Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 66
1.
Surv Ophthalmol ; 69(4): 585-605, 2024.
Article En | MEDLINE | ID: mdl-38432359

Laser photocoagulation (LPC) and/or intravitreal anti-vascular endothelial growth factor (anti-VEGF) injections constitute the current standard treatment for retinopathy of prematurity (ROP). This network meta-analysis focus on whether a ranking of interventions may be established for different dose levels of intravitreal injection of anti-VEGF agents (aflibercept, bevacizumab, conbercept, ranibizumab) as primary treatments for ROP versus laser in terms of retreatment rate as primary outcome, and time to retreatment and refractive error as secondary endpoints, since best anti-VEGF dosage remains under debate. Sixty-eight studies (15 randomized control trials and 53 nonrandomized studies) of 12,356 eyes of 6445 infants were retrieved from databases (2005 Jan. - 2023 June). Studies were evaluated for model fit, risk of bias and confidence of evidence in Network Meta-Analysis (CINeMA). Bayesian NMA showed that anti-VEGF drugs were not inferior to laser in terms of retreatment rate. For intravitreal bevacizumab (IVB), doses half of the conventional infant dose showed a low risk of retreatment rate (risk ratio (RR) of 1.43; 95% credible interval (CrI): 0.508, 4.03). On probability ranking as surface under the cumulative ranking curve (SUCRA) plot, half dose of bevacizumab had a better position than conventional and augmented (1.2-2 times the regular dose) doses. A similar probability trend was observed for half vs. conventional doses of aflibercept and ranibizumab. Conventional infant dose of conbercept showed the lowest risk for retreatment (RR 0.846; 95% CrI: 0.245, 2.91). For secondary endpoints, lower doses of anti-VEGF agents were associated with shorter times to retreatment. The largest changes were noted for the augmented doses of bevacizumab and ranibizumab (0.3 mg) with means of 14.1 weeks (95% CrI: 6.65, 21.6) and 12.8 weeks (95% CrI: 3.19, 20.9), respectively. Finally, NMA demonstrated better refractive profile for anti-VEGF than laser therapy, especially for the conventional infant doses of bevacizumab and ranibizumab which exhibited a significantly better refractive profile than LPC, with mean differences of 1.67 (spherical equivalent - diopters) (95% CrI: 0.705, 2.67) and 2.19 (95% CrI: 0.782, 3.59), respectively. In the SUCRA plots, LPC had a markedly different position with a higher probability for myopia. Further clinical trials comparing different intravitreal doses of anti-VEGF agents are needed, but our findings suggest that low doses of these drugs retain efficacy and may reduce ocular and systemic undesired events.


Angiogenesis Inhibitors , Bevacizumab , Intravitreal Injections , Laser Coagulation , Network Meta-Analysis , Ranibizumab , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins , Retinopathy of Prematurity , Vascular Endothelial Growth Factor A , Humans , Retinopathy of Prematurity/drug therapy , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/therapeutic use , Receptors, Vascular Endothelial Growth Factor/administration & dosage , Bevacizumab/administration & dosage , Bevacizumab/therapeutic use , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/therapeutic use , Ranibizumab/administration & dosage , Ranibizumab/therapeutic use , Laser Coagulation/methods , Infant, Newborn
2.
Mucosal Immunol ; 14(2): 377-388, 2021 03.
Article En | MEDLINE | ID: mdl-32887938

Several mucins are implicated in idiopathic pulmonary fibrosis (IPF); however, there is no evidence regarding the role of MUC4 in the development of IPF. Here we demonstrated that MUC4 was overexpressed in IPF patients (n = 22) compared with healthy subjects (n = 21) and located in pulmonary arteries, bronchial epithelial cells, fibroblasts, and hyperplastic alveolar type II cells. Decreased expression of MUC4 using siRNA-MUC4 inhibited the mesenchymal/myofibroblast transformations of alveolar type II A549 cells and lung fibroblasts, as well as cell senescence and fibroblast proliferation induced by TGF-ß1. The induction of the overexpression of MUC4 increased the effects of TGF-ß1 on mesenchymal/myofibroblast transformations and cell senescence. MUC4 overexpression and siRNA-MUC4 gene silencing increased or decreased, respectively, the phosphorylation of TGFßRI and SMAD3, contributing to smad-binding element activation. Immunoprecipitation analysis and confocal immunofluorescence showed the formation of a protein complex between MUC4ß/p-TGFßRI and p-SMAD3 in the cell membrane after TGF-ß1 stimulation and in lung tissue from IPF patients. Bleomycin-induced lung fibrosis was reduced in mice transiently transfected with siRNA-MUC4. This study shows that MUC4 expression is enhanced in IPF and promotes fibrotic processes in collaboration with TGF-ß1 canonical pathway that could be an attractive druggable target for human IPF.


Fibroblasts/pathology , Idiopathic Pulmonary Fibrosis/metabolism , Lung/pathology , Mucin-4/metabolism , Respiratory Mucosa/metabolism , A549 Cells , Cellular Senescence , Gene Expression Regulation, Neoplastic , Humans , Idiopathic Pulmonary Fibrosis/immunology , Molecular Targeted Therapy , Mucin-4/genetics , RNA, Small Interfering/genetics , Respiratory Mucosa/pathology , Signal Transduction , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Up-Regulation
3.
Thorax ; 75(2): 132-142, 2020 02.
Article En | MEDLINE | ID: mdl-31801904

BACKGROUND: Serum KL6/mucin 1 (MUC1) has been identified as a potential biomarker in idiopathic pulmonary fibrosis (IPF), but the role of MUC1 intracellular bioactivation in IPF is unknown. OBJECTIVE: To characterise MUC1 intracellular bioactivation in IPF. METHODS AND RESULTS: The expression and phosphorylation of Thr41 and Tyr46 on the intracellular MUC1-cytoplasmic tail (CT) was increased in patients with IPF (n=22) compared with healthy subjects (n=21) and localised to fibroblasts and hyperplastic alveolar type II cells. Transforming growth factor (TGF)-ß1 phosphorylated SMAD3 and thereby increased the phosphorylation of MUC1-CT Thr41 and Tyr46 in lung fibroblasts and alveolar type II cells, activating ß-catenin to form a phospho-Smad3/MUC1-CT and MUC1-CT/ß-catenin nuclear complex. This nuclear complex promoted alveolar epithelial type II and fibroblast to myofibroblast transitions, as well as cell senescence and fibroblast proliferation. The inhibition of MUC1-CT nuclear translocation using the inhibitor, GO-201 or silencing MUC1 by siRNA, reduced myofibroblast transition, senescence and proliferation in vitro. Bleomycin-induced lung fibrosis was reduced in mice treated with GO-201 and in MUC1-knockout mice. The profibrotic lectin, galectin-3, directly activated MUC1-CT and served as a bridge between the TGF-ß receptor and the MUC1-C domain, indicating TGF-ß1-dependent and TGF-ß1-independent intracellular bioactivation of MUC1. CONCLUSIONS: MUC1 intracellular bioactivation is enhanced in IPF and promotes fibrotic processes that could represent potential druggable targets for IPF.


Gene Expression Regulation/genetics , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/genetics , Mucin-1/genetics , Transforming Growth Factor beta1/genetics , Animals , Biopsy, Needle , Bleomycin/pharmacology , Cells, Cultured , Disease Models, Animal , Female , Fibroblasts/drug effects , Humans , Idiopathic Pulmonary Fibrosis/pathology , Immunohistochemistry , Male , Mice , Mice, Knockout , Molecular Targeted Therapy/methods , RNA, Messenger/genetics , Risk Assessment , Signal Transduction/genetics , Smad3 Protein/genetics
4.
PLoS One ; 14(1): e0210188, 2019.
Article En | MEDLINE | ID: mdl-30608978

Recent evidence indicates that AZD8999 (LAS190792), a novel muscarinic acetylcholine receptor antagonist and ß2-adrenoceptor agonist (MABA) in development for chronic respiratory diseases, induces potent and sustained relaxant effects in human bronchi by adressing both muscarinic acetylcholine receptors and ß2-adrenoceptor. However, the anti-inflammatory effects of the AZD8999 monotherapy or in combination with corticosteroids are unknown. This study investigates the anti-inflammatory effects of AZD8999 in monotherapy and combined with fluticasone propionate in neutrophils from healthy and chronic obstructive pulmonary disease (COPD) patients. Peripheral blood neutrophils from healthy and COPD patients were incubated with AZD8999 and fluticasone propionate, individually or in combination, for 1h followed by lipopolysaccharide (LPS) stimulation for 6h. The IL-8, MMP9, IL-1ß, and GM-CSF release was measured in cell culture supernatants. AZD8999 shows ~ 50% maximum inhibitory effect and similar potency inhibiting the released cytokines in neutrophils from healthy and COPD patients. However, while fluticasone propionate suppresses mediator release in neutrophils from healthy patients, COPD neutrophils are less sensitive. The combination of non-effective concentrations of AZD8999 (0.01nM) with non-effective concentrations of fluticasone propionate (0.1nM) shows synergistic anti-inflammatory effects. The studied mechanisms that may be involved in the synergistic anti-inflammatory effects of this combination include the increase of glucocorticoid receptor (GR)α and MKP1 expression, the induction of glucocorticoid response element (GRE) activation and the decrease of ERK1/2, P38 and GR-Ser226 phosphorylations compared with monotherapies. In summary, AZD8999 shows anti-inflammatory effects in neutrophils from COPD patients and induces synergistic anti-inflammatory effects when combined with fluticasone propionate, supporting the use of MABA/ICS combination therapy in COPD.


Anti-Inflammatory Agents/pharmacology , Cyclohexanes/pharmacology , Neutrophils/drug effects , Pulmonary Disease, Chronic Obstructive/drug therapy , Quinolines/pharmacology , Thiophenes/pharmacology , Adrenergic beta-2 Receptor Agonists/pharmacology , Adrenergic beta-2 Receptor Agonists/therapeutic use , Aged , Anti-Inflammatory Agents/therapeutic use , Cyclohexanes/therapeutic use , Dose-Response Relationship, Drug , Drug Synergism , Drug Therapy, Combination/methods , Female , Fluticasone/pharmacology , Fluticasone/therapeutic use , Healthy Volunteers , Humans , Male , Middle Aged , Muscarinic Antagonists/pharmacology , Muscarinic Antagonists/therapeutic use , Neutrophils/immunology , Neutrophils/metabolism , Pulmonary Disease, Chronic Obstructive/immunology , Quinolines/therapeutic use , Receptors, Adrenergic, beta-2/metabolism , Receptors, Muscarinic/metabolism , Thiophenes/therapeutic use
5.
Allergy ; 74(1): 111-121, 2019 01.
Article En | MEDLINE | ID: mdl-29978485

BACKGROUND: The loss of corticosteroid efficacy is an important issue in severe asthma management and may lead to poor asthma control and deterioration of airflow. Recent data indicate that Mucin 1 (MUC1) membrane mucin can mediate corticosteroid efficacy in chronic rhinosinusitis, but the role of MUC1 in uncontrolled severe asthma is unknown. The objective was to analyze the previously unexplored role of MUC1 on corticosteroid efficacy in asthma. METHODS: Mucin 1 expression was evaluated by real-time PCR in human bronchial epithelial cells (HBEC) and blood neutrophils from uncontrolled severe asthma (n = 27), controlled mild asthma (n = 16), and healthy subjects (n = 13). IL-8, MMP9, and GM-CSF were measured by ELISA in HBEC and neutrophils. An asthma model of ovalbumin (OVA) was used in MUC1 KO and WT C57BL/6 mice according to ARRIVE guidelines. RESULTS: Mucin 1-CT expression was downregulated in bronchial epithelial cells and peripheral blood neutrophils from severe asthma patients compared with mild asthma and healthy subjects (P < 0.05). Daily dose of inhaled corticosteroids (ICS) inversely correlated with MUC1 expression in neutrophils from mild and severe asthma (ρ = -0.71; P < 0.0001). Dexamethasone showed lower anti-inflammatory effects in severe asthma peripheral blood neutrophils and HBECs stimulated with lipopolysaccharide (LPS) than in cells from mild asthma. Glucocorticoid receptor (GR)-α phosphorylated at serine 226 was increased in cells from severe asthma, and the MUC1-CT/GRα complex was downregulated in severe asthma cells. OVA asthma model in MUC1 KO mice was resistant to the anti-inflammatory effects of dexamethasone. CONCLUSION: Mucin 1-CT modulates corticosteroid efficacy in vitro and in vivo asthma models.


Adrenal Cortex Hormones/pharmacology , Asthma/drug therapy , Drug Resistance , Mucin-1/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Asthma/metabolism , Case-Control Studies , Cells, Cultured , Dexamethasone/pharmacology , Epithelial Cells/metabolism , Humans , Mice , Neutrophils/metabolism , Receptors, Glucocorticoid/metabolism
6.
Respir Res ; 19(1): 226, 2018 Nov 20.
Article En | MEDLINE | ID: mdl-30458870

BACKGROUND: Lung inflammation in COPD is poorly controlled by inhaled corticosteroids (ICS). Strategies to improve ICS efficacy or the search of biomarkers who may select those patients candidates to receive ICS in COPD are needed. Recent data indicate that MUC1 cytoplasmic tail (CT) membrane mucin can mediate corticosteroid efficacy in chronic rhinosinusitis. The objective of this work was to analyze the previously unexplored role of MUC1 on corticosteroid efficacy in COPD in vitro and in vivo models. METHODS: MUC1-CT expression was measured by real time PCR, western blot, immunohistochemistry and immunofluorescence. The inflammatory mediators IL-8, MMP9, GM-CSF and MIP3α were measured by ELISA. The effect of MUC1 on inflammation and corticosteroid anti-inflammatory effects was measured using cell siRNA in vitro and Muc1-KO in vivo animal models. RESULTS: MUC1-CT expression was downregulated in lung tissue, bronchial epithelial cells and lung neutrophils from smokers (n = 11) and COPD (n = 11) patients compared with healthy subjects (n = 10). MUC1 was correlated with FEV1% (ρ = 0.7479; p < 0.0001) in smokers and COPD patients. Cigarette smoke extract (CSE) decreased the expression of MUC1 and induced corticosteroid resistance in human primary bronchial epithelial cells and human neutrophils. MUC1 Gene silencing using siRNA-MUC1 impaired the anti-inflammatory effects of dexamethasone and reduced glucocorticoid response element activation. Dexamethasone promoted glucocorticoid receptor alpha (GRα) and MUC1-CT nuclear translocation and co-localization that was inhibited by CSE. Lung function decline and inflammation induced by lipopolysaccharide and cigarette smoke in Muc1 KO mice was resistant to dexamethasone. CONCLUSIONS: These results confirm a role for MUC1-CT mediating corticosteroid efficacy in COPD.


Adrenal Cortex Hormones/therapeutic use , Drug Resistance/physiology , Mucin-1/metabolism , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/metabolism , Adrenal Cortex Hormones/pharmacology , Aged , Animals , Biomarkers/blood , Biomarkers/metabolism , Drug Resistance/drug effects , Female , Gene Silencing/drug effects , Gene Silencing/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Mucin-1/genetics , Pulmonary Disease, Chronic Obstructive/genetics , Sputum/metabolism
7.
Thorax ; 73(6): 519-529, 2018 06.
Article En | MEDLINE | ID: mdl-29440315

BACKGROUND: Pulmonary hypertension (PH) is a common disorder in patients with idiopathic pulmonary fibrosis (IPF) and portends a poor prognosis. Recent studies using vasodilators approved for PH have failed in improving IPF mainly due to ventilation (V)/perfusion (Q) mismatching and oxygen desaturation. Janus kinase type 2 (JAK2) is a non-receptor tyrosine kinase activated by a broad spectrum of profibrotic and vasoactive mediators, but its role in PH associated to PH is unknown. OBJECTIVE: The study of JAK2 as potential target to treat PH in IPF. METHODS AND RESULTS: JAK2 expression was increased in pulmonary arteries (PAs) from IPF (n=10; 1.93-fold; P=0.0011) and IPF+PH (n=9; 2.65-fold; P<0.0001) compared with PA from control subjects (n=10). PA remodelling was evaluated in human pulmonary artery endothelial cells (HPAECs) and human pulmonary artery smooth muscle cells (HPASMCs) from patients with IPF in vitro treated with the JAK2 inhibitor JSI-124 or siRNA-JAK2 and stimulated with transforming growth factor beta. Both JSI-124 and siRNA-JAK2 inhibited the HPAEC to mesenchymal transition and the HPASMCs to myofibroblast transition and proliferation. JAK2 inhibition induced small PA relaxation in precision-cut lung slice experiments. PA relaxation was dependent of the large conductance calcium-activated potassium channel (BKCa). JAK2 inhibition activated BKCa channels and reduced intracellular Ca2+. JSI-124 1 mg/kg/day, reduced bleomycin-induced lung fibrosis, PA remodelling, right ventricular hypertrophy, PA hypertension and V/Q mismatching in rats. The animal studies followed the ARRIVE guidelines. CONCLUSIONS: JAK2 participates in PA remodelling and tension and may be an attractive target to treat IPF associated to PH.


Hypertension, Pulmonary/drug therapy , Idiopathic Pulmonary Fibrosis/drug therapy , Janus Kinase 2/antagonists & inhibitors , Triterpenes/pharmacology , Vascular Remodeling/drug effects , Animals , Blotting, Western , Cell Proliferation/drug effects , Cells, Cultured , Endothelial Cells , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Janus Kinase 2/metabolism , Myocytes, Smooth Muscle , Phenotype , RNA, Small Interfering/pharmacology , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Signal Transduction , Transforming Growth Factor beta/pharmacology
8.
Respir Res ; 19(1): 24, 2018 02 06.
Article En | MEDLINE | ID: mdl-29409529

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is the most rapidly progressive and fatal fibrotic disorder, with no curative therapies. The signal transducer and activator of transcription 3 (STAT3) protein is activated in lung fibroblasts and alveolar type II cells (ATII), thereby contributing to lung fibrosis in IPF. Although activation of Janus kinase 2 (JAK2) has been implicated in proliferative disorders, its role in IPF is unknown. The aim of this study was to analyze JAK2 activation in IPF, and to determine whether JAK2/STAT3 inhibition is a potential therapeutic strategy for this disease. METHODS AND RESULTS: JAK2/p-JAK2 and STAT3/pSTAT3 expression was evaluated using quantitative real time-PCR, western blotting, and immunohistochemistry. Compared to human healthy lung tissue (n = 10) both proteins were upregulated in the lung tissue of IPF patients (n = 12). Stimulating primary ATII and lung fibroblasts with transforming growth factor beta 1 or interleukin (IL)-6/IL-13 activated JAK2 and STAT3, inducing epithelial to mesenchymal and fibroblast to myofibroblast transitions. Dual p-JAK2/p-STAT3 inhibition with JSI-124 or silencing of JAK2 and STAT3 genes suppressed ATII and the fibroblast to myofibroblast transition, with greater effects than the sum of those obtained using JAK2 or STAT3 inhibitors individually. Dual rather than single inhibition was also more effective for inhibiting fibroblast migration, preventing increases in fibroblast senescence and Bcl-2 expression, and ameliorating impaired autophagy. In rats administered JSI-124, a dual inhibitor of p-JAK2/p-STAT3, at a dose of 1 mg/kg/day, bleomycin-induced lung fibrosis was reduced and collagen deposition in the lung was inhibited, as were JAK2 and STAT3 activation and several markers of fibrosis, autophagy, senescence, and anti-apoptosis. CONCLUSIONS: JAK2 and STAT3 are activated in IPF, and their dual inhibition may be an attractive strategy for treating this disease.


Idiopathic Pulmonary Fibrosis/enzymology , Idiopathic Pulmonary Fibrosis/pathology , Janus Kinase 2/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , A549 Cells , Adult , Aged , Animals , Enzyme Activation/drug effects , Enzyme Activation/physiology , Female , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/pathology , Humans , Janus Kinase 2/antagonists & inhibitors , Male , Middle Aged , Rats , STAT3 Transcription Factor/antagonists & inhibitors , Triterpenes/pharmacology
9.
Curr Pharm Des ; 23(14): 2073-2083, 2017.
Article En | MEDLINE | ID: mdl-28201975

Chronic respiratory diseases affect millions of people every day. According to the World Health Organization estimates, ~235 million people suffer from asthma, ~64 million suffer from chronic obstructive pulmonary disease (COPD), and millions more suffer from allergic rhinitis around the world. In recent last years, the first phosphodiesterase 4 (PDE4) inhibitor, roflumilast, was approved as a treatment to reduce the risk of exacerbations in stable and severe COPD associated with chronic bronchitis and a history of exacerbations. PDE4 exists as four subtypes (A, B, C, and D) each with a capacity to degrade cAMP, a second messenger involved in inflammatory responses. PDE4 inhibitors inhibit PDE4 activity, consequently increasing cAMP levels. This results in an anti-inflammatory effect, improving lung function. Roflumilast is a selective and non-specific PDE4 inhibitor, with the potential to inhibit all PDE4 isoforms to some degree. Despite the pharmacological effects of roflumilast, its lack of specificity can induce side effects, such as diarrhea, nausea, and dizziness. Thus, there is a continuing need to develop more specific inhibitors of the individual PDE4 subtypes. PDE4B and D inhibitors have been investigated the most, because the levels of these two subtypes are upregulated in moderate and severe COPD. Current and new evidences show that PDE4B and D inhibitors are the most studied, because their expressions are up-regulated in moderate and severe COPD. This review highlights the major advantages of the selective specific inhibition of PDE4A, B, C, and D versus selective, non-specific inhibitors as treatments for chronic respiratory diseases.


Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , Pulmonary Disease, Chronic Obstructive/drug therapy , Animals , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism
10.
J Allergy Clin Immunol ; 139(3): 855-862.e13, 2017 Mar.
Article En | MEDLINE | ID: mdl-27639937

BACKGROUND: Current evidence suggests that membrane-tethered mucins could mediate corticosteroid efficacy, interacting with glucocorticoid receptor (GR) in patients with chronic rhinosinusitis with nasal polyps (CRSwNP). Mucin 4 (MUC4)-tethered mucin is expressed in nasal polyp (NP) epithelial cells and upregulated under inflammatory conditions. Moreover, MUC4ß has the capacity to interact with other intracellular proteins. We hypothesized that MUC4 modulates corticosteroid efficacy of patients with CRSwNP. OBJECTIVE: We sought to analyze the role of MUC4 in corticosteroid effectiveness in different cohorts of patients with CRSwNP and elucidate the possible mechanisms involved. METHODS: Eighty-one patients with CRSwNP took oral corticosteroids for 15 days. Corticosteroid resistance was evaluated by using nasal endoscopy. Expression of MUC4 and MUC4ß was evaluated by means of real-time PCR, Western blotting, and immunohistochemistry. BEAS-2B knockdown with RNA interference for MUC4 (small interfering RNA [siRNA]-MUC4) was used to analyze the role of MUC4 in the anti-inflammatory effects of dexamethasone. RESULTS: Twenty-two patients had NPs resistant to oral corticosteroids. MUC4 expression was upregulated in these patients. In siRNA-MUC4 BEAS-2B airway epithelial cells dexamethasone produced higher anti-inflammatory effects, increased inhibition of phospho-extracellular signal-regulated kinase 1/2, increased mitogen-activated protein kinase phosphatase 1 expression, and increased glucocorticoid response element activation. Immunoprecipitation and immunofluorescence experiments revealed that MUC4ß forms a complex with GRα in the nuclei of NP epithelial cells from corticosteroid-resistant patients. CONCLUSION: MUC4ß participates in the corticosteroid resistance process, inhibiting normal GRα nuclear function. The high expression of MUC4 in patients with CRSwNP might participate in corticosteroid resistance.


Anti-Inflammatory Agents/therapeutic use , Drug Resistance , Mucin-4/immunology , Nasal Polyps/drug therapy , Pregnenediones/therapeutic use , Rhinitis/drug therapy , Sinusitis/drug therapy , Adult , Aged , Anti-Inflammatory Agents/pharmacology , Cell Line , Cells, Cultured , Chronic Disease , Dexamethasone/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , HEK293 Cells , Humans , Male , Middle Aged , Mucin-4/genetics , Young Adult
11.
Respir Res ; 17(1): 145, 2016 11 08.
Article En | MEDLINE | ID: mdl-27825347

BACKGROUND: Inhaled corticosteroid (ICS) with long-acting beta-2 agonists is a well-documented combination therapy for chronic obstructive pulmonary disease (COPD) based on its additive anti-inflammatory properties. By contrast, the recommendation of ICS in combination with long-acting muscarinic antagonist (LAMA) is not evidence-based. In this study, neutrophils obtained from COPD patients were used to compare the anti-inflammatory effects of aclidinium bromide (a long-acting muscarinic antagonist) with corticosteroids and their potential additive effect. METHODS: Human sputum and blood neutrophils were isolated from healthy individuals (n = 37), patients with stable COPD (n = 52) and those with exacerbated COPD (n = 16). The cells were incubated with corticosteroid fluticasone propionate (0.1 nM-1 µM), aclidinium bromide (0.1 nM-1 µM) or a combination thereof and stimulated with 1 µg of lipopolysaccharide/ml or 5 % cigarette smoke extract. Levels of the pro-inflammatory mediators interleukin-8, matrix metalloproteinase-9, CCL-5, granulocyte-macrophage colony-stimulating factor and interleukin-1ß were measured and the mechanisms of corticosteroid resistance evaluated at the end of the incubation. RESULTS: The non-neuronal cholinergic system was over-expressed in neutrophils from COPD patients, as evidenced by increases in the expression of muscarinic receptors (M2, M4 and M5), choline acetyltransferase and vesicular acetylcholine transporter. Aclidinium bromide demonstrated anti-inflammatory effects on neutrophils from COPD patients, reversing their resistance to corticosteroids. Additive effects of combined aclidinium bromide and fluticasone propionate in blocking M2 receptor levels, inhibiting phosphoinositide 3-kinase-δ and enhancing the glucocorticoid response element transcription factor were demonstrated and were accompanied by an increase in the corticosteroid-induced expression of anti-inflammatory-related genes. CONCLUSIONS: LAMAs potentiate the anti-inflammatory effects of corticosteroids in neutrophils from COPD patients in vitro, thus providing a scientific rationale for their use in combination with corticosteroids in the treatment of COPD.


Anti-Inflammatory Agents/pharmacology , Bronchodilator Agents/pharmacology , Drug Resistance/drug effects , Fluticasone/pharmacology , Muscarinic Antagonists/pharmacology , Neutrophils/drug effects , Non-Neuronal Cholinergic System/drug effects , Pulmonary Disease, Chronic Obstructive/drug therapy , Tropanes/pharmacology , Aged , Case-Control Studies , Choline O-Acetyltransferase/metabolism , Dose-Response Relationship, Drug , Drug Synergism , Drug Therapy, Combination , Female , Humans , Inflammation Mediators/metabolism , Male , Middle Aged , Neutrophils/metabolism , Pulmonary Disease, Chronic Obstructive/blood , Receptors, Muscarinic/drug effects , Receptors, Muscarinic/metabolism , Sputum/metabolism , Vesicular Acetylcholine Transport Proteins/metabolism
12.
PLoS One ; 11(10): e0164399, 2016.
Article En | MEDLINE | ID: mdl-27723827

Different image techniques have been used to analyze mucociliary clearance (MCC) in humans, but current small animal MCC analysis using in vivo imaging has not been well defined. Bitter taste receptor (T2R) agonists increase ciliary beat frequency (CBF) and cause bronchodilation but their effects in vivo are not well understood. This work analyzes in vivo nasal and bronchial MCC in guinea pig animals using three dimension (3D) micro-CT-SPECT images and evaluates the effect of T2R agonists. Intranasal macroaggreggates of albumin-Technetium 99 metastable (MAA-Tc99m) and lung nebulized Tc99m albumin nanocolloids were used to analyze the effect of T2R agonists on nasal and bronchial MCC respectively, using 3D micro-CT-SPECT in guinea pig. MAA-Tc99m showed a nasal mucociliary transport rate of 0.36 mm/min that was increased in presence of T2R agonist to 0.66 mm/min. Tc99m albumin nanocolloids were homogeneously distributed in the lung of guinea pig and cleared with time-dependence through the bronchi and trachea of guinea pig. T2R agonist increased bronchial MCC of Tc99m albumin nanocolloids. T2R agonists increased CBF in human nasal ciliated cells in vitro and induced bronchodilation in human bronchi ex vivo. In summary, T2R agonists increase MCC in vivo as assessed by 3D micro-CT-SPECT analysis.


Albumins , Lung , Mucociliary Clearance/physiology , Nanoparticles , Receptors, G-Protein-Coupled/agonists , Single Photon Emission Computed Tomography Computed Tomography/methods , X-Ray Microtomography/methods , Albumins/pharmacokinetics , Albumins/pharmacology , Animals , Guinea Pigs , Humans , Lung/diagnostic imaging , Mucociliary Clearance/drug effects
13.
Eur Respir J ; 47(6): 1737-49, 2016 06.
Article En | MEDLINE | ID: mdl-27009174

Sildenafil improves the 6-min walking distance in patients with idiopathic pulmonary fibrosis (IPF) and right-sided ventricular systolic dysfunction.We analysed the previously unexplored role of sildenafil on vasoconstriction and remodelling of pulmonary arteries from patients with IPF and pulmonary hypertension (PH) ex vivo Pulmonary arteries from 18 donors without lung disease, nine IPF, eight PH+IPF and four PH patients were isolated to measure vasodilator and anti-contractile effects of sildenafil in isometric organ bath. Ventilation/perfusion was explored in an animal model of bleomycin lung fibrosis.Sildenafil relaxed serotonin (5-HT) pre-contracted pulmonary arteries in healthy donors and IPF patients and, to a lesser extent, in PH+IPF and PH. Sildenafil inhibited 5-HT dose-response contraction curve mainly in PH+IPF and PH, but not in healthy donors. Sildenafil did not impair the ventilation/perfusion mismatching induced by bleomycin. Pulmonary arteries from PH+IPF patients showed a marked expression of phosphodiesterse-5 and extracellular matrix components. Sildenafil inhibited pulmonary artery endothelial and smooth muscle cell to mesenchymal transition by inhibition of extracellular regulated kinases 1 and 2 (ERK1/2) and SMAD3 phosphorylation.These results suggest an absence of direct relaxant effect and a prominent anti-contractile and anti-remodelling role of sildenafil in PH+IPF pulmonary arteries that could explain the beneficial effects of sildenafil in IPF with PH phenotype.


Hypertension, Pulmonary/drug therapy , Pulmonary Fibrosis/drug therapy , Sildenafil Citrate/therapeutic use , Animals , Bleomycin/chemistry , Bleomycin/pharmacology , Disease Models, Animal , Endothelium, Vascular/pathology , Extracellular Matrix/metabolism , Fibroblasts/chemistry , Humans , Lung/pathology , Male , Myocytes, Smooth Muscle/metabolism , Myofibroblasts/metabolism , Pulmonary Artery/physiopathology , Pulmonary Disease, Chronic Obstructive/physiopathology , Rats , Rats, Wistar , Serotonin/chemistry , Signal Transduction , Sildenafil Citrate/pharmacology , Transforming Growth Factor beta1/metabolism , Vasoconstriction , Vasodilator Agents
14.
PLoS One ; 10(7): e0133453, 2015.
Article En | MEDLINE | ID: mdl-26192616

Fibrotic remodeling is a process common to chronic lung diseases such as chronic obstructive pulmonary disease (COPD), pulmonary fibrosis, acute respiratory distress syndrome and asthma. Based on preclinical studies phosphodiesterase 4 (PDE4) inhibitors may exhibit beneficial anti-inflammatory and anti-remodeling properties for the treatment of these respiratory disorders. Effects of PDE4 inhibitors on changes in the lung metabolome in models of pulmonary fibrotic remodeling have not yet been explored. This work studies the effects of the PDE4 inhibitor roflumilast on changes in the lung metabolome in the common murine model of bleomycin-induced lung fibrosis by nuclear magnetic resonance (NMR) metabolic profiling of intact lung tissue. Metabolic profiling reveals strong differences between fibrotic and non-fibrotic tissue. These differences include increases in proline, glycine, lactate, taurine, phosphocholine and total glutathione and decreases in global fatty acids. In parallel, there was a loss in plasma BH4. This profile suggests that bleomycin produces alterations in the oxidative equilibrium, a strong inflammatory response and activation of the collagen synthesis among others. Roflumilast prevented most of these metabolic effects associated to pulmonary fibrosis suggesting a favorable anti-fibrotic profile.


Aminopyridines/pharmacology , Benzamides/pharmacology , Lung/drug effects , Metabolome , Pulmonary Fibrosis/metabolism , Animals , Bleomycin , Cyclopropanes/pharmacology , Disease Models, Animal , Lung/metabolism , Lung/pathology , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology
15.
Respir Res ; 16: 12, 2015 Feb 05.
Article En | MEDLINE | ID: mdl-25652132

BACKGROUND: Chronic obstructive pulmonary disease (COPD) is characterised by chronic pulmonary inflammation punctuated by periods of viral exacerbations. Recent evidence suggests that the combination of roflumilast with corticosteroids may improve the compromised anti-inflammatory properties of corticosteroids in COPD. We analyzed differential and combination anti-inflammatory effects of dexamethasone and roflumilast N-oxide in human bronchial epithelial cells (HBECs) stimulated with viral toll like receptor (TLR) agonists. METHODS: Lung tissue and HBECs were isolated from healthy (n = 15), smokers (n = 12) and smokers with COPD (15). TLR3 expression was measured in lung tissue and in HBECs. IL-8 secretion was measured in cell cultures after TLR3 stimulation with poly I:C 10 µg/mL. RESULTS: We found that TLR3 expression was increased by 1.95 fold (protein) and 2.5 fold (mRNA) in lung tissues from smokers with COPD and inversely correlated with lung function. The TLR3 agonist poly I:C 10 µg/mL increased the IL-8 release in HBECs that was poorly inhibited by dexamethasone in smokers (24.5%) and smokers with COPD (21.6%). In contrast, roflumilast showed similar inhibitory effects on IL-8 release in healthy (58.8%), smokers (56.6%) and smokers with COPD (50.5%). The combination of roflumilast N-oxide and dexamethasone showed additive inhibitory effects. Mechanistically, roflumilast N-oxide when combined with dexamethasone increased the expression of MKP1, and enhanced the inhibitory effects on phospho-p38, AP1 and NFκB activities which may explain the additive anti-inflammatory effects. CONCLUSIONS: Altogether, our data provide in vitro evidence for a possible clinical utility to add roflumilast on top of inhaled corticosteroid in COPD.


Adrenal Cortex Hormones/pharmacology , Aminopyridines/pharmacology , Anti-Inflammatory Agents/pharmacology , Benzamides/pharmacology , Bronchi/drug effects , Dexamethasone/pharmacology , Drug Resistance , Epithelial Cells/drug effects , Poly I-C/pharmacology , Pulmonary Disease, Chronic Obstructive/drug therapy , Toll-Like Receptor 3/agonists , Aged , Bronchi/immunology , Bronchi/metabolism , Case-Control Studies , Cells, Cultured , Cyclopropanes/pharmacology , Drug Therapy, Combination , Epithelial Cells/immunology , Epithelial Cells/metabolism , Female , Humans , Interleukin-8/immunology , Interleukin-8/metabolism , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/metabolism , Signal Transduction/drug effects , Smoking/adverse effects , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/immunology , Toll-Like Receptor 3/metabolism
16.
Eur J Pharmacol ; 715(1-3): 172-80, 2013 Sep 05.
Article En | MEDLINE | ID: mdl-23747655

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


Eosinophils/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Apoptosis/drug effects , Calcium/metabolism , Cell Movement/drug effects , Eosinophil Cationic Protein/biosynthesis , Eosinophils/cytology , Eosinophils/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interleukin-5/pharmacology , Leukotriene C4/biosynthesis , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Peroxidase/metabolism , Superoxides/metabolism
17.
Arterioscler Thromb Vasc Biol ; 33(1): 96-104, 2013 Jan.
Article En | MEDLINE | ID: mdl-23117657

OBJECTIVE: Angiotensin-II (Ang-II) promotes the interaction of mononuclear cells with arterioles and neutrophils with postcapillary venules. To investigate the mechanisms underlying this dissimilar response, the involvement of fractalkine (CX(3)CL1) was explored. METHODS AND RESULTS: Enhanced CX(3)CL1 expression was detected in both cremasteric arterioles and postcapillary venules 24 hours after Ang-II intrascrotal injection. Arteriolar leukocyte adhesion was the unique parameter significantly reduced (83%) in animals lacking CX(3)CL1 receptor (CX(3)CR1). Human umbilical arterial and venous endothelial cell stimulation with 1 µmol/L Ang-II increased CX(3)CL1 expression, yet neutralization of CX(3)CL1 activity only significantly inhibited Ang-II-induced mononuclear cell-human umbilical arterial endothelial cell interactions (73%) but not with human umbilical venous endothelial cells. The use of small interfering RNA revealed the involvement of tumor necrosis factor-α in Ang-II-induced CX(3)CL1 upregulation and mononuclear cell arrest. Nox5 knockdown with small interfering RNA or pharmacological inhibition of extracellular signal-regulated kinases1/2, p38 mitogen-activated protein kinase, and nuclear factor-κB also abolished these responses. Finally, when human umbilical arterial endothelial cells were costimulated with Ang-II, tumor necrosis factor-α, and interferon-γ, CX(3)CL1 expression and mononuclear cell adhesiveness were more pronounced than when each stimulus was provided alone. CONCLUSIONS: These results suggest that Ang-II induces functional CX(3)CL1 expression in arterial but not in venous endothelia. Thus, targeting endothelial CX(3)CL1-mononuclear leukocyte CX(3)CR1 interactions may constitute a new therapeutic strategy in the treatment of Ang-II-associated cardiovascular diseases.


Angiotensin II/metabolism , Arteries/metabolism , Chemokine CX3CL1/metabolism , Endothelial Cells/metabolism , Veins/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Arteries/drug effects , Arterioles/drug effects , Arterioles/metabolism , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/prevention & control , CX3C Chemokine Receptor 1 , Cell Adhesion , Cells, Cultured , Chemokine CX3CL1/genetics , Disease Models, Animal , Endothelial Cells/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Interferon-gamma/metabolism , Leukocyte Rolling , Leukocytes/metabolism , Losartan/pharmacology , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 2 , NADPH Oxidase 4 , NADPH Oxidase 5 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , NF-kappa B/metabolism , RNA Interference , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Signal Transduction , Time Factors , Transfection , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Veins/drug effects , Venules/drug effects , Venules/metabolism
18.
Thorax ; 68(2): 177-86, 2013 Feb.
Article En | MEDLINE | ID: mdl-23143793

BACKGROUND: Cigarette smoking is an important risk factor for the development of cardiovascular disease, yet the pathways through which this may operate are poorly understood. Therefore, the mechanism underlying cigarette smoke (CS)-induced arterial endothelial dysfunction and the potential link with fractalkine/CX(3)CL1 upregulation were investigated. METHODS AND RESULTS: Stimulation of human arterial umbilical endothelial cells (HUAECs) with pathophysiological concentrations of CS extract (1% CSE) increased CX(3)CL1 expression. Neutralisation of CX(3)CL1 activity under dynamic flow conditions significantly inhibited CSE-induced mononuclear cell adhesion to HUAECs (67%). The use of small interfering RNA (siRNA) revealed that nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 5 (Nox5) but not Nox2 or Nox4 is the main NADPH isoform involved in CSE-induced CX(3)CL1 upregulation and mononuclear cell arrest. Knock down of HUAEC tumour necrosis factor α expression with siRNA or pharmacological inhibition of p38 mitogen-activated protein kinase and nuclear factor κB also abolished these responses. Interestingly, circulating monocytes and lymphocytes from patients with chronic obstructive pulmonary disease (COPD) (n=29) versus age-matched controls (n=23) showed CX(3)CR1overexpression. Furthermore, CX(3)CL1 neutralisation dramatically diminished their enhanced adhesiveness to CSE-stimulated HUAECs. Finally, when animals were exposed for 3 days to CS, a mild inflammatory response in the lung was observed which was accompanied by enhanced CX(3)CL1 expression in the cremasteric arterioles, an organ distant from the lung. CS exposure resulted in increased leukocyte-arteriolar endothelial cell adhesion which was significantly reduced (51%) in animals lacking CX(3)CL1 receptor (CX(3)CR1). CONCLUSIONS: These results suggest that CS induces functional CX(3)CL1 expression in arterial endothelium and leukocytes from patients with COPD show increased CX(3)CL1-dependent adhesiveness. Therefore, targeting the CX(3)CL1/CX(3)CR1 axis might prevent COPD-associated cardiovascular disorders.


Chemokine CX3CL1/physiology , Endothelium, Vascular/cytology , Smoking/physiopathology , Animals , Cell Adhesion , Endothelium, Vascular/physiology , Humans , Membrane Proteins/physiology , Mice , Microcirculation/physiology , NADPH Oxidase 5 , NADPH Oxidases/physiology , RNA, Small Interfering/physiology , Up-Regulation/physiology
19.
Eur Respir J ; 41(6): 1264-74, 2013 Jun.
Article En | MEDLINE | ID: mdl-23018909

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


Fibroblasts/cytology , Gene Expression Regulation , Myofibroblasts/cytology , Smoking/adverse effects , Tropanes/pharmacology , Actins/metabolism , Bronchi/drug effects , Cells, Cultured , Cholinergic Antagonists/pharmacology , Collagen Type I/metabolism , Cyclic AMP/metabolism , Fibroblasts/drug effects , Fibrosis , Fluoresceins/pharmacology , Humans , Inflammation , Lung/cytology , Lung/drug effects , Microscopy, Fluorescence , Myofibroblasts/drug effects , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Smoke , Time Factors
20.
J Immunol ; 189(1): 411-24, 2012 Jul 01.
Article En | MEDLINE | ID: mdl-22661092

Mononuclear cell migration into the vascular subendothelium constitutes an early event of the atherogenic process. Because the effect of retinoid X receptor (RXR)α on arterial mononuclear leukocyte recruitment is poorly understood, this study investigated whether RXR agonists can affect this response and the underlying mechanisms involved. Decreased RXRα expression was detected after 4 h stimulation of human umbilical arterial endothelial cells with TNF-α. Interestingly, under physiological flow conditions, TNF-α-induced endothelial adhesion of human mononuclear cells was concentration-dependently inhibited by preincubation of the human umbilical arterial endothelial cells with RXR agonists such as bexarotene or 9-cis-retinoid acid. RXR agonists also prevented TNF-α-induced VCAM-1 and ICAM-1 expression, as well as endothelial growth-related oncogene-α and MCP-1 release. Suppression of RXRα expression with a small interfering RNA abrogated these responses. Furthermore, inhibition of MAPKs and NF-κB pathways were involved in these events. RXR agonist-induced antileukocyte adhesive effects seemed to be mediated via RXRα/peroxisome proliferator-activated receptor (PPAR)γ interaction, since endothelial PPARγ silencing abolished their inhibitory responses. Furthermore, RXR agonists increased RXR/PPARγ interaction, and combinations of suboptimal concentrations of both nuclear receptor ligands inhibited TNF-α-induced mononuclear leukocyte arrest by 60-65%. In vivo, bexarotene dose-dependently inhibited TNF-α-induced leukocyte adhesion to the murine cremasteric arterioles and decreased VCAM-1 and ICAM-1 expression. Therefore, these results reveal that RXR agonists can inhibit the initial inflammatory response that precedes the atherogenic process by targeting different steps of the mononuclear recruitment cascade. Thus, RXR agonists may constitute a new therapeutic tool in the control of the inflammatory process associated with cardiovascular disease.


Cell Migration Inhibition/immunology , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/pathology , PPAR gamma/metabolism , Retinoid X Receptor alpha/agonists , Bexarotene , Cell Communication/drug effects , Cell Communication/immunology , Cell Line , Cell Migration Inhibition/drug effects , Down-Regulation/drug effects , Down-Regulation/immunology , Endothelium, Vascular/metabolism , Humans , Intercellular Adhesion Molecule-1/blood , Leukocytes, Mononuclear/drug effects , Microcirculation/drug effects , Microcirculation/immunology , Muscle, Skeletal/blood supply , Muscle, Skeletal/drug effects , Muscle, Skeletal/immunology , PPAR gamma/physiology , Retinoid X Receptor alpha/biosynthesis , Retinoid X Receptor alpha/physiology , Tetrahydronaphthalenes/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/physiology , Umbilical Arteries/drug effects , Umbilical Arteries/immunology , Umbilical Arteries/pathology , Vascular Cell Adhesion Molecule-1/blood
...