Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Int J Immunogenet ; 49(4): 260-270, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35862251

ABSTRACT

Despite strong genetic implications of NLRP3 inflammasome, its examination as genetic determinant of ischaemic stroke (IS) remains to be done in Punjab, which has been investigated in this study. In this case control study, 400 subjects (200 IS patients, 200 stroke free controls) were included. Contributions of 5 single nucleotide polymorphisms (SNPs) including a functional SNP within NLRP3 gene (rs10754558, rs4612666, rs2027432, rs3738488 and rs1539019) for the risk of IS were investigated through genetic models after correcting the effect of significant variables. Plasma levels of three pro-inflammatory markers, that is, C-reactive protein (CRP), interleukin-1beta (IL-1ß) and interleukin-18 (IL-18) were measured by enzyme-linked immunosorbent assays (ELISA). Minor alleles of 3 out of 5 SNPs (rs10754558, rs4612666 and rs1539019) exhibited association with IS risk in additive, recessive and multiplicative models. Multivariable regression analysis confirmed that higher levels of systolic blood pressure (ß ± SE: 1.42 ± 0.57, p = .013), CRP (ß ± SE: 1.22 ± 0.41, p = .003), IL-1ß (ß ± SE: 1.78 ± 0.88, p = .043) and IL-18 (ß ± SE: 1.13 ± 0.49, p = .021) were independent risk predictors for IS. Haplotype analysis revealed a susceptibility putative haplotype GTGTA, which approximately doubled the IS risk (OR: 1.98, 95% CI: 1.12-3.78, p = .04) in dominant mode after adjusting the effect with confounding variables. This susceptibility putative haplotype GTGTA was significantly associated with increased concentrations of CRP (ß = 1.21, p = .014) and IL-1ß (ß = 1.53, p = .034) in dose-dependent manner (less in carriers of 1 copy than those who had 2 copies of GTGTA). The present study has revealed a susceptibility putative haplotype GTGTA within NLRP3 gene, carriers of which have double the risk of IS by having increased plasma levels of CRP and IL-1ß in a dose-dependent manner.


Subject(s)
Brain Ischemia , Ischemic Stroke , Stroke , Brain Ischemia/genetics , Case-Control Studies , Genetic Predisposition to Disease , Haplotypes , Humans , Inflammasomes/genetics , Interleukin-18/genetics , Interleukin-1beta/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Polymorphism, Single Nucleotide , Stroke/genetics
2.
Cytokine ; 113: 68-73, 2019 01.
Article in English | MEDLINE | ID: mdl-29934047

ABSTRACT

BACKGROUND: Lymphocytes play a key role in asthma pathophysiology, secreting various cytokines involved in chronic inflammation. CHF6001 is a highly potent and selective phosphodiesterase type 4 (PDE4) inhibitor designed for inhaled administration and has been shown to reduce the late asthmatic response. However, the effect of PDE4 inhibition on the different cytokines produced by lung lymphocytes from asthma patients has not been examined. METHODS: This study investigated the anti-inflammatory effects of CHF6001 and the corticosteroid, 17-BMP, on T-cell receptor (TCR) stimulated Th1, Th2 and Th17 cytokine release from bronchoalveolar lavage (BAL) cells from mild (n = 12) and moderate asthma (n = 12) patients. RESULTS: CHF6001 inhibited IFNγ, IL-2 and IL-17, but not IL-13, secretion from both mild and moderate asthma patient BAL cells; there was a greater effect on IFNγ and IL-2 than IL-17. The corticosteroid inhibited all four cytokines from both patient groups, but was less effective in cells from more severe patients. CHF6001 had a greater inhibitory effect on IFNγ and IL-2 than 17-BMP. CONCLUSION: The PDE4 inhibitor CHF6001 had a greater effect on Th1 cytokines from TCR-stimulated BAL cells than corticosteroid. This pharmacological effect suggests the therapeutic potential for PDE4 inhibitors to be used in the subset of more severe asthma patients with increased airway levels of IFNγ.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Asthma/drug therapy , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Lung/drug effects , Phosphodiesterase 4 Inhibitors/therapeutic use , Sulfonamides/therapeutic use , para-Aminobenzoates/therapeutic use , Adult , Asthma/metabolism , Bronchoalveolar Lavage/methods , Bronchoalveolar Lavage Fluid , Cell Line , Cytokines/metabolism , Female , Humans , Lung/metabolism , Lymphocytes/drug effects , Lymphocytes/metabolism , Male , Middle Aged , Receptors, Antigen, T-Cell/metabolism
3.
Respir Res ; 17: 9, 2016 Jan 25.
Article in English | MEDLINE | ID: mdl-26809346

ABSTRACT

BACKGROUND: CD8 lymphocytes play an important role in the pathogenesis of COPD. Corticosteroids and phosphodiesterase 4 (PDE4) inhibitors are anti-inflammatory drugs used for COPD treatment. Little is known of the combined effect of these drugs on COPD CD8 cells. We studied the effect of corticosteroid combined with PDE4 inhibitors on cytokine release form circulating and pulmonary CD8 cells, and on glucocorticoid (GR) nuclear translocation. METHODS: The effect of dexamethasone alone and in combination with the PDE4 inhibitors roflumilast and GSK256066 on cytokine release from circulating and pulmonary CD8 cells was measured. The effect of the compounds on nuclear translocation of GR and cyclic AMP-responsive element-binding protein (CREB) was studied using immunofluorescence. RESULTS: Dexamethasone inhibited cytokine release from COPD CD8 cells in a concentration dependent manner. PDE4 inhibitors enhanced this anti-inflammatory effect in an additive manner. PDE4 inhibitors did not increase corticosteroid induced GR nuclear translocation. PDE4 inhibitors, but not corticosteroid, increased phospho-CREB nuclear translocation. CONCLUSION: The combination of corticosteroids and PDE4 inhibitors results in an additive anti-inflammatory effect in COPD CD8 cells. This enhanced anti-inflammatory effect could translate to important clinical benefits for patients with COPD.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cytokines/immunology , Phosphodiesterase 4 Inhibitors/administration & dosage , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/pathology , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/immunology , Adult , Aged , Anti-Inflammatory Agents/administration & dosage , Cells, Cultured , Dose-Response Relationship, Drug , Drug Synergism , Drug Therapy, Combination/methods , Glucocorticoids/immunology , Humans , Middle Aged , Pulmonary Disease, Chronic Obstructive/drug therapy , Treatment Outcome
4.
Pulm Pharmacol Ther ; 35: 67-74, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26344428

ABSTRACT

Asthma is increasing globally and current treatments only manage a proportion of patients. There is an urgent need to develop new therapies. Lymphocytes are thought to play a central role in the pathophysiology of asthma through the production of inflammatory mediators. This is thought to be via the transcription factor NFAT which in turn can be activated through Ca(2+) release-activated Ca(2+) (CRAC) channels. The aim of this work was to investigate the role of CRAC in clinical and pre-clinical models of allergic asthma. Initial data demonstrated that the NFAT pathway is increased in stimulated lymphocytes from asthmatics. To confirm a role for the channel we showed that a selective inhibitor, Synta 66, blocked mediator production from lymphocytes. Synta 66 inhibited CD2/3/28 induced IL-2, IL-7, IL-13 & IFNΥ in a concentration-dependent manner in healthy and severe asthma donors, with over 60% inhibition observed for all cytokines. NFAT pathway was also increased in a pre-clinical asthma model. In this model we have demonstrated that CRAC played a central role in the airway inflammation and late asthmatic response (LAR). In conclusion, our data provides evidence that suggests targeting CRAC channels could be of therapeutic benefit for asthma sufferers.


Subject(s)
Asthma/metabolism , Calcium Channels/metabolism , Adult , Allergens/toxicity , Animals , Asthma/pathology , Bronchoalveolar Lavage Fluid/cytology , Calcium Channels/drug effects , Cells, Cultured , Cytokines/biosynthesis , Dose-Response Relationship, Drug , Female , Humans , In Vitro Techniques , Lung/pathology , Lymphocytes/drug effects , Lymphocytes/metabolism , Male , Middle Aged , NFATC Transcription Factors/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Rats
5.
J Pharmacol Exp Ther ; 348(1): 12-24, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24163441

ABSTRACT

In asthma and chronic obstructive pulmonary disease (COPD) multiple mediators act on Gαq-linked G-protein-coupled receptors (GPCRs) to cause bronchoconstriction. However, acting on the airway epithelium, such mediators may also elicit inflammatory responses. In human bronchial epithelial BEAS-2B cells (bronchial epithelium + adenovirus 12-SV40 hybrid), regulator of G-protein signaling (RGS) 2 mRNA and protein were synergistically induced in response to combinations of long-acting ß2-adrenoceptor agonist (LABA) (salmeterol, formoterol) plus glucocorticoid (dexamethasone, fluticasone propionate, budesonide). Equivalent responses occurred in primary human bronchial epithelial cells. Concentrations of glucocorticoid plus LABA required to induce RGS2 expression in BEAS-2B cells were consistent with the levels achieved therapeutically in the lungs. As RGS2 is a GTPase-activating protein that switches off Gαq, intracellular free calcium ([Ca(2+)]i) flux was used as a surrogate of responses induced by histamine, methacholine, and the thromboxane receptor agonist U46619 [(Z)-7-[(1S,4R,5R,6S)-5-[(E,3S)-3-hydroxyoct-1-enyl]-3-oxabicyclo[2.2.1]heptan-6-yl]hept-5-enoic acid]. This was significantly attenuated by salmeterol plus dexamethasone pretreatment, or RGS2 overexpression, and the protective effect of salmeterol plus dexamethasone was abolished by RGS2 RNA silencing. Although methacholine and U46619 induced interleukin-8 (IL-8) release and this was inhibited by RGS2 overexpression, the repression of U46619-induced IL-8 release by salmeterol plus dexamethasone was unaffected by RGS2 knockdown. Given a role for Gαq-mediated pathways in inducing IL-8 release, we propose that RGS2 acts redundantly with other effector processes to repress IL-8 expression. Thus, RGS2 expression is a novel effector mechanism in the airway epithelium that is induced by glucocorticoid/LABA combinations. This could contribute to the efficacy of glucocorticoid/LABA combinations in asthma and COPD.


Subject(s)
Adrenergic beta-2 Receptor Agonists/administration & dosage , Epithelial Cells/metabolism , Gene Expression Regulation/drug effects , Glucocorticoids/administration & dosage , RGS Proteins/genetics , Respiratory Mucosa/metabolism , Drug Combinations , Epithelial Cells/drug effects , Humans , RGS Proteins/biosynthesis , RGS Proteins/physiology , Respiratory Mucosa/cytology , Respiratory Mucosa/drug effects , Time Factors
6.
Clin Sci (Lond) ; 126(3): 223-32, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23905758

ABSTRACT

There are increased numbers of pulmonary CD8 lymphocytes in COPD (chronic obstructive pulmonary disease). CRAC (calcium release-activation calcium) channels play a central role in lymphocyte activation though the regulation of the transcription factor NFAT (nuclear factor of activated T-cells). We studied the expression of NFAT in lungs from COPD patients compared with controls, and evaluated the effects of CRAC channel inhibition compared with corticosteroids on NFAT activation and cytokine production in CD8 cells from COPD patients. The effects of the corticosteroid dexamethasone, the calcineurin inhibitor cyclosporin and the CRAC channel inhibitor Synta 66 were studied on cytokine production and NFAT activation using peripheral blood and isolated pulmonary CD8 cells. NFAT1 and CD8 co-expression in the lungs was compared in COPD patients and controls using combined immunohistochemistry and immunofluorescence. NFAT inhibition with either cyclosporin or Synta 66 resulted in significantly greater maximal inhibition of cytokines than dexamethasone in both peripheral blood and pulmonary CD8 cells [e.g. >95% inhibition of IFNγ (interferon γ) production from pulmonary CD8 cells using cyclosporin and Synta 66 compared with <50% using dexamethasone]. The absolute number of pulmonary CD8 cells co-expressing NFAT1 was significantly raised in lungs from COPD patients compared with controls, but the percentage of CD8 cells co-expressing NFAT1 was similar between COPD patients and controls (80.7% compared with 78.5% respectively, P=0.3). Inhibition of NFAT using the CRAC channel Synta 66 produces greater anti-inflammatory effects on CD8 cells from COPD patients than corticosteroids. NFAT is expressed at a high level in pulmonary CD8 cells in COPD.


Subject(s)
Anti-Inflammatory Agents/pharmacology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/pathology , Calcium Channel Blockers/pharmacology , NFATC Transcription Factors/antagonists & inhibitors , Pulmonary Disease, Chronic Obstructive/drug therapy , Aged , Aged, 80 and over , Calcium Channel Blockers/therapeutic use , Case-Control Studies , Cells, Cultured , Cyclosporine/pharmacology , Cyclosporine/therapeutic use , Cytokines/metabolism , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Drug Evaluation, Preclinical , Female , Glucocorticoids/pharmacology , Glucocorticoids/therapeutic use , Humans , Male , Middle Aged , NFATC Transcription Factors/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology
7.
Eur Respir J ; 42(1): 28-41, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23060629

ABSTRACT

The p38 mitogen-activated protein kinase (MAPK) pathway is upregulated in chronic obstructive pulmonary disease (COPD). To date, dual labelling to identify cell-type-specific presence of phosphorylated (phospho-)p38 MAPK has not been carried out. Phospho-p38 MAPK was quantified in a variety of cell types in the lung tissue of 20 COPD patients, 12 smokers and 12 nonsmokers using immunohistochemistry. Paired blood and sputum neutrophils (from seven subjects with COPD), and CD8 and epithelial cells (from three subjects with COPD) were cultured with a p38 MAPK inhibitor. Supernatant tumour necrosis factor-α and CXCL8 levels were analysed by ELISA. Sputum and blood neutrophil cytospins were analysed for phospho-p38 MAPK. Phospho-p38 MAPK was increased in bronchial epithelial cells, macrophages and CD20+ and CD8+ lymphocytes in COPD lungs. Sputum and lung tissue neutrophils were devoid of phospho-p38 in all patient groups. The p38 MAPK inhibitor SB100 attenuated pro-inflammatory mediator release in COPD lung CD8 cells and airway epithelia, but there was no effect on COPD sputum neutrophils. Our data indicate cell-specific anti-inflammatory effects of p38 MAPK inhibition in the lung.


Subject(s)
Gene Expression Regulation, Enzymologic , Pulmonary Disease, Chronic Obstructive/enzymology , Pulmonary Disease, Chronic Obstructive/physiopathology , p38 Mitogen-Activated Protein Kinases/metabolism , Aged , CD8-Positive T-Lymphocytes/cytology , Cells, Cultured/cytology , Cytokines/metabolism , Epithelial Cells/enzymology , Female , Humans , Immunohistochemistry , Inflammation , Macrophages/enzymology , Male , Middle Aged , Neutrophils/enzymology , Neutrophils/metabolism , Phosphorylation , Smoking
8.
J Pharmacol Exp Ther ; 347(1): 173-80, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23912333

ABSTRACT

Alveolar macrophages produce neutrophil chemoattractants; this cellular cross-talk contributes to neutrophilic airway inflammation in chronic obstructive pulmonary disease (COPD). We have investigated the chemotaxis cross-talk mechanisms between these cells using COPD alveolar macrophages. Using conditioned media from stimulated COPD alveolar macrophages, we investigated the relative contributions of growth-related oncogene (CXCL1), interleukin-8 (CXCL8), and regulated on activation normal T cell expressed and secreted (CCL5) to neutrophil chemotaxis and evaluated the effect of blocking the chemokine receptors CXCR1 and CXCR2 on chemotaxis caused by macrophage-conditioned media. Furthermore, we evaluated whether corticosteroid treatment of stimulated alveolar macrophages inhibited the chemotaxis ability of conditioned media. Alveolar macrophages isolated from COPD (n = 8) and smoker (S) (n = 8) lungs were treated with ultra-pure lipopolysaccharide in the presence and absence of dexamethasone (1 µM). Supernatants were used for neutrophil chemotaxis assays. SB656933 (2-hydroxy-N,N-dimethyl-3-{2-[[(R)-1-(5-methyl-furan-2-yl)-propyl]amino]-3,4-dioxo-cyclobut-1-enylamino}-benzamide) (CXCR2 antagonist) and Sch527123 [1-(2-chloro-3-fluorophenyl)-3-(4-chloro-2-hydroxy-3-piperazin-1-ylsulfonylphenyl)urea, 3-(2-chloro-3-fluoro-phenyl)-1-(4-chloro-2-hydroxy-3-piperazin-1-ylsulfonyl-phenyl)urea] (dual CXCR1 and CXCR2 antagonist) and blocking antibodies for CXCL8, CXCL1, and CCL5 were assessed. Conditioned media caused neutrophil chemotaxis in COPD and smokers (60.5 and 79.9% of total cells, respectively). Dexamethasone did not significantly reduce neutrophil chemotaxis in COPD or S. SB656933 and Sch527123 inhibited chemotaxis in a concentration-dependent manner, with the dual antagonist Sch527123 causing greater inhibition of chemotaxis. CXCL8 antibody inhibited neutrophil chemotaxis to basal levels, although there was no significant effect of blocking either CXCL1 or CCL5 (P > 0.05). CXCL8 plays a major role in neutrophil chemotaxis caused by alveolar macrophage-derived conditioned media, and this is most effectively inhibited by dual antagonism of CXCR1 and CXCR2. Corticosteroids do not inhibit chemotaxis caused by macrophage-derived chemokines.


Subject(s)
Interleukin-8/physiology , Macrophages, Alveolar/physiology , Neutrophils/physiology , Pulmonary Disease, Chronic Obstructive/metabolism , Receptors, Interleukin-8A/physiology , Receptors, Interleukin-8B/physiology , Aged , Cells, Cultured , Chemotaxis/drug effects , Chemotaxis/physiology , Female , Humans , Interleukin-8/antagonists & inhibitors , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/pathology , Male , Middle Aged , Neutrophils/drug effects , Neutrophils/pathology , Phenylurea Compounds/pharmacology , Pulmonary Disease, Chronic Obstructive/pathology , Receptors, Interleukin-8A/antagonists & inhibitors , Receptors, Interleukin-8B/antagonists & inhibitors , Sulfonamides/pharmacology
9.
Vaccines (Basel) ; 11(2)2023 Jan 25.
Article in English | MEDLINE | ID: mdl-36851139

ABSTRACT

Atherosclerosis is the formation of plaque within arteries due to overt assemblage of fats, cholesterol and fibrous material causing a blockage of the free flow of blood leading to ischemia. It is harshly impinging on health statistics worldwide because of being principal cause of high morbidity and mortality for several diseases including rheumatological, heart and brain disorders. Atherosclerosis is perpetuated by pro-inflammatory and exacerbated by pro-coagulatory mediators. Besides several other pathways, the formation of neutrophil extracellular traps (NETs) and the activation of the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome contribute significantly to the initiation and propagation of atherosclerotic plaque for its worst outcomes. The present review highlights the contribution of these two disturbing processes in atherosclerosis, inflammation and atherothrombosis in their individual as well as collaborative manner.

10.
Am J Physiol Lung Cell Mol Physiol ; 303(10): L929-38, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22983351

ABSTRACT

Interleukin-8 (IL-8/CXCL8) is an important neutrophil chemoattractant known to be elevated in the airways of cigarette smokers and in patients with chronic obstructive pulmonary disease (COPD). We examined the acute effect of aqueous cigarette smoke extract (CSE) on IL-8 expression in primary human pulmonary cells, in particular in normal human bronchial smooth muscle cells (HBSMCs). IL-8 mRNA levels increased upon CSE exposure in a concentration- and time-dependent manner, and such an effect was accompanied by IL-8 secretion. CSE-evoked elevation of IL-8 mRNA was mimicked by its component acrolein. Both CSE and acrolein induced p38 mitogen-activated protein kinase (MAPK) phosphorylation, accompanied by the phosphorylation of MAPK-activated kinase 2 (MK2), a known downstream substrate of the p38 MAPK, both in HBSMCs and in human airway epithelial cells. Furthermore, pharmacological inhibition of p38 MAPK or MK2 strongly accelerated the decay of IL-8 mRNA levels upon stimulation with CSE or acrolein and subsequent blockade of mRNA neosynthesis with actinomycin D in pulmonary structural cells (HBSMCs and airways epithelial cells) as well as in human alveolar macrophages. Conversely, pharmacological inhibition of ERK1/2 signaling inhibited CSE-induced steady-state levels of IL-8 mRNA without affecting mRNA stability, thus suggesting inhibition at the transcriptional level. In sum, p38 MAPK/MK2 signaling is an important posttranscriptional mechanism underlying upregulation of IL-8 mRNA levels elicited by CSE and acrolein. Given the pivotal role of IL-8 in neutrophil chemotaxis and activation, our results shed light on the mechanisms through which cigarette smoke can initiate inflammation in the lung.


Subject(s)
Acrolein/toxicity , Bronchi/metabolism , Epithelial Cells/metabolism , Interleukin-8/biosynthesis , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System/drug effects , Myocytes, Smooth Muscle/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA Stability/drug effects , RNA, Messenger/biosynthesis , Respiratory Mucosa/metabolism , Tobacco Smoke Pollution/adverse effects , p38 Mitogen-Activated Protein Kinases/metabolism , Bronchi/pathology , Cells, Cultured , Chemotaxis/drug effects , Dactinomycin/pharmacology , Epithelial Cells/pathology , Female , Humans , Male , Middle Aged , Myocytes, Smooth Muscle/pathology , Neutrophil Activation/drug effects , Neutrophils/metabolism , Neutrophils/pathology , Nucleic Acid Synthesis Inhibitors/pharmacology , Pneumonia/metabolism , Pneumonia/pathology , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Respiratory Mucosa/pathology
11.
Respir Res ; 13: 20, 2012 Mar 14.
Article in English | MEDLINE | ID: mdl-22417244

ABSTRACT

BACKGROUND: There are increased numbers of activated lymphocytes in the lungs of chronic obstructive pulmonary disease (COPD) patients. The clinical benefits of corticosteroids in COPD patients are limited. Our hypothesis is that lymphocytes play a role in this corticosteroid insensitivity. OBJECTIVES: To investigate the effects of the corticosteroid dexamethasone on lung lymphocyte cytokine production from patients with COPD compared to controls. METHODS: Cultured airway lymphocytes obtained by bronchoscopy from healthy non-smokers (HNS), smokers (S) and COPD patients were stimulated with phytohaemagglutinin (PHA) & phorbol myristate acetate (PMA), +/- dexamethasone. Supernatants were assayed for interleukin (IL)-2 and interferon (IFN)γ. Immunofluoresence was used to analyse changes in CD8 glucocorticoid receptor (GRα and GRß) expression. RESULTS: The inhibition of PHA/PMA stimulated IFNγ production by dexamethasone was reduced in COPD patients compared to HNS (p < 0.05 at concentrations from 0.1-1 µM). There was also a significant reduction (p < 0.05) in the mean inhibitory effect at 1 µM in COPD patients (54.1%) compared to smokers (72.1%), and in smokers compared to HNS (85.5%). There was a numerically reduced effect of dexamethasone on IL-2 production that did not reach statistical significance. There was no difference in GRα and GRß expression in follicular CD8 cells between COPD patients (50.9% and 30.4% respectively) and smokers (52.9% and 29.7% respectively). CONCLUSIONS: IFNγ production from COPD airway lymphocytes is corticosteroid insensitive. This phenomenon may be important in the poor clinical response often observed with corticosteroids.


Subject(s)
Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Pulmonary Disease, Chronic Obstructive/drug therapy , T-Lymphocytes/drug effects , Aged , Bronchoscopy , Cells, Cultured , Female , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/metabolism , Interleukin-2/biosynthesis , Interleukin-2/metabolism , Male , Middle Aged , Phytohemagglutinins/pharmacology , Pulmonary Disease, Chronic Obstructive/metabolism , Receptors, Glucocorticoid/analysis , Smoking , T-Lymphocytes/metabolism , Tetradecanoylphorbol Acetate/pharmacology
12.
J Pharmacol Exp Ther ; 330(2): 575-85, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19435930

ABSTRACT

The mRNA-destabilizing protein tristetraprolin (TTP) negatively regulates adenine- and uridine-rich element (ARE)-containing mRNAs. In A549 pulmonary cells, TTP mRNA and both a approximately 40- and a approximately 45-kDa phosphorylated version of TTP protein were rapidly induced in response to interleukin (IL)-1beta. Analysis with IkappaBalphaDeltaN, a dominant version of inhibitor of kappaBalpha (IkappaBalpha), as well as dominant-negative and small-molecule IkappaB kinase (IKK) inhibitors demonstrated that IL-1beta-induced TTP is nuclear factor-kappaB (NF-kappaB)-dependent. Likewise, TTP expression and formation of the approximately 45-kDa phosphorylated form of TTP are blocked by the p38 mitogen-activated protein kinase (MAPK) inhibitor 4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)1H-imidazole (SB203580). By contrast, and despite a 3- to 4-fold induction of TTP mRNA, the anti-inflammatory glucocorticoid dexamethasone only modestly induced expression of the approximately 40-kDa form of TTP. In the context of IL-1beta, dexamethasone exerted a marginal repressive effect on TTP mRNA expression and more considerably reduced TTP protein. Given a requirement for p38 MAPK in the induction of TTP by IL-1beta, this repressive effect may be explained by repression of the p38 MAPK pathway by dexamethasone. Knockdown of TTP protein by siRNA elevated IL-1beta-induced expression of granulocyte macrophage-colony-stimulating factor (GM-CSF) and IL-8, demonstrating a role for TTP in feedback control. Likewise, knockdown of TTP increased GM-CSF expression in the presence of IL-1beta plus dexamethasone, suggesting that feedback control by TTP also occurs in the context of IL-1beta plus dexamethasone. Taken together, our data demonstrate that NF-kappaB and p38 MAPK are critical to the induction of TTP by IL-1beta and that TTP induction provides feedback control of the ARE-containing genes GM-CSF and IL-8.


Subject(s)
Dexamethasone/pharmacology , Interleukin-1beta/physiology , NF-kappa B/physiology , Respiratory Mucosa/metabolism , Tristetraprolin/biosynthesis , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cattle , Cell Line, Tumor , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Respiratory Mucosa/drug effects , Tristetraprolin/genetics
13.
Cell Signal ; 20(7): 1338-48, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18436431

ABSTRACT

Since protein kinase C (PKC) isoforms are variously implicated in the activation of NF-kappaB, we have investigated the role of PKC in the activation of NF-kappaB-dependent transcription by the diacyl glycerol (DAG) mimetic, phorbol 12-myristate 13-acetate (PMA), and by tumour necrosis factor (TNF) alpha in pulmonary A549 cells. The PKC selective inhibitors, Ro31-8220, Gö6976, GF109203X and Gö6983, revealed no effect on TNFalpha-induced NF-kappaB DNA binding and a similar lack of effect on serine 32/36 phosphorylated IkappaBalpha and the loss of total IkappaBalpha indicates that activation of the core IKK-IkappaBalpha-NF-kappaB cascade by TNFalpha does not involve PKC. In contrast, differential sensitivity of an NF-kappaB-dependent reporter to Ro31-8220, Gö6976, GF109203X and Gö6983 (EC(50)s 0.46 microM, 0.34 microM, >10 microM and >10 microM respectively) suggests a role for protein kinase D in transcriptional activation by TNFalpha. Compared with TNFalpha, PMA weakly induces NF-kappaB DNA binding and this effect was not associated with serine 32/36 phosphorylation of IkappaBalpha. However, PMA-stimulated NF-kappaB DNA binding was inhibited by Ro31-8220 (10 microM), GF109203X (10 microM) and Gö6983 (10 microM), but not by Gö6976 (10 microM), suggesting a role for novel PKC isoforms. Furthermore, a lack of positive effect of calcium mobilising agents on both NF-kappaB DNA binding and on transcriptional activation argues against major roles for classical PKCs. This, combined with the ability of both GF109203X and Gö6983 to prevent enhancement of TNFalpha-induced NF-kappaB-dependent transcription by PMA, further indicates a role for novel PKCs in NF-kappaB transactivation. Finally, siRNA-mediated knockdown of PKCdelta and epsilon expression did not affect TNFalpha-induced NF-kappaB-dependent transcription. However, knockdown of PKCdelta expression significantly inhibited PMA-stimulated luciferase activity, whereas knockdown of PKCepsilon was without effect. Furthermore, combined knockdown of PKCdelta and epsilon revealed an increased inhibitory effect on PMA-stimulated NF-kappaB-dependent transcription suggesting that PMA-induced NF-kappaB-dependent transcription is driven by novel PKC isoforms, particularly PKCdelta and epsilon.


Subject(s)
NF-kappa B/metabolism , Phorbol Esters/pharmacology , Protein Kinase C-delta/metabolism , Protein Kinase C-epsilon/metabolism , Transcription, Genetic/drug effects , Calcium/metabolism , Carbazoles/pharmacology , Cell Line, Tumor , DNA/metabolism , Humans , Indoles/pharmacology , Intracellular Space/drug effects , Intracellular Space/metabolism , Isoenzymes/metabolism , Lamins/metabolism , Maleimides/pharmacology , Models, Biological , Protein Binding/drug effects , Protein Kinase C/metabolism , Protein Kinase Inhibitors , RNA, Small Interfering/metabolism , Transfection , Tumor Necrosis Factor-alpha/pharmacology
14.
Pulm Pharmacol Ther ; 22(5): 446-54, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19022391

ABSTRACT

SCOPE OF THE REVIEW: Our knowledge of the multifunctional nature of airway smooth muscle (ASM) has expanded rapidly in the last decade, but the underlying molecular mechanisms and how current therapies for obstructive airway diseases, such as asthma and chronic obstructive pulmonary disease (COPD), affect these are still being elucidated. Our current knowledge has built on the pharmacology of human ASM contraction and relaxation established prior to that and which is reviewed in detail elsewhere in this issue. The advent of methods to isolate and culture ASM cells, especially human ASM cells, has made it possible to study how they may contribute to airway remodelling through their synthetic, proliferative, and migratory capacities. Now the underlying molecular mechanisms of ASM growth factor secretion, extracellular matrix (ECM) production, proliferation and migration, as well as contraction and relaxation, are being determined. A complex network of signalling pathways leading to gene transcription in ASM cells permits this functional plasticity in healthy and diseased airways. This review is an overview of the effects of current therapies, and some of those in development, on key signalling pathways and transcription factors involved in these ASM functions.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Asthma/drug therapy , Muscle, Smooth/drug effects , Respiratory System/drug effects , Signal Transduction/drug effects , Transcriptional Activation/drug effects , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Agonists/therapeutic use , Animals , Anti-Asthmatic Agents/therapeutic use , Asthma/metabolism , Cholinergic Antagonists/pharmacology , Cholinergic Antagonists/therapeutic use , Cromolyn Sodium/pharmacology , Cromolyn Sodium/therapeutic use , Drug Therapy, Combination , Glucocorticoids/pharmacology , Glucocorticoids/therapeutic use , Humans , Leukotriene Antagonists/pharmacology , Leukotriene Antagonists/therapeutic use , Muscle, Smooth/metabolism , Muscle, Smooth/physiology , Nedocromil/pharmacology , Nedocromil/therapeutic use , Respiratory System/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism
15.
Mol Pharmacol ; 73(1): 203-14, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17901197

ABSTRACT

Addition of an inhaled long-acting beta(2)-adrenoceptor agonist (LABA) to an inhaled corticosteroid (ICS) is more effective at improving asthma control and reducing exacerbations than increasing the dose of ICS. Given that LABA monotherapy is not anti-inflammatory, pathways may exist by which LABAs enhance ICS actions. In the current study, the glucocorticoid dexamethasone had no effect on beta(2)-adrenoceptor agonist-induced cAMP-response element-dependent transcription in the human bronchial epithelial cell line BEAS-2B. In contrast, simple glucocorticoid response element (GRE)-dependent transcription induced by dexamethasone, budesonide, and fluticasone was synergistically enhanced by beta(2)-adrenoceptor agonists, including salmeterol and formoterol, to a level that could not be achieved by glucocorticoid alone. This enhancement was mimicked by other cAMP-elevating agents, and a cAMP mimetic, and was blocked by an inhibitor of cAMP-dependent protein kinase (PKA). Thus, beta(2)-adrenoceptor agonists synergistically enhance simple GRE-dependent transcription via the classical cAMP-PKA pathway. Consistent with the clinical situation, the addition of a beta(2)-adrenoceptor agonist to a glucocorticoid is steroid-sparing in that maximal GRE-dependent responses, evoked by glucocorticoid, are achieved at approximately 10-fold lower concentrations in the presence of beta(2)-adrenoceptor agonist. Finally, analysis of dexamethasone-inducible genes, including glucocorticoid-inducible leucine zipper (GILZ), aminopeptidase N, FKBP51, PAI-1, tristetraprolin, DNB5, p57KIP2, metallothionein 1X, and MKP-1, revealed enhanced inducibility of some genes by glucocorticoid/beta(2)-adrenoceptor agonist combinations in a manner that was consistent with the GRE-reporter. Because such effects also occur in primary human airway smooth muscle cells, we propose that enhancement of glucocorticoid-inducible gene expression may contribute to the superior efficacy of LABA/ICS combination therapies, over ICS alone, in asthma treatment.


Subject(s)
Adrenergic beta-2 Receptor Agonists , Adrenergic beta-Agonists/pharmacology , Bronchi/drug effects , Glucocorticoids/pharmacology , Muscle, Smooth, Vascular/drug effects , Transcription, Genetic/drug effects , Bronchi/cytology , Bronchi/metabolism , Humans , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism
16.
Article in English | MEDLINE | ID: mdl-29535517

ABSTRACT

Background: COPD is a progressive inflammatory airway disease characterized by increased numbers of alveolar macrophages in the lungs. Bacterial colonization of the lungs is a common feature in COPD and can promote inflammation through continual and repeated Toll-like receptor (TLR) stimulation. We have studied the response of COPD alveolar macrophages to repetitive stimulation with TLR2 and TLR4 ligands. We investigated the effect of sequential stimulation with different ligands to determine whether this results in tolerance or amplification of the immune response. Methods: We stimulated alveolar macrophages from COPD patients (n=9) and smokers (n=8) with the TLR4 agonist lipopolysaccharide (LPS) or the TLR2 agonist Pam3CSK4 for 24 hours before restimulating again for 24 hours. Cytokine protein release and gene expression were investigated. Results: Repetitive stimulation of COPD and smokers macrophages with LPS for both 24-hour periods caused a reduction in tumor necrosis factor α, CCL5, and IL-10 production compared to cells that were not exposed initially to LPS. IL-6 and CXCL8 production were not significantly altered following repetitive LPS stimulation. The same pattern was observed for repeated stimulation with Pam3CSK4. Using COPD macrophages, LPS followed by Pam3CSK4 stimulation increased the levels of all cytokines compared to media followed by Pam3CSK4. Conclusion: TLR tolerance in COPD alveolar macrophages occurs after repetitive stimulation with the same TLR ligand, but this only occurs for selected cytokines. CXCL8 production is not reduced after repetitive TLR stimulation with the same ligand; this may be an important mechanism for the increased CXCL8 levels that have been observed in COPD. We showed that TLR4 stimulation followed by TLR2 stimulation does not cause tolerance, but enhances cytokine production. This may be a relevant mechanism by which bacteria cause excessive inflammation in COPD patients.


Subject(s)
Lipopeptides/pharmacology , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophages, Alveolar/drug effects , Pulmonary Disease, Chronic Obstructive/metabolism , Toll-Like Receptor 2/agonists , Toll-Like Receptor 4/agonists , Aged , Case-Control Studies , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Female , Humans , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Ligands , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/immunology , Signal Transduction/drug effects , Smoking/adverse effects , Smoking/immunology , Smoking/metabolism , Time Factors , Toll-Like Receptor 2/immunology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism
17.
J Neurosci Rural Pract ; 7(4): 577-579, 2016.
Article in English | MEDLINE | ID: mdl-27695240

ABSTRACT

Morvan's syndrome is a rare autoimmune disorder characterized by triad of peripheral nerve hyperexcitability, autonomic dysfunction, and central nervous system symptoms. Antibodies against contactin-associated protein-like 2 (CASPR2), a subtype of voltage-gated potassium channel (VGKC) complex, are found in a significant proportion of patients with Morvan's syndrome and are thought to play a key role in peripheral as well as central clinical manifestations. We report a patient of Morvan's syndrome with positive CASPR2-anti-VGKC antibody having syndrome of inappropriate antidiuretic hormone as a cause of persistent hyponatremia.

18.
FEBS Lett ; 579(1): 285-91, 2005 Jan 03.
Article in English | MEDLINE | ID: mdl-15620728

ABSTRACT

We have investigated the effect of the sulfhydryl-reactive reagent, methyl thiosulfonate ethylammonium (MTSEA), on ligand binding to the human melanocortin-4 (MC4) receptor stably expressed in HEK-293 cells. MTSEA inhibited binding of the agonist, 125I-NDPalpha-MSH, and the antagonist, 125I-SHU9119, in a concentration-dependent manner. Pre-incubation of cells with either the agonist or antagonist protected from subsequent MTSEA inhibition of radioligand binding. Mutation of Cys130 in transmembrane helix 3 to alanine, whilst not affecting ligand binding, led to a complete loss of the inhibitory effect of MTSEA. Since other types of sulfhydryl-reactive reagents had no effect on ligand binding, we conclude that covalent modification of Cys130 by MTSEA disrupts ligand binding by neutralising a close-by negative charge, most likely on Asp126.


Subject(s)
Cysteine/drug effects , Ethyl Methanesulfonate/analogs & derivatives , Ethyl Methanesulfonate/pharmacology , Receptor, Melanocortin, Type 4/chemistry , Receptor, Melanocortin, Type 4/drug effects , Amino Acid Motifs/genetics , Amino Acid Sequence , Binding Sites/genetics , Cysteine/chemistry , Cysteine/genetics , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Protein Structure, Secondary/genetics , Receptor, Melanocortin, Type 4/metabolism
19.
Eur Respir Rev ; 24(137): 510-5, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26324813

ABSTRACT

Bacterial and viral infections (exacerbations) are particularly problematic in those with underlying respiratory disease, including post-viral infection, asthma, chronic obstructive pulmonary disease and pulmonary fibrosis. Patients experiencing exacerbations tend to be at the more severe end of the disease spectrum and are often difficult to treat. Most of the unmet medical need remains in this patient group. Airway macrophages are one of the first cell populations to encounter airborne pathogens and, in health, exist in a state of reduced responsiveness due to interactions with the respiratory epithelium and specific factors found in the airway lumen. Granulocyte-macrophage colony-stimulating factor, interleukin-10, transforming growth factor-ß, surfactant proteins and signalling via the CD200 receptor, for example, all raise the threshold above which airway macrophages can be activated. We highlight that following severe respiratory inflammation, the airspace microenvironment does not automatically re-set to baseline and may leave airway macrophages more restrained than they were at the outset. This excessive restraint is mediated in part by the clearance of apoptotic cells and components of extracellular matrix. This implies that one strategy to combat respiratory exacerbations would be to retune airway macrophage responsiveness to allow earlier bacterial recognition.


Subject(s)
Cellular Microenvironment , Lung/immunology , Macrophages, Alveolar/immunology , Pneumonia, Bacterial/immunology , Pneumonia, Viral/immunology , Adaptation, Physiological , Animals , Host-Pathogen Interactions , Humans , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Lung/metabolism , Lung/microbiology , Lung/pathology , Lung/virology , Macrophage Activation , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/microbiology , Macrophages, Alveolar/pathology , Macrophages, Alveolar/virology , Phenotype , Pneumonia, Bacterial/diagnosis , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/microbiology , Pneumonia, Viral/diagnosis , Pneumonia, Viral/metabolism , Pneumonia, Viral/virology , Risk Assessment , Risk Factors , Signal Transduction
20.
Mucosal Immunol ; 8(5): 1021-1030, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25603826

ABSTRACT

Much of the biology surrounding macrophage functional specificity has arisen through examining inflammation-induced polarizing signals, but this also occurs in homeostasis, requiring tissue-specific environmental triggers that influence macrophage phenotype and function. The TAM receptor family of receptor tyrosine kinases (Tyro3, Axl and MerTK) mediates the non-inflammatory removal of apoptotic cells by phagocytes through the bridging phosphatidylserine-binding molecules growth arrest-specific 6 (Gas6) or Protein S. We show that one such TAM receptor (Axl) is exclusively expressed on mouse airway macrophages, but not interstitial macrophages and other lung leukocytes, under homeostatic conditions and is constitutively ligated to Gas6. Axl expression is potently induced by granulocyte-macrophage colony-stimulating factor expressed in the healthy and inflamed airway, and by type I interferon or Toll-like receptor-3 stimulation on human and mouse macrophages, indicating potential involvement of Axl in apoptotic cell removal under inflammatory conditions. Indeed, an absence of Axl does not cause sterile inflammation in health, but leads to exaggerated lung inflammatory disease upon influenza infection. These data imply that Axl allows specific identification of airway macrophages, and that its expression is critical for macrophage functional compartmentalization in the airspaces or lung interstitium. We propose that this may be a critical feature to prevent excessive inflammation because of secondary necrosis of apoptotic cells that have not been cleared by efferocytosis.


Subject(s)
Lung/immunology , Macrophages/immunology , Orthomyxoviridae Infections/immunology , Pneumonia/immunology , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , Animals , Female , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Lung/pathology , Macrophages/pathology , Mice , Mice, Knockout , Organ Specificity , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/pathology , Pneumonia/genetics , Pneumonia/pathology , Protein S/genetics , Protein S/immunology , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Axl Receptor Tyrosine Kinase
SELECTION OF CITATIONS
SEARCH DETAIL