Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Front Physiol ; 12: 585895, 2021.
Article in English | MEDLINE | ID: mdl-33790802

ABSTRACT

Supplemental O2 (hyperoxia), necessary for maintenance of oxygenation in premature infants, contributes to neonatal and pediatric airway diseases including asthma. Airway smooth muscle (ASM) is a key resident cell type, responding to hyperoxia with increased contractility and remodeling [proliferation, extracellular matrix (ECM) production], making the mechanisms underlying hyperoxia effects on ASM significant. Recognizing that fetal lungs experience a higher extracellular Ca2+ ([Ca2+]o) environment, we previously reported that the calcium sensing receptor (CaSR) is expressed and functional in human fetal ASM (fASM). In this study, using fASM cells from 18 to 22 week human fetal lungs, we tested the hypothesis that CaSR contributes to hyperoxia effects on developing ASM. Moderate hyperoxia (50% O2) increased fASM CaSR expression. Fluorescence [Ca2+]i imaging showed hyperoxia increased [Ca2+]i responses to histamine that was more sensitive to altered [Ca2+]o, and promoted IP3 induced intracellular Ca2+ release and store-operated Ca2+ entry: effects blunted by the calcilytic NPS2143. Hyperoxia did not significantly increase mitochondrial calcium which was regulated by CaSR irrespective of oxygen levels. Separately, fASM cell proliferation and ECM deposition (collagens but not fibronectin) showed sensitivity to [Ca2+]o that was enhanced by hyperoxia, but blunted by NPS2143. Effects of hyperoxia involved p42/44 ERK via CaSR and HIF1α. These results demonstrate functional CaSR in developing ASM that contributes to hyperoxia-induced contractility and remodeling that may be relevant to perinatal airway disease.

2.
Curr Opin Pharmacol ; 51: 19-28, 2020 04.
Article in English | MEDLINE | ID: mdl-32375079

ABSTRACT

Understanding and targeting of GPCRs remain a critical aspect of airway pharmacology and therapeutics for diseases such as asthma or COPD. Most attention has been on the large Class A GPCRs towards improved bronchodilation and blunting of remodeling. Better known in the central or peripheral nervous system, there is increasing evidence that Class C GPCRs which include metabotropic glutamate and GABA receptors, the calcium sensing receptor, sweet/umami taste receptors and a number of orphan receptors, can contribute to airway structure and function. In this review, we will summarize current state of knowledge regarding the pharmacology of Class C GPCRs, their expression and potential functions in the airways, and the application of pharmacological agents targeting this group in the context of airway diseases.


Subject(s)
Lung Diseases/drug therapy , Lung Diseases/metabolism , Lung/metabolism , Receptors, G-Protein-Coupled/metabolism , Respiratory System Agents/administration & dosage , Animals , Drug Delivery Systems/trends , Humans , Lung/drug effects , Receptors, Calcium-Sensing/antagonists & inhibitors , Receptors, Calcium-Sensing/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, GABA-B/metabolism
3.
Inflamm Res ; 68(5): 369-377, 2019 May.
Article in English | MEDLINE | ID: mdl-30852628

ABSTRACT

Tristetraprolin (TTP) is an anti-inflammatory molecule known to post-transcriptionally regulate cytokine production and is, therefore, an attractive drug target for chronic respiratory diseases driven by inflammation, such as asthma and chronic obstructive pulmonary disease. Our recent in vitro studies in primary human airway smooth (ASM) cells have confirmed the essential anti-inflammatory role played by TTP as a critical partner in a cytokine regulatory network. However, several unanswered questions remain. While prior in vitro studies have suggested that TTP is regulated in a cAMP-mediated manner, raising the possibility that this may be one of the ways in which ß2-agonists achieve beneficial effects beyond bronchodilation, the impact of ß2-agonists on ASM cells is unknown. Furthermore, the effect of prostaglandin E2 (PGE2) on TTP expression in ASM cells has not been reported. We address this herein and reveal, for the first time, that TTP is not regulated by cAMP-activating agents nor following treatment with long-acting ß2-agonists. However, PGE2 does induce TTP mRNA expression and protein upregulation in ASM cells. Although the underlying mechanism of action remains undefined, we can confirm that PGE2-induced TTP upregulation is not mediated via cAMP, or EP2/EP4 receptor activation, and occurred in a manner independent of the p38 MAPK-mediated pathway. Taken together, these data confirm that ß2-agonists do not upregulate TTP in human ASM cells and indicate that another way in which PGE2 may achieve beneficial effects in asthma and COPD may be via upregulation of the master controller of inflammation-TTP.


Subject(s)
Dinoprostone/pharmacology , Myocytes, Smooth Muscle/drug effects , Tristetraprolin/biosynthesis , Adrenergic beta-2 Receptor Agonists/pharmacology , Azetidines/pharmacology , Bronchi/cytology , Cells, Cultured , Cyclic AMP/metabolism , Dual Specificity Phosphatase 1/genetics , Formoterol Fumarate/pharmacology , Humans , Isoindoles/pharmacology , Myocytes, Smooth Muscle/metabolism , RNA, Messenger/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Salmeterol Xinafoate/pharmacology , Sulfonamides/pharmacology , Tristetraprolin/genetics , Up-Regulation/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Pulm Pharmacol Ther ; 45: 34-39, 2017 08.
Article in English | MEDLINE | ID: mdl-28414142

ABSTRACT

The xanthine doxofylline has been examined in clinical trials and shown to have efficacy and greater tolerability than theophylline in asthma and chronic obstructive pulmonary disease. The 'novofylline' doxofylline has demonstrated bronchodilatory and anti-inflammatory actions in in vivo and ex vivo experimental models of respiratory disease. However, there are limited studies in vitro. We address this herein and examine whether doxofylline has anti-inflammatory impact on primary cultures of airway smooth muscle (ASM) cells. We conduct a series of investigations comparing and contrasting doxofylline with the archetypal xanthine, theophylline, and the specific phosphodiesterase (PDE) 4 inhibitor, cilomilast. We confirm that the xanthine drugs do not have action as PDE inhibitors in ASM cells. Unlike cilomilast, doxofylline (and theophylline) do not increase cAMP production in ASM cells induced by long-acting ß2-agonist formoterol. Similar to theophylline, and consistent with the lack of cAMP potentiation, doxofylline does not augment formoterol-induced upregulation of the anti-inflammatory protein mitogen-activated protein kinase phosphatase 1 (MKP-1). However, when we examine the effect of doxofylline on secretion of the interleukin 8 from ASM cells stimulated by tumour necrosis factor (an in vitro surrogate measure of inflammation), there was no repression of inflammation. This is in contrast to the anti-inflammatory impact exerted by theophylline and cilomilast in confirmatory experiments. In summary, our study is the first to examine the effect of doxofylline on ASM cells in vitro and highlights some distinct differences between two key members of xanthine drug family, doxofylline and theophylline.


Subject(s)
Formoterol Fumarate/pharmacology , Myocytes, Smooth Muscle/drug effects , Theophylline/analogs & derivatives , Theophylline/pharmacology , Anti-Inflammatory Agents/pharmacology , Bronchi/cytology , Bronchi/drug effects , Bronchodilator Agents/pharmacology , Cells, Cultured , Cyclic AMP/metabolism , Cyclohexanecarboxylic Acids/administration & dosage , Cyclohexanecarboxylic Acids/pharmacology , Dual Specificity Phosphatase 1/metabolism , Humans , Inflammation/drug therapy , Inflammation/pathology , Interleukin-8/metabolism , Myocytes, Smooth Muscle/metabolism , Nitriles/administration & dosage , Nitriles/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Tumor Necrosis Factor-alpha/metabolism
5.
Am J Respir Cell Mol Biol ; 56(4): 532-538, 2017 04.
Article in English | MEDLINE | ID: mdl-27997807

ABSTRACT

Roflumilast is an orally active phosphodiesterase 4 inhibitor approved for use in chronic obstructive pulmonary disease. Roflumilast N-oxide (RNO) is the active metabolite of roflumilast and has a demonstrated antiinflammatory impact in vivo and in vitro. To date, the effect of RNO on the synthetic function of airway smooth muscle (ASM) cells is unknown. We address this herein and investigate the effect of RNO on ß2-adrenoceptor-mediated, cAMP-dependent responses in ASM cells in vitro, and whether RNO enhances steroid-induced repression of inflammation. RNO (0.001-1,000 nM) alone had no effect on AMP production from ASM cells, and significant potentiation of the long-acting ß2-agonist formoterol-induced cAMP could only be achieved at the highest concentration of RNO tested (1,000 nM). At this concentration, RNO exerted a small, but not significantly different, potentiation of formoterol-induced expression of antiinflammatory mitogen-activated protein kinase phosphatase 1. Consequently, tumor necrosis factor-induced IL-8 secretion was unaffected by RNO in combination with formoterol. However, because there was the potential for phosphodiesterase 4 inhibitors and long-acting ß2-agonists to interact with corticosteroids to achieve superior antiinflammatory efficacy, we examined whether RNO, alone or in combination with formoterol, enhanced the antiinflammatory effect of dexamethasone by measuring the impact on IL-8 secretion. Although RNO alone did not significantly enhance the cytokine repression achieved with steroids, RNO in combination with formoterol significantly enhanced the antiinflammatory effect of dexamethasone in ASM cells. This was linked to increased mitogen-activated protein kinase phosphatase 1 expression in ASM cells, suggesting that a molecular mechanism is responsible for augmented antiinflammatory actions of combination therapeutic approaches that include RNO.


Subject(s)
Aminopyridines/pharmacology , Anti-Inflammatory Agents/pharmacology , Benzamides/pharmacology , Dexamethasone/pharmacology , Formoterol Fumarate/pharmacology , Lung/cytology , Myocytes, Smooth Muscle/metabolism , Cyclic AMP/biosynthesis , Cyclopropanes/pharmacology , Dual Specificity Phosphatase 1/genetics , Dual Specificity Phosphatase 1/metabolism , Humans , Interleukin-8/metabolism , Myocytes, Smooth Muscle/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects
6.
Sci Rep ; 6: 37297, 2016 11 16.
Article in English | MEDLINE | ID: mdl-27849062

ABSTRACT

Protein phosphatase 2A (PP2A) activity can be enhanced pharmacologically by PP2A-activating drugs (PADs). The sphingosine analog FTY720 is the best known PAD and we have shown that FTY720 represses production of pro-inflammatory cytokines responsible for respiratory disease pathogenesis. Whether its phosphorylated form, FTY720-P, also enhances PP2A activity independently of the sphingosine 1-phosphate (S1P) pathway was unknown. Herein, we show that FTY720-P enhances TNF-induced PP2A phosphatase activity and significantly represses TNF-induced interleukin 6 (IL-6) and IL-8 mRNA expression and protein secretion from A549 lung epithelial cells. Comparing FTY720 and FTY720-P with S1P, we show that unlike S1P, the sphingosine analogs do not induce cytokine production on their own. In fact, FTY720 and FTY720-P significantly repress S1P-induced IL-6 and IL-8 production. We then examined their impact on expression of cyclooxygenase 2 (COX-2) and resultant prostaglandin E2 (PGE2) production. S1P did not increase production of this pro-inflammatory enzyme because COX-2 mRNA gene expression is NF-κB-dependent, and unlike TNF, S1P did not activate NF-κB. However, TNF-induced COX-2 mRNA expression and PGE2 secretion is repressed by FTY720 and FTY720-P. Hence, FTY720-P enhances PP2A activity and that PADs can repress production of pro-inflammatory cytokines and enzymes in A549 lung epithelial cells in a manner devoid of S1P agonism.


Subject(s)
Epithelial Cells/drug effects , Inflammation/prevention & control , Organophosphates/pharmacology , Protein Phosphatase 2/metabolism , Sphingosine/analogs & derivatives , A549 Cells , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Enzyme Activation/drug effects , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inflammation/genetics , Inflammation/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Lung/pathology , Lysophospholipids/pharmacology , Sphingosine/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
7.
Int J Biochem Cell Biol ; 78: 279-287, 2016 09.
Article in English | MEDLINE | ID: mdl-27477309

ABSTRACT

Corticosteroids are effective anti-inflammatory therapies widely utilized in chronic respiratory diseases. But these medicines can lose their efficacy during respiratory infection resulting in disease exacerbation. Further in vitro research is required to understand how infection worsens lung function control in order to advance therapeutic options to treat infectious exacerbation in the future. In this study, we utilize a cellular model of bacterial exacerbation where we pretreat A549 lung epithelial cells with the synthetic bacterial lipoprotein Pam3CSK4 (a TLR2 ligand) to mimic bacterial infection and tumor necrosis factor α (TNFα) to simulate inflammation. Under these conditions, Pam3CSK4 induces corticosteroid insensitivity; demonstrated by substantially reduced ability of the corticosteroid dexamethasone to repress TNFα-induced interleukin 6 secretion. We then explored the molecular mechanism responsible and found that corticosteroid insensitivity induced by bacterial mimics was not due to altered translocation of the glucocorticoid receptor into the nucleus, nor an impact on the NF-κB pathway. Moreover, Pam3CSK4 did not affect corticosteroid-induced upregulation of anti-inflammatory MAPK deactivating phosphatase-MKP-1. However, Pam3CSK4 can induce oxidative stress and we show that a proportion of the MKP-1 produced in response to corticosteroid in the context of TLR2 ligation was rendered inactive by oxidation. Thus to combat inflammation in the context of bacterial exacerbation we sought to discover effective strategies that bypassed this road-block. We show for the first time that known (FTY720) and novel (theophylline) activators of the phosphatase PP2A can serve as non-steroidal anti-inflammatory alternatives and/or corticosteroid-sparing approaches in respiratory inflammation where corticosteroid insensitivity exists.


Subject(s)
Adrenal Cortex Hormones/pharmacology , Anti-Inflammatory Agents/pharmacology , Drug Resistance/drug effects , Lipopeptides/pharmacology , Lung/cytology , Protein Phosphatase 2/metabolism , Toll-Like Receptor 2/metabolism , Cell Line , Dual Specificity Phosphatase 1/metabolism , Enzyme Activation/drug effects , Humans , Ligands , Lipopeptides/metabolism , Oxidation-Reduction/drug effects , Protein Binding , Up-Regulation/drug effects
8.
Am J Respir Cell Mol Biol ; 54(6): 792-801, 2016 06.
Article in English | MEDLINE | ID: mdl-26574643

ABSTRACT

Theophylline is an old drug experiencing a renaissance owing to its beneficial antiinflammatory effects in chronic respiratory diseases, such as asthma and chronic obstructive pulmonary disease. Multiple modes of antiinflammatory action have been reported, including inhibition of the enzymes that degrade cAMP-phosphodiesterase (PDE). Using primary cultures of airway smooth muscle (ASM) cells, we recently revealed that PDE4 inhibitors can potentiate the antiinflammatory action of ß2-agonists by augmenting cAMP-dependent expression of the phosphatase that deactivates mitogen-activated protein kinase (MAPK)-MAPK phosphatase (MKP)-1. Therefore, the aim of this study was to address whether theophylline repressed cytokine production in a similar, PDE-dependent, MKP-1-mediated manner. Notably, theophylline did not potentiate cAMP release from ASM cells treated with the long-acting ß2-agonist formoterol. Moreover, theophylline (0.1-10 µM) did not increase formoterol-induced MKP-1 messenger RNA expression nor protein up-regulation, consistent with the lack of cAMP generation. However, theophylline (at 10 µM) was antiinflammatory and repressed secretion of the neutrophil chemoattractant cytokine IL-8, which is produced in response to TNF-α. Because theophylline's effects were independent of PDE4 inhibition or antiinflammatory MKP-1, we then wished to elucidate the novel mechanisms responsible. We investigated the impact of theophylline on protein phosphatase (PP) 2A, a master controller of multiple inflammatory signaling pathways, and show that theophylline increases TNF-α-induced PP2A activity in ASM cells. Confirmatory results were obtained in A549 lung epithelial cells. PP2A activators have beneficial effects in ex vivo and in vivo models of respiratory disease. Thus, our study is the first to link theophylline with PP2A activation as a novel mechanism to control respiratory inflammation.


Subject(s)
Enzyme Activators/pharmacology , Interleukin-8/metabolism , Lung/cytology , Myocytes, Smooth Muscle/enzymology , Phosphodiesterase Inhibitors/pharmacology , Protein Phosphatase 2/metabolism , Theophylline/pharmacology , A549 Cells , Anti-Inflammatory Agents/pharmacology , Cyclic AMP/biosynthesis , Dual Specificity Phosphatase 1/genetics , Dual Specificity Phosphatase 1/metabolism , Formoterol Fumarate/pharmacology , Gene Knockdown Techniques , Humans , Myocytes, Smooth Muscle/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects
9.
Cancer Biol Ther ; 16(5): 780-9, 2015.
Article in English | MEDLINE | ID: mdl-25774547

ABSTRACT

Mitogen activated protein kinase phosphatase-1 (MKP-1) has emerged as an important protein mediating breast cancer oncogenesis and chemoresistance to cancer chemotherapies, especially proteasome inhibitors. In this in vitro study, we utilized the breast cancer epithelial cell lines MCF-7 and MDA-MB-231, in comparison to MCF-10A control cells, to examine the impact of MKP-1 on breast cancer cell growth and repression by proteasome inhibitors. We confirm that proteasome inhibitors MG-132 and bortezomib induce MKP-1 protein upregulation and we show that one of the ways in which bortezomib increases MKP-1 in breast cancer cells, in addition to inhibition of ubiquitin-proteasome system, is via upregulation of MKP-1 mRNA expression in p38 MAPK-mediated manner. Notably, these effects are specific to cancer cells, as bortezomib activated p38 MAPK and induced MKP-1 in MCF-7 and MDA-MB-231 breast cancer cells, but not in control cells (MCF-10A). We took a dual approach toward targeting MKP-1 to show that bortezomib-induced effects are enhanced. Firstly, treatment with the non-specific MKP-1 inhibitor triptolide reduces breast cancer cell growth and augments proteasome inhibitor-induced effects. Secondly, specific knock-down of MKP-1 with siRNA significantly repressed cell viability by reduced cyclin D1 expression, and enhanced repression of cancer cell growth by proteasome inhibitors. Taken together, these results indicate that removing the unwanted (MKP-1-inducing) effects of bortezomib significantly improves the efficacy of proteasome inhibition in breast cancer cells. Thus, future development of drugs targeting MKP-1 offer promise of combination therapies with reduced toxicity and enhanced cell death in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Cell Cycle Proteins/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Phosphoprotein Phosphatases/genetics , Cell Line, Tumor , Cell Survival , Female , Humans , Transfection
10.
Am J Respir Cell Mol Biol ; 52(5): 634-40, 2015 May.
Article in English | MEDLINE | ID: mdl-25296132

ABSTRACT

ß2-agonists are principally used in asthma to provide bronchodilation; however, they also have antiinflammatory properties, due, in part, to their ability to up-regulate mitogen-activated protein kinase phosphatase (MKP) 1 in a cAMP-dependent manner. Phosphodiesterases (PDEs) are attractive targets for potentiating the antiinflammatory response. There are 11 subfamilies of PDE enzymes; among these, inhibition of PDE3 and PDE4 are the main targets for airway smooth muscle (ASM). PDE enzymes are important intracellular regulators that catalyze the breakdown of cyclic adenosine monophosphate (cAMP) and/or 3',5'-cyclic guanosine monophosphate to their inactive forms. Given that MKP-1 is cAMP dependent, and inhibition of PDE acts to increase ß2-agonist-induced cAMP, it is possible that the presence of PDE inhibitors may enhance ß2-adrenoceptor-mediated responses. We address this herein by comparing the ability of a panel of inhibitors against PDE3 (cilostamide, cilostazol, milrinone) or PDE4 (cilomilast, piclamilast, rolipram) to increase cAMP, MKP-1 mRNA expression, and protein up-regulation in ASM cells induced in response to the ß2-agonist formoterol. Our data show that inhibitors of PDE4, but not PDE3, increase ß2-agonist-induced cAMP and induce MKP-1 mRNA expression and protein up-regulation. When cAMP was increased, there was a concomitant increase in MKP-1 levels and significant inhibition of TNF-α-induced CXCL8 (IL-8). This result was consistent with all PDE4 inhibitors examined but not for the PDE3 inhibitors. These findings reinforce cAMP-dependent control of MKP-1 expression, and suggest that PDE4 is the predominant PDE isoform responsible for formoterol-induced cAMP breakdown in ASM cells. Our study is the first to demonstrate that PDE4 inhibitors augment antiinflammatory effects of ß2-agonists via increased MKP-1 expression in ASM cells.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Anti-Inflammatory Agents/pharmacology , Bronchi/drug effects , Dual Specificity Phosphatase 1/metabolism , Ethanolamines/pharmacology , Muscle, Smooth/drug effects , Myocytes, Smooth Muscle/drug effects , Phosphodiesterase 3 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/pharmacology , Adenylyl Cyclases/metabolism , Bronchi/enzymology , Bronchi/immunology , Cells, Cultured , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Dual Specificity Phosphatase 1/genetics , Formoterol Fumarate , Humans , Interleukin-8/metabolism , Muscle, Smooth/enzymology , Muscle, Smooth/immunology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/immunology , RNA, Messenger/metabolism , Second Messenger Systems/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation
11.
PLoS One ; 8(3): e59635, 2013.
Article in English | MEDLINE | ID: mdl-23533638

ABSTRACT

Mitogen-activated protein kinase phosphatase 1 (MKP-1) represses MAPK-driven signalling and plays an important anti-inflammatory role in asthma and airway remodelling. Although MKP-1 is corticosteroid-responsive and increased by cAMP-mediated signalling, the upregulation of this critical anti-inflammatory protein by long-acting ß2-agonists and clinically-used corticosteroids has been incompletely examined to date. To address this, we investigated MKP-1 gene expression and protein upregulation induced by two long-acting ß2-agonists (salmeterol and formoterol), alone or in combination with the corticosteroid fluticasone propionate (abbreviated as fluticasone) in primary human airway smooth muscle (ASM) cells in vitro. ß2-agonists increased MKP-1 protein in a rapid but transient manner, while fluticasone induced sustained upregulation. Together, long-acting ß2-agonists increased fluticasone-induced MKP-1 and modulated ASM synthetic function (measured by interleukin 6 (IL-6) and interleukin 8 (IL-8) secretion). As IL-6 expression (like MKP-1) is cAMP/adenylate cyclase-mediated, the long-acting ß2-agonist formoterol increased IL-6 mRNA expression and secretion. Nevertheless, when added in combination with fluticasone, ß2-agonists significantly repressed IL-6 secretion induced by tumour necrosis factor α (TNFα). Conversely, as IL-8 is not cAMP-responsive, ß2-agonists significantly inhibited TNFα-induced IL-8 in combination with fluticasone, where fluticasone alone was without repressive effect. In summary, long-acting ß2-agonists increase fluticasone-induced MKP-1 in ASM cells and repress synthetic function of this immunomodulatory airway cell type.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Androstadienes/pharmacology , Dual Specificity Phosphatase 1/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , Blotting, Western , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Fluticasone , Humans , Real-Time Polymerase Chain Reaction
12.
Biochim Biophys Acta ; 1823(10): 1658-65, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22743041

ABSTRACT

Sphingosine 1-phosphate (S1P), a bioactive sphingolipid elevated in asthmatic airways, is increasingly recognized as playing an important role in respiratory disease. S1P activates receptor-mediated signaling to modulate diverse cellular functions and promote airway inflammation. Although many of the stimulatory pathways activated by S1P have been delineated, especially mitogen-activated protein kinases (MAPK), the question of whether S1P exerts negative feedback control on its own signaling cascade via upregulation of phosphatases remains unexplored. We show that S1P rapidly and robustly upregulates mRNA and protein expression of the MAPK deactivator-MAPK phosphatase 1 (MKP-1). Utilizing the pivotal airway structural cell, airway smooth muscle (ASM), we confirm that S1P activates all members of the MAPK family and, in part, S1P upregulates MKP-1 expression in a p38 MAPK-dependent manner. MKP-1 is a cAMP response element binding (CREB) protein-responsive gene and here, we reveal for the first time that an adenylate cyclase/PKA/CREB-mediated pathway also contributes to S1P-induced MKP-1. Thus, by increasing MKP-1 expression via parallel p38 MAPK- and CREB-mediated pathways, S1P temporally regulates MAPK signaling pathways by upregulating the negative feedback controller MKP-1. This limits the extent and duration of pro-inflammatory MAPK signaling and represses cytokine secretion in ASM cells. Taken together, our results demonstrate that S1P stimulates both kinases and the phosphatase MKP-1 to control inflammation in ASM cells and may provide a greater understanding of the molecular mechanisms responsible for the pro-asthmatic functions induced by the potent bioactive sphingolipid S1P in the lung.


Subject(s)
Bronchi/cytology , Cyclic AMP Response Element-Binding Protein/metabolism , Dual Specificity Phosphatase 1/biosynthesis , Lysophospholipids/pharmacology , Myocytes, Smooth Muscle/enzymology , Signal Transduction/drug effects , Sphingosine/analogs & derivatives , p38 Mitogen-Activated Protein Kinases/metabolism , Adenylyl Cyclases/metabolism , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation/drug effects , Enzyme Induction/drug effects , Feedback, Physiological/drug effects , Humans , Interleukin-6/metabolism , MAP Kinase Signaling System/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Phosphorylation/drug effects , Sphingosine/pharmacology , Time Factors , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...