised by airflow obstruction. Currently approved bronchodilators such as long-acting ß(2) adrenoceptor agonists are the mainstay treatments but often fail to relieve symptoms of chronic obstructive pulmonary disease and severe asthma and safety concerns have been raised over long-term use. The aim of the study was to identify the receptor involved in prostaglandin E(2) (PGE(2))-induced relaxation in guinea pig, murine, monkey, rat and human airways in vitro. METHODS: Using an extensive range of pharmacological tools, the relaxant potential of PGE(2) and selective agonists for the EP(1-4) receptors in the presence and absence of selective antagonists in guinea pig, murine, monkey, rat and human isolated airways was investigated. RESULTS: In agreement with previous studies, it was found that the EP(2) receptor mediates PGE(2)-induced relaxation of guinea pig, murine and monkey trachea and that the EP(4) receptor mediates PGE(2)-induced relaxation of the rat trachea. These data have been confirmed in murine airways from EP(2) receptor-deficient mice (Ptger2). In contrast to previous publications, a role for the EP(4) receptor in relaxant responses in human airways in vitro was found. Relaxant activity of AH13205 (EP(2) agonist) was also demonstrated in guinea pig but not human airway tissue, which may explain its failure in clinical studies. CONCLUSION: Identification of the receptor mediating PGE(2)-induced relaxation represents a key step in developing a novel bronchodilator therapy. These data explain the lack of bronchodilator activity observed with selective EP(2) receptor agonists in clinical studies.


Asunto(s)
Asma/tratamiento farmacológico , Broncodilatadores/farmacología , Dinoprostona/farmacología , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Receptores de Prostaglandina E/agonistas , Tráquea/efectos de los fármacos , Alprostadil/análogos & derivados , Alprostadil/farmacología , Animales , Asma/fisiopatología , Compuestos Bicíclicos Heterocíclicos con Puentes , Dinoprostona/análogos & derivados , Ácidos Grasos Insaturados , Cobayas , Humanos , Hidrazinas/farmacología , Macaca fascicularis , Éteres Metílicos/farmacología , Ratones , Ratones Endogámicos C57BL , Naftalenos/farmacología , Fenilbutiratos/farmacología , Ácidos Prostanoicos/farmacología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Ratas , Ratas Sprague-Dawley , Receptores de Prostaglandina E/fisiología , Análisis de Regresión , Especificidad de la Especie , Tráquea/fisiología , Xantonas/farmacología
5.
Front Med (Lausanne) ; 8: 711194, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34422866

RESUMEN

Introduction: Pulmonary fibrosis includes a spectrum of diseases and is incurable. There is a variation in disease course, but it is often progressive leading to increased breathlessness, impaired quality of life, and decreased life expectancy. Detection of pulmonary fibrosis is challenging, which contributes to considerable delays in diagnosis and treatment. More knowledge about the diagnostic journey from patients' perspective is needed to improve the diagnostic pathway. The aims of this study were to evaluate the time to diagnosis of pulmonary fibrosis, identify potential reasons for delays, and document patients emotions. Methods: Members of European patient organisations, with a self-reported diagnosis of pulmonary fibrosis, were invited to participate in an online survey. The survey assessed the diagnostic pathway retrospectively, focusing on four stages: (1) time from initial symptoms to first appointment in primary care; (2) time to hospital referral; (3) time to first hospital appointment; (4) time to final diagnosis. It comprised open-ended and closed questions focusing on time to diagnosis, factors contributing to delays, diagnostic tests, patient emotions, and information provision. Results: Two hundred and seventy three participants (214 idiopathic pulmonary fibrosis, 28 sarcoidosis, 31 other) from 13 countries responded. Forty percent of individuals took ≥1 year to receive a final diagnosis. Greatest delays were reported in stage 1, with only 50.2% making an appointment within 3 months. For stage 2, 73.3% reported a hospital referral within three primary care visits. However, 9.9% reported six or more visits. After referral, 76.9% of patients were assessed by a specialist within 3 months (stage 3) and 62.6% received a final diagnosis within 3 months of their first hospital visit (stage 4). Emotions during the journey were overall negative. A major need for more information and support during and after the diagnostic process was identified. Conclusion: The time to diagnose pulmonary fibrosis varies widely across Europe. Delays occur at each stage of the diagnostic pathway. Raising awareness about pulmonary fibrosis amongst the general population and healthcare workers is essential to shorten the time to diagnosis. Furthermore, there remains a need to provide patients with sufficient information and support at all stages of their diagnostic journey.

6.
J Immunol ; 181(5): 3503-14, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18714023

RESUMEN

The transcription factor NF-kappaB plays a pivotal role in regulating inflammatory gene expression. Its effects are optimized by various coactivators, including histone acetyltransferases (HATs) such as CREB-binding protein/p300 and p300/CBP-associated factor (p/CAF). The molecular mechanisms regulating cofactor recruitment are poorly understood. In this study, we describe a novel role for protein kinase C (PKC) betaIotaIota in augmenting NF-kappaB-mediated TNF-alpha-induced transcription of the target gene CCL11 in human airway smooth muscle cells by phosphorylating the HAT p/CAF. Studies using reporters, overexpression strategies, kinase-dead and HAT-defective mutants, and chromatin immunoprecipitation showed that PKCbetaII activation was not involved in NF-kappaB translocation, but facilitated NF-kappaB-mediated CCL11 transcription by colocalizing with and phosphorylating p/CAF, and thereby acetylating histone H4 and promoting p65 association with the CCL11 promoter. The effect was dependent on p/CAF's HAT activity. Furthermore, mouse embryonic fibroblasts from PKCbeta knockout mice showed markedly reduced TNF-alpha-induced CCL11 expression and NF-kappaB reporter activity that was restored on PKCbetaII overexpression, suggesting a critical role for this pathway. These data suggest a novel important biological role for PKCbetaIotaIota in NF-kappaB-mediated CCL11 transcription by p/CAF activation and histone H4 acetylation.


Asunto(s)
Quimiocina CCL11/genética , Proteína p300 Asociada a E1A/metabolismo , Histonas/metabolismo , FN-kappa B/fisiología , Regiones Promotoras Genéticas , Proteína Quinasa C/fisiología , Factor de Transcripción ReIA/fisiología , Transcripción Genética , Acetilación , Adulto , Animales , Células Cultivadas , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Miocitos del Músculo Liso , Proteína Quinasa C beta , Transporte de Proteínas , Sistema Respiratorio , Factor de Necrosis Tumoral alfa
7.
J Immunol ; 181(6): 4265-71, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18768884

RESUMEN

The liver X receptors (LXRalpha/beta) are orphan nuclear receptors that are expressed in a large number of cell types and have been shown to have anti-inflammatory properties. Nuclear receptors have previously proved to be amenable targets for small molecular mass pharmacological agents in asthma, and so the effect of an LXR ligand was assessed in models of allergic airway inflammation. LXR agonist, GW 3965, was profiled in rat and mouse models of allergic asthma. In the Brown Norway rats, GW 3965 (3-30 mg/kg) was unable to reduce the bronchoalveolar lavage eosinophilia associated with this model and had no impact on inflammatory biomarkers (eotaxin and IL-1beta). The compound did significantly stimulate ABCA-1 (ATP-binding cassette A1) mRNA expression, indicating that there was adequate exposure/LXR activation. In the mouse model, the LXR ligand surprisingly increased airway reactivity, an effect that was apparent in both the Ag and nonchallenged groups. This increase was not associated with a change in lung tissue inflammation or number of mucus-containing cells. There was, however, a marked increase in airway smooth muscle thickness in both treated groups. We demonstrated an increase in contractile response to exogenous methacholine in isolated airways taken from LXR agonist-treated animals compared with the relevant control tissue. We corroborated these findings in a human system by demonstrating increased proliferation of cultured airway smooth muscle. This phenomenon, if evidenced in man, would indicate that LXR ligands may directly increase airway reactivity, which could be detrimental, especially in patients with existing respiratory disease and with already compromised lung function.


Asunto(s)
Asma/inmunología , Asma/metabolismo , Benzoatos/administración & dosificación , Bencilaminas/administración & dosificación , Hiperreactividad Bronquial/metabolismo , Proteínas de Unión al ADN/agonistas , Músculo Liso/crecimiento & desarrollo , Músculo Liso/metabolismo , Receptores Citoplasmáticos y Nucleares/agonistas , Regulación hacia Arriba/inmunología , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Asma/patología , Asma/fisiopatología , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/fisiopatología , Proliferación Celular/efectos de los fármacos , Proteínas de Unión al ADN/fisiología , Relación Dosis-Respuesta Inmunológica , Humanos , Receptores X del Hígado , Masculino , Ratones , Ratones Endogámicos BALB C , Músculo Liso/inmunología , Receptores Nucleares Huérfanos , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Ratas , Ratas Endogámicas BN , Receptores Citoplasmáticos y Nucleares/fisiología , Regulación hacia Arriba/efectos de los fármacos
8.
Pulm Pharmacol Ther ; 22(5): 426-35, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19114116

RESUMEN

Compelling evidence identifies airway smooth muscle (ASM) not only as a target but also a cellular source for a diverse range of mediators underlying the processes of airway narrowing and airway hyperresponsiveness in diseases such as asthma. These include the growing family of plasma membrane phospholipid-derived polyunsaturated fatty acids broadly characterised by the prostaglandins, leukotrienes, lipoxins, isoprostanes and lysophospholipids. In this review, we describe the enzymatic and non-enzymatic biosynthetic pathways of these lipid mediators and how these are influenced by drug treatment, oxidative stress and airways disease. Additionally, we outline their cognate receptors, many of which are expressed by ASM. We describe potential deleterious and protective roles for these lipid mediators in airway inflammatory and remodelling processes by describing their effects on diverse functions of ASM in asthma that have the potential to contribute to asthma pathogenesis and symptoms. These functions include contractile tone development, cytokine and extracellular matrix production, and cellular proliferation and migration.


Asunto(s)
Bronquios/fisiología , Eicosanoides/fisiología , Lisofosfolípidos/fisiología , Músculo Liso/fisiología , Sistema Respiratorio/metabolismo , Animales , Eicosanoides/biosíntesis , Humanos , Lisofosfolípidos/biosíntesis , Modelos Químicos , Contracción Muscular/fisiología , Músculo Liso/metabolismo
9.
Sci Rep ; 9(1): 6020, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30988425

RESUMEN

Phenotypic changes in lung fibroblasts are believed to contribute to the development of Idiopathic Pulmonary Fibrosis (IPF), a progressive and fatal lung disease. Long intergenic non-coding RNAs (lincRNAs) have been identified as novel regulators of gene expression and protein activity. In non-stimulated cells, we observed reduced proliferation and inflammation but no difference in the fibrotic response of IPF fibroblasts. These functional changes in non-stimulated cells were associated with changes in the expression of the histone marks, H3K4me1, H3K4me3 and H3K27ac indicating a possible involvement of epigenetics. Following activation with TGF-ß1 and IL-1ß, we demonstrated an increased fibrotic but reduced inflammatory response in IPF fibroblasts. There was no significant difference in proliferation following PDGF exposure. The lincRNAs, LINC00960 and LINC01140 were upregulated in IPF fibroblasts. Knockdown studies showed that LINC00960 and LINC01140 were positive regulators of proliferation in both control and IPF fibroblasts but had no effect upon the fibrotic response. Knockdown of LINC01140 but not LINC00960 increased the inflammatory response, which was greater in IPF compared to control fibroblasts. Overall, these studies demonstrate for the first time that lincRNAs are important regulators of proliferation and inflammation in human lung fibroblasts and that these might mediate the reduced inflammatory response observed in IPF-derived fibroblasts.


Asunto(s)
Fibroblastos/patología , Fibrosis Pulmonar Idiopática/genética , Pulmón/patología , ARN Largo no Codificante/genética , Células Cultivadas , Epigénesis Genética , Femenino , Fibroblastos/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/patología , Pulmón/metabolismo , Masculino , Persona de Mediana Edad , Transcriptoma
10.
Front Immunol ; 9: 2906, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30619270

RESUMEN

There is accumulating evidence to indicate that long non-coding RNAs (lncRNAs) are important regulators of the inflammatory response. In this report, we have employed next generation sequencing to identify 14 lncRNAs that are differentially expressed in human lung fibroblasts following the induction of inflammation using interleukin-1ß (IL-1ß). Knockdown of the two most highly expressed lncRNAs, IL7AS, and MIR3142HG, showed that IL7AS negatively regulated IL-6 release whilst MIR3142HG was a positive regulator of IL-8 and CCL2 release. Parallel studies in fibroblasts derived from patients with idiopathic pulmonary fibrosis showed similar increases in IL7AS levels, that also negatively regulate IL-6 release. In contrast, IL-1ß-induced MIR3142HG expression, and its metabolism to miR-146a, was reduced by 4- and 9-fold in IPF fibroblasts, respectively. This correlated with a reduced expression of inflammatory mediators whilst MIR3142HG knockdown showed no effect upon IL-8 and CCL2 release. Pharmacological studies showed that IL-1ß-induced IL7AS and MIR3142HG production and release of IL-6, IL-8, and CCL2 in both control and IPF fibroblasts were mediated via an NF-κB-mediated pathway. In summary, we have cataloged those lncRNAs that are differentially expressed following IL-1ß-activation of human lung fibroblasts, shown that IL7AS and MIR3142HG regulate the inflammatory response and demonstrated that the reduced inflammatory response in IPF fibroblast is correlated with attenuated expression of MIR3142HG/miR-146a.


Asunto(s)
Fibroblastos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Fibrosis Pulmonar Idiopática/genética , Inflamación/genética , Interleucina-1beta/farmacología , ARN Largo no Codificante/genética , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Fibrosis Pulmonar Idiopática/patología , Inflamación/metabolismo , Inflamación/patología , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Masculino , Persona de Mediana Edad
11.
Pharmacol Ther ; 169: 35-46, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27612548

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a chronic fibrosing interstitial pneumonia of unknown cause, characterised by progressive worsening in lung function and dyspnoea with an associated prognosis similar to or worse than many cancers. As a better understanding emerges around the pathogenesis and mechanisms driving disease pathology, a host of novel agents are being tested both pre-clinically and clinically. However even with this deeper understanding and positive pre-clinical supportive data, negative trial outcomes are frequently reported, highlighting the problems faced in treating such a heterogeneous disease with a varied clinical course. Recently, two therapies that slow disease progression, nintedanib and pirfenidone, have been approved for the treatment of IPF, yet the clinical unmet need is still high for IPF patients given their failure to stop disease progression and their potential side-effect profiles. Efforts are being made to not only understand the underlying pathways and genetics that might influence the clinical course of the disease, but also the non-invasive biomarkers that reflect the activity of specific pathways which in turn may highlight progressive treatment plans for individual patients. The cumulative data may be based on the identification of subgroups of patients via biomarker analysis of ongoing clinical trials, or investigation of cohorts of patients over time to understand the relative role of these mediators in their disease progression. Below we review the ongoing quest for novel therapeutic approaches and highlight, where appropriate attempts have been made to identify patients for which a specific pathway or mediator may be driving disease progression.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Medicina de Precisión/métodos , Animales , Antiinflamatorios no Esteroideos/efectos adversos , Biomarcadores/metabolismo , Progresión de la Enfermedad , Humanos , Fibrosis Pulmonar Idiopática/fisiopatología , Indoles/efectos adversos , Indoles/uso terapéutico , Pronóstico , Piridonas/efectos adversos , Piridonas/uso terapéutico
12.
Br J Pharmacol ; 141(4): 600-9, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14744812

RESUMEN

1. In the present study, we examined the effect of E-ring 8-isoprostanes on cholinergic neurotransmission in guinea-pig trachea and identified the receptor(s) involved. As isoprostanes are isomeric with prostaglandins, PGE(2) and sulprostone (a selective EP(3)-receptor agonist) were examined in parallel. 2. 8-Iso-PGE(1), 8-iso-PGE(2) (0.1 nm-1 microM), sulprostone (1 nm-1 microM) and PGE(2) (1 microM) suppressed EFS-evoked [(3)H]ACh release from guinea-pig trachea in a concentration-dependent manner, producing 39.5, 53.9, 61.2 and 59.9% inhibition, respectively, at 1 microM. It should be noted that an established maximum effective concentration was not determined. 3. Neither SQ 29,548 (1 microm; a TP-receptor antagonist) nor AH 6809 (10 microM; an EP(1)-/EP(2)-/DP-receptor antagonist) reversed the inhibitory effect of these compounds. 4. L-798,106, a novel and highly selective EP(3)-receptor antagonist, produced a parallel shift to the right of the concentration-response curves that described the inhibitory action of sulprostone on EFS-evoked contractile responses in guinea-pig vas deferens (an established EP(3)-receptor-expressing tissue), from which a mean pA(2) of 7.48 was derived. On guinea-pig trachea, L-798,106 also antagonised sulprostone-induced inhibition of EFS-induced twitch responses, with similar potency (mean pA(2)=7.82). 5. The inhibitory effects of 8-iso-PGE(1), 8-iso-PGE(2), sulprostone and PGE(2) on EFS-induced [(3)H]ACh release was blocked by L-798,106 at a concentration (10 microM) that binds only weakly to human recombinant EP(1)-, EP(2)- and EP(4)-receptor subtypes expressed in HEK 293 cells. 6. These data suggest that E-ring 8-isoprostanes, PGE(2) and sulprostone inhibit EFS-evoked [(3)H]ACh release from cholinergic nerves innervating guinea-pig trachea, by interacting with prejunctional prostanoid receptors of the EP(3)-subtype.


Asunto(s)
Acetilcolina/metabolismo , Dinoprostona/análogos & derivados , Isoprostanos/farmacología , Sistema Nervioso Parasimpático/metabolismo , Receptores de Prostaglandina E/agonistas , Tráquea/inervación , Tráquea/metabolismo , Animales , Dinoprostona/farmacología , Estimulación Eléctrica , Cobayas , Técnicas In Vitro , Masculino , Contracción Muscular/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Sistema Nervioso Parasimpático/efectos de los fármacos , Prostaglandinas E Sintéticas/farmacología , Subtipo EP3 de Receptores de Prostaglandina E , Transmisión Sináptica/efectos de los fármacos , Tráquea/efectos de los fármacos , Conducto Deferente/efectos de los fármacos , Conducto Deferente/inervación , Xantonas/farmacología
13.
Br J Pharmacol ; 141(7): 1141-50, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15023863

RESUMEN

1. The prostanoid receptor(s) on human airways smooth muscle (HASM) cells that mediates the inhibitory effect of PGE(2) on interleukin (IL)-1 beta-induced granulocyte/macrophage colony-stimulating factor (GM-CSF) release has been classified. 2. IL-1 beta evoked the release of GM-CSF from HASM cells, which was suppressed by PGE(2), 16,16-dimethyl PGE(2) (nonselective), misoprostol (EP(2)/EP(3)-selective), ONO-AE1-259 and butaprost (both EP(2)-selective) with pIC(50) values of 8.61, 7.13, 5.64, 8.79 and 5.43, respectively. EP-receptor agonists that have selectivity for the EP(1)-(17-phenyl-omega-trinor PGE(2)) and EP(3)-receptor (sulprostone) subtypes as well as cicaprost (IP-selective), PGD(2), PGF(2 alpha) and U-46619 (TP-selective) were poorly active or inactive at concentrations up to 10 microM. 3. AH 6809, a drug that can be used to selectively block EP(2)-receptors in HASM cells, antagonised the inhibitory effect of PGE(2), 16,16-dimethyl PGE(2) and ONO-AE1-259 with apparent pA(2) values of 5.85, 6.09 and 6.1 respectively. In contrast, the EP(4)-receptor antagonists, AH 23848B and L-161,982, failed to displace to the right the concentration-response curves that described the inhibition of GM-CSF release evoked by PGE(2) and ONO-AE1-259. 4. Inhibition of GM-CSF release by PGE(2) and 8-Br-cAMP was abolished in cells infected with an adenovirus vector encoding an inhibitor protein of cAMP-dependent protein kinase (PKA) but not by H-89, a purported small molecule inhibitor of PKA. 5. We conclude that prostanoid receptors of the EP(2)-subtype mediate the inhibitory effect of PGE(2) on GM-CSF release from HASM cells by recruiting a PKA-dependent pathway. In addition, the data illustrate that caution should be exercised when using H-89 in studies designed to assess the role of PKA in biological processes.


Asunto(s)
Alprostadil/análogos & derivados , Dinoprostona/análogos & derivados , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Prostaglandinas E Sintéticas/farmacología , Receptores de Prostaglandina E/clasificación , Receptores de Prostaglandina E/efectos de los fármacos , Transducción de Señal/fisiología , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Adenoviridae/genética , Adenoviridae/metabolismo , Adolescente , Adulto , Anciano , Alprostadil/farmacología , Supervivencia Celular , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Dinoprostona/farmacología , Inhibidores Enzimáticos/farmacología , Femenino , Expresión Génica/efectos de los fármacos , Vectores Genéticos/genética , Factor Estimulante de Colonias de Granulocitos/antagonistas & inhibidores , Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Humanos , Indometacina/farmacología , Interleucina-1/antagonistas & inhibidores , Interleucina-1/farmacología , Isoquinolinas/farmacología , Masculino , Persona de Mediana Edad , Misoprostol/farmacología , Miocitos del Músculo Liso/metabolismo , Prostaglandinas E Sintéticas/química , Prostaglandinas E Sintéticas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores de Prostaglandina E/antagonistas & inhibidores , Receptores de Prostaglandina E/fisiología , Subtipo EP2 de Receptores de Prostaglandina E , Subtipo EP4 de Receptores de Prostaglandina E , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología , Tráquea/citología , Xantonas/farmacología
14.
Front Pharmacol ; 4: 173, 2014 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-24454287

RESUMEN

Idiopathic pulmonary fibrosis is a progressive disease of unknown etiology characterized by a dysregulated wound healing response that leads to fatal accumulation of fibroblasts and extracellular matrix (ECM) in the lung, which compromises tissue architecture and lung function capacity. Injury to type II alveolar epithelial cells is thought to be the key event for the initiation of the disease, and so far both genetic factors, such as mutations in telomerase and MUC5B genes as well as environmental components, like cigarette smoking, exposure to asbestos and viral infections have been implicated as potential initiating triggers. The injured epithelium then enters a state of senescence-associated secretory phenotype whereby it produces both pro-inflammatory and pro-fibrotic factors that contribute to the wound healing process in the lung. Immune cells, like macrophages and neutrophils as well as activated myofibroblasts then perpetuate this cascade of epithelial cell apoptosis and proliferation by release of pro-fibrotic transforming growth factor beta and continuous deposition of ECM stiffens the basement membrane, altogether having a deleterious impact on epithelial cell function. In this review, we describe the role of the epithelium as both a physical and immunological barrier between environment and self in the homeostatic versus diseased lung and explore the potential mechanisms of epithelial cell injury and the impact of loss of epithelial cell permeability and function on cytokine production, inflammation, and myofibroblast activation in the fibrotic lung.

15.
Inflamm Allergy Drug Targets ; 12(2): 99-108, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23517646

RESUMEN

Chronic respiratory diseases such as asthma and chronic obstructive pulmonary disease (COPD) represent a significant health burden worldwide and are a major unmet medical need. Asthma affects over 300 million people and leads to 250,000 deaths per year, with an increasing prevalence particularly in developing countries. Although a large proportion of asthmatics are maintained on beta agonists and corticosteroids, there still remains a group of patients where these medicines fail to modulate symptoms and who may therefore benefit from monoclonal antibody based drugs that are aimed at controlling the disease. COPD is a cigarette smoke-driven chronic inflammatory airway disease with an increasing global prevalence. Given that current therapies fail to prevent disease progression or mortality, this patient population is also a focus for the development of monoclonal antibody therapies. At present anti-IgE (omalizumab, Xolair®) is the only monoclonal antibody based drug approved in the respiratory space for the treatment of asthma. However, an increasing number of antibodies targeting key mediators/pathways of disease are in clinical development for both asthma and COPD, including targeting the Th2 pathway for asthma (anti-IL-4/5/13) and the pro-inflammatory cytokine IL-1 for COPD. This review will examine the antibody engineering approaches used to develop the next generation of antibodies, with a focus on respiratory disease.


Asunto(s)
Anticuerpos Antiidiotipos/química , Anticuerpos Antiidiotipos/uso terapéutico , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/uso terapéutico , Asma/tratamiento farmacológico , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Animales , Humanos
16.
Front Pharmacol ; 4: 174, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24478701

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a fatal lung disorder of unknown etiology characterized by accumulation of lung fibroblasts and extracellular matrix deposition, ultimately leading to compromised tissue architecture and lung function capacity. IPF has a heterogeneous clinical course; however the median survival after diagnosis is only 3-5 years. The pharmaceutical and biotechnology industry has made many attempts to find effective treatments for IPF, but the disease has so far defied all attempts at therapeutic intervention. Clinical trial failures may arise for many reasons, including disease heterogeneity, lack of readily measurable clinical end points other than overall survival, and, perhaps most of all, a lack of understanding of the underlying molecular mechanisms of the progression of IPF. The precise link between inflammation and fibrosis remains unclear, but it appears that immune cells can promote fibrosis by releasing fibrogenic factors. So far, however, therapeutic approaches targeting macrophages, neutrophils, or lymphocytes have failed to alter disease pathogenesis. A new cell to garner research interest in fibrosis is the mast cell. Increased numbers of mast cells have long been known to be present in pulmonary fibrosis and clinically correlations between mast cells and fibrosis have been reported. More recent data suggests that mast cells may contribute to the fibrotic process by stimulating fibroblasts resident in the lung, thus driving the pathogenesis of the disease. In this review, we will discuss the mast cell and its physiological role in tissue repair and remodeling, as well as its pathological role in fibrotic diseases such as IPF, where the process of tissue repair and remodeling is thought to be dysregulated.

17.
Fibrogenesis Tissue Repair ; 6(1): 20, 2013 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-24279676

RESUMEN

Repairing damaged tissues is an essential homeostatic mechanism that enables clearance of dead or damaged cells after injury, and the maintenance of tissue integrity. However, exaggeration of this process in the lung can lead to the development of fibrotic scar tissue. This is characterized by excessive accumulation of extracellular matrix (ECM) components such as fibronectin, proteoglycans, hyaluronic acid, and interstitial collagens. After tissue injury, or a breakdown of tissue integrity, a cascade of events unfolds to maintain normal tissue homeostasis. Inflammatory mediators are released from injured epithelium, leading to both platelet activation and inflammatory cell migration. Inflammatory cells are capable of releasing multiple pro-inflammatory and fibrogenic mediators such as transforming growth factor (TGF)ß and interleukin (IL)-13, which can trigger myofibroblast proliferation and recruitment. The myofibroblast population is also expanded as a result of epithelial cells undergoing epithelial-to-mesenchymal transition and of the activation of resident fibroblasts, leading to ECM deposition and tissue remodeling. In the healthy lung, wound healing then proceeds to restore the normal architecture of the lung; however, fibrosis can develop when the wound is severe, the tissue injury persists, or the repair process becomes dysregulated. Understanding the processes regulating aberrant wound healing and the matrix in the chronic fibrotic lung disease idiopathic pulmonary fibrosis (IPF), is key to identifying new treatments for this chronic debilitating disease. This review focuses primarily on the emerging role of enzymes in the lungs of patients with IPF. Elevated expression of a number of enzymes that can directly modulate the ECM has been reported, and recent data indicates that modulating the activity of these enzymes can have a downstream effect on fibrotic tissue remodeling.

18.
EMBO Mol Med ; 4(12): 1244-60, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23165884

RESUMEN

The importance of NF-κB activation and deficient anti-viral interferon induction in the pathogenesis of rhinovirus-induced asthma exacerbations is poorly understood. We provide the first in vivo evidence in man and mouse that rhinovirus infection enhanced bronchial epithelial cell NF-κB p65 nuclear expression, NF-κB p65 DNA binding in lung tissue and NF-κB-regulated airway inflammation. In vitro inhibition of NF-κB reduced rhinovirus-induced pro-inflammatory cytokines but did not affect type I/III interferon induction. Rhinovirus-infected p65-deficient mice exhibited reduced neutrophilic inflammation, yet interferon induction, antiviral responses and virus loads were unaffected, indicating that NF-κB p65 is required for pro-inflammatory responses, but redundant in interferon induction by rhinoviruses in vivo. Conversely, IFNAR1(-/-) mice exhibited enhanced neutrophilic inflammation with impaired antiviral immunity and increased rhinovirus replication, demonstrating that interferon signalling was critical to antiviral immunity. We thus provide new mechanistic insights into rhinovirus infection and demonstrate the therapeutic potential of targeting NF-κB p65 (to suppress inflammation but preserve anti-viral immunity) and type I IFN signalling (to enhance deficient anti-viral immunity) to treat rhinovirus-induced exacerbations of airway diseases.


Asunto(s)
Inmunidad Innata , Interferón Tipo I/metabolismo , FN-kappa B/metabolismo , Fosfoproteínas/genética , Rhinovirus/fisiología , Animales , Asma/inmunología , Asma/metabolismo , Asma/virología , Células Cultivadas , Proteínas del Citoesqueleto , Células Epiteliales/metabolismo , Células Epiteliales/virología , Femenino , Células HeLa , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Interferón Tipo I/inmunología , Pulmón/inmunología , Pulmón/metabolismo , Ratones , Proteínas de Microfilamentos , Fosfoproteínas/metabolismo , Receptor de Interferón alfa y beta/metabolismo , Rhinovirus/inmunología , Rhinovirus/metabolismo
19.
Br J Pharmacol ; 157(3): 436-50, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19371341

RESUMEN

BACKGROUND AND PURPOSE: Endothelin-1 (ET-1) is implicated in airway inflammation in asthma, but the mechanisms of its effects are poorly understood. We studied the effect of ET-1 on expression of the chemokine, monocyte chemotactic protein-1 (MCP-1), in primary cultures of human airway smooth muscle cells. EXPERIMENTAL APPROACH: MCP-1 release was measured by elisa. Pharmacological antagonists/inhibitors, reverse transcriptase-polymerase chain reaction (RT-PCR) and Western blotting were used to study ET receptors and kinase cascades. Transcriptional regulation was studied by real-time RT-PCR, transient transfection studies and chromatin immunoprecipitation assay. Major findings were confirmed in cells from three donors and mechanistic studies in cells from one donor. KEY RESULTS: ET-1 increased MCP-1 release through an ET(A) and ET(B) receptor-dependent mechanism. ET-1 increased MCP-1 mRNA levels but not mRNA stability suggesting it was acting transcriptionally. ET-1 increased the activity of an MCP-1 promoter-reporter construct. Serial deletions of the MCP-1 promoter mapped ET-1 effects to a region between -213 and -128 base pairs upstream of the translation start codon, containing consensus sequences for activator protein-1 (AP-1) and nuclear factor-kappaB (NF-kappaB). ET-1 promoted binding of AP-1 c-Jun subunit and NF-kappaB p65 subunit to the MCP-1 promoter. Blocking the inhibitor of kappaB kinase-2 with 2-[(aminocarbonyl)amino]-5-[4-fluorophenyl]-3-thiophenecarboxamide (TPCA-1) decreased ET-1-stimulated MCP-1 production. p38 and p44/p42 mitogen-activated protein kinases were involved in upstream signalling. CONCLUSIONS AND IMPLICATIONS: ET-1 regulated MCP-1 transcriptionally, via NF-kappaB and AP-1. The upstream signalling involved ET(A), ET(B) receptors, p38 and p44/p42 mitogen-activated protein kinases. These may be targets for novel asthma therapies.


Asunto(s)
Quimiocina CCL2/metabolismo , Endotelina-1/fisiología , Miocitos del Músculo Liso/metabolismo , FN-kappa B/fisiología , Sistema Respiratorio/citología , Factor de Transcripción AP-1/fisiología , Células Cultivadas , Quimiocina CCL2/genética , Endotelina-1/farmacología , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Proteína Quinasa 3 Activada por Mitógenos/fisiología , Miocitos del Músculo Liso/citología , Fosfatidilinositol 3-Quinasas/fisiología , Receptor de Endotelina A/fisiología , Receptor de Endotelina B/fisiología , Transducción de Señal , Transcripción Genética , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
20.
Mol Pharmacol ; 67(2): 383-93, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15528403

RESUMEN

8-Isoprostanes are bioactive lipid mediators formed via the nonenzymatic peroxidation of arachidonic acid by free radicals and reactive oxygen species. However, their cognate receptors, biological actions, and signaling pathways are poorly studied. Here, we report the effect of a variety of E- and Falpha-ring 8-isoprostanes on the release of granulocyte/macrophage colony-stimulating factor (GM-CSF) and granulocyte colony-stimulating factor (G-CSF) from human airway smooth muscle (HASM) cells stimulated with interleukin-1beta (IL-1beta). The elaboration of GM-CSF and G-CSF by IL-1beta was inhibited and augmented, respectively, in a concentration-dependent manner by 8-iso-prostaglandin (PG) E(1) and 8-iso-PGE(2), but not by 8-iso-PGF(1alpha), 8-iso-PGF(2alpha), and 8-iso-PGF(3)alpha. AH 6809 (6-isopropoxy-9-oxoxanthine-2-carboxylic acid), an EP(1)-/EP(2)-/DP-receptor blocking drug, antagonized the inhibitory effect of 8-iso-PGE(1) and 8-iso-PGE(2) on GM-CSF output with an affinity consistent with an interaction at prostanoid receptors of the EP(2)-subtype. In contrast, the facilitation by 8-iso-PGE(1) and 8-iso-PGE(2) of G-CSF release was unaffected by AH 6809 and the selective EP(4)-receptor antagonist L-161,982 [4'-[3-butyl-5-oxo-1-(2-trifluoromethyl-phenyl)-1,5-dihydro-[1,2,4]triazol-4-ylmethyl]-biphenyl-2-sulfonic acid (3-methyl-thiophene-2-carbonyl)-amide]. However, when used in combination, AH 6809 and L-161,982 displaced 5-fold to the right the 8-iso-PGE and 8-iso-PGE concentration-response curves. The opposing (1)effect of E-ring (2)8-isoprostanes on GM-CSF and G-CSF release was mimicked by 8-bromo-cAMP and abolished in cells infected with an adenovirus vector encoding an inhibitor protein of cAMP-dependent protein kinase (PKA). Together, these data demonstrate that E-ring 8-isoprostanes regulate the secretion of GM-CSF and G-CSF from HASM cells by a cAMP- and PKA-dependent mechanism. Moreover, antagonist studies revealed that 8-iso-PGE(1) and 8-iso-PGE(2) act solely via EP(2) -receptors to inhibit GM-CSF release, whereas both EP(2)- and EP(4)-receptor subtypes positively regulate G-CSF output.


Asunto(s)
Factores Estimulantes de Colonias/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Dinoprost/análogos & derivados , Dinoprost/farmacología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Receptores de Prostaglandina E/agonistas , Adolescente , Adulto , Células Cultivadas , Dinoprost/química , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Receptores de Prostaglandina E/metabolismo , Receptores de Prostaglandina E/fisiología , Subtipo EP2 de Receptores de Prostaglandina E , Subtipo EP4 de Receptores de Prostaglandina E , Tráquea/efectos de los fármacos , Tráquea/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA