Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Am J Physiol Lung Cell Mol Physiol ; 314(3): L421-L431, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29097425

ABSTRACT

Injurious dust exposures in the agricultural workplace involve the release of inflammatory mediators and activation of epidermal growth factor receptor (EGFR) in the respiratory epithelium. Amphiregulin (AREG), an EGFR ligand, mediates tissue repair and wound healing in the lung epithelium. Omega-3 fatty acids such as docosahexaenoic acid (DHA) are also known modulators of repair and resolution of inflammatory injury. This study investigated how AREG, DHA, and EGFR modulate lung repair processes following dust-induced injury. Primary human bronchial epithelial (BEC) and BEAS-2B cells were treated with an aqueous extract of swine confinement facility dust (DE) in the presence of DHA and AREG or EGFR inhibitors. Mice were exposed to DE intranasally with or without EGFR inhibition and DHA. Using a decellularized lung scaffolding tissue repair model, BEC recolonization of human lung scaffolds was analyzed in the context of DE, DHA, and AREG treatments. Through these investigations, we identified an important role for AREG in mediating BEC repair processes. DE-induced AREG release from BEC, and DHA treatment following DE exposure, enhanced this release. Both DHA and AREG also enhanced BEC repair capacities and rescued DE-induced recellularization deficits. In vivo, DHA treatment enhanced AREG production following DE exposure, whereas EGFR inhibitor-treated mice exhibited reduced AREG in their lung homogenates. These data indicate a role for AREG in the process of tissue repair after inflammatory lung injury caused by environmental dust exposure and implicate a role for DHA in regulating AREG-mediated repair signaling in BEC.


Subject(s)
Amphiregulin/metabolism , Bronchi/cytology , Docosahexaenoic Acids/pharmacology , Dust/analysis , Environmental Exposure/adverse effects , Epithelial Cells/cytology , Lung Injury/prevention & control , Animals , Bronchi/drug effects , Bronchi/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , ErbB Receptors/metabolism , Humans , Lung Injury/etiology , Lung Injury/metabolism , Lung Injury/pathology , Male , Mice , Mice, Inbred C57BL , Signal Transduction , Swine
2.
J Pharmacol Exp Ther ; 367(1): 101-107, 2018 10.
Article in English | MEDLINE | ID: mdl-30068728

ABSTRACT

A clade of New World monkeys (NWMs) exhibits considerable diversity in both oxytocin (OT) ligand and oxytocin receptor (OTR) structure. Most notable is the variant Pro8-OT, with proline instead of leucine at the eighth position, resulting in a rigid bend in the peptide backbone. A higher proportion of species that express Pro8-OT also engage in biparental care and social monogamy. When marmosets (genus Callithrix), a biparental and monogamous Pro8-OT NWM species, are administered the ancestral Leu8-OT, there is no change in social behavior compared with saline treatment. However, when Pro8-OT is administered, marmosets' sociosexual and prosocial behaviors are altered. The studies here tested the hypothesis that OTR binding affinities and OT-induced intracellular Ca2+ potencies would favor the native OT ligand in OTRs from four primate species, each representing a unique combination of ancestral lineage, breeding system, and native OT ligand: humans (Leu8-OT, monogamous, apes), macaques (Leu8-OT, nonmonogamous, Old World monkey), marmosets (Pro8-OT, monogamous, NWM), and titi monkeys (Leu8-OT, monogamous, NWM). OTRs were expressed in immortalized Chinese hamster ovary cells and tested for intact-cell binding affinities for Pro8-OT, Leu8-OT, and arginine vasopressin (AVP), as well as intracellular Ca2+ signaling after stimulation with Pro8-OT, Leu8-OT, and AVP. Contrary to our hypothesis, Pro8-OT bound at modestly higher affinities and stimulated calcium signaling at modestly higher potencies compared with Leu8-OT in all four primate OTRs. Thus, differences downstream from a ligand-receptor binding event are more likely to explain the different behavioral responses to these two ligands.


Subject(s)
Behavior, Animal/physiology , Oxytocin/metabolism , Receptors, Oxytocin/metabolism , Vasopressins/metabolism , Animals , Arginine Vasopressin/metabolism , CHO Cells , Calcium/metabolism , Calcium Signaling/physiology , Callithrix/metabolism , Cell Line , Cricetulus , Ligands , Primates/metabolism , Receptors, Vasopressin/metabolism , Social Behavior
3.
Exp Lung Res ; 44(10): 443-454, 2018 12.
Article in English | MEDLINE | ID: mdl-30862200

ABSTRACT

PURPOSE OF THE STUDY: Workers in enclosed hogbarns experience an increased incidence of airway inflammation and obstructive lung disease, and an aqueous hogbarn dust extract (HDE) induces multiple inflammation-related responses in cultured airway epithelial cells. Epidermal growth factor receptor (EGFR) phosphorylation and activation has been identified as one important mediator of inflammatory cytokine release from these cells. The studies here investigated both early and late phase adaptive changes in EGFR binding properties and subcellular localization induced by exposure of cells to HDE. MATERIALS AND METHODS: Cell surface EGFRs were quantified as binding to intact cells on ice. EGFR phosphorylation, expression, and localization were assessed with anti-EGFR antibodies and either blotting or confocal microscopy. RESULTS: In BEAS-2B and primary human bronchial epithelial cells, HDE induced decreases in cell surface EGFR binding following both 15-min and 18-h exposures. In contrast, H292 cells exhibited only the 15-min decrease, with binding near the control level at 18 hr. Confocal microscopy showed that the 15-min decrease in binding is due to EGFR endocytosis. Although total EGFR immunoreactivity decreased markedly at 18 hr in confocal microscopy with BEAS-2B cells, immunoblots showed no loss of EGFR protein. HDE stimulated EGFR phosphorylation at both 15 min and 18 hr in BEAS-2B cells and primary cells, but only at 15 min in H292 cells, indicating that the different EGFR binding changes among these cell types is likely related to their different time-dependent changes in phosphorylation. CONCLUSIONS: These studies extend the evidence for EGFRs as important cellular targets for components of HDE and they reveal novel patterns of EGFR phosphorylation and binding changes that vary among airway epithelial cell types. The results provide both impetus and convenient assays for identifying the EGFR-activating components and pathways that likely contribute to hogbarn dust-induced lung disease in agricultural workers.


Subject(s)
Epithelial Cells/metabolism , ErbB Receptors/metabolism , Lung Diseases/etiology , Occupational Diseases/etiology , Particulate Matter/adverse effects , Animals , Cell Line , Dust , Humans , Phosphorylation , Respiratory Mucosa/metabolism , Swine
4.
Am J Physiol Lung Cell Mol Physiol ; 311(1): L101-10, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27190062

ABSTRACT

Agricultural dust exposure results in significant lung inflammation, and individuals working in concentrated animal feeding operations (CAFOs) are at risk for chronic airway inflammatory diseases. Exposure of bronchial epithelial cells to aqueous extracts of hog CAFO dusts (HDE) leads to inflammatory cytokine production that is driven by protein kinase C (PKC) activation. cAMP-dependent protein kinase (PKA)-activating agents can inhibit PKC activation in epithelial cells, leading to reduced inflammatory cytokine production following HDE exposure. ß2-Adrenergic receptor agonists (ß2-agonists) activate PKA, and we hypothesized that ß2-agonists would beneficially impact HDE-induced adverse airway inflammatory consequences. Bronchial epithelial cells were cultured with the short-acting ß2-agonist salbutamol or the long-acting ß2-agonist salmeterol prior to stimulation with HDE. ß2-Agonist treatment significantly increased PKA activation and significantly decreased HDE-stimulated IL-6 and IL-8 production in a concentration- and time-dependent manner. Salbutamol treatment significantly reduced HDE-induced intracellular adhesion molecule-1 expression and neutrophil adhesion to epithelial cells. Using an established intranasal inhalation exposure model, we found that salbutamol pretreatment reduced airway neutrophil influx and IL-6, TNF-α, CXCL1, and CXCL2 release in bronchoalveolar lavage fluid following a one-time exposure to HDE. Likewise, when mice were pretreated daily with salbutamol prior to HDE exposure for 3 wk, HDE-induced neutrophil influx and inflammatory mediator production were also reduced. The severity of HDE-induced lung pathology in mice repetitively exposed to HDE for 3 wk was also decreased with daily salbutamol pretreatment. Together, these results support the need for future clinical investigations to evaluate the utility of ß2-agonist therapies in the treatment of airway inflammation associated with CAFO dust exposure.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Air Pollutants/toxicity , Albuterol/pharmacology , Pneumonia/drug therapy , Salmeterol Xinafoate/pharmacology , Animals , Cell Line , Cytokines/metabolism , Drug Evaluation, Preclinical , Dust , Humans , Male , Mice , Mice, Inbred C57BL , Pneumonia/etiology , Pneumonia/immunology , Respiratory Mucosa/drug effects , Respiratory Mucosa/pathology
5.
Respir Res ; 17(1): 103, 2016 08 22.
Article in English | MEDLINE | ID: mdl-27549302

ABSTRACT

BACKGROUND: Pirfenidone was recently approved for treatment of idiopathic pulmonary fibrosis. However, the therapeutic dose of pirfenidone is very high, causing side effects that limit its doses and therapeutic effectiveness. Understanding the molecular mechanisms of action of pirfenidone could improve its safety and efficacy. Because activated fibroblasts are critical effector cells associated with the progression of fibrosis, this study investigated the genes that change expression rapidly in response to pirfenidone treatment of pulmonary fibroblasts and explored their contributions to the anti-fibrotic effects of pirfenidone. METHODS: We used the GeneChip microarray to screen for genes that were rapidly up-regulated upon exposure of human lung fibroblast cells to pirfenidone, with confirmation for specific genes by real-time PCR and western blots. Biochemical and functional analyses were used to establish their anti-fibrotic effects in cellular and animal models of pulmonary fibrosis. RESULTS: We identified Regulator of G-protein Signaling 2 (RGS2) as an early pirfenidone-induced gene. Treatment with pirfenidone significantly increased RGS2 mRNA and protein expression in both a human fetal lung fibroblast cell line and primary pulmonary fibroblasts isolated from patients without or with idiopathic pulmonary fibrosis. Pirfenidone treatment or direct overexpression of recombinant RGS2 in human lung fibroblasts inhibited the profibrotic effects of thrombin, whereas loss of RGS2 exacerbated bleomycin-induced pulmonary fibrosis and mortality in mice. Pirfenidone treatment reduced bleomycin-induced pulmonary fibrosis in wild-type but not RGS2 knockout mice. CONCLUSIONS: Endogenous RGS2 exhibits anti-fibrotic functions. Upregulated RGS2 contributes significantly to the anti-fibrotic effects of pirfenidone.


Subject(s)
Fibroblasts/drug effects , Idiopathic Pulmonary Fibrosis/drug therapy , Lung/drug effects , Pyridones/pharmacology , RGS Proteins/metabolism , Animals , Bleomycin , Calcium Signaling/drug effects , Cell Line , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Profiling/methods , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Lung/metabolism , Lung/pathology , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , RGS Proteins/deficiency , RGS Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thrombin/pharmacology , Time Factors , Transfection , Up-Regulation
6.
Am J Respir Cell Mol Biol ; 53(1): 42-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25368964

ABSTRACT

G protein-coupled receptors (GPCRs) are important regulators of cell functions in asthma. We recently reported that regulator of G-protein signaling (RGS) 2, a selective modulator of Gq-coupled GPCRs, is a key regulator of airway hyper-responsiveness (AHR), the pathophysiologic hallmark of asthma. Because RGS2 protein levels in airway cells were significantly lower in patients with asthma compared with patients without asthma, we further investigated the potential pathological importance of RGS2 repression in asthma. The human RGS2 gene maps to chromosome 1q31. We first screened patients with asthma for RGS2 gene promoter single-nucleotide polymorphisms (SNPs) and found significant differences in the distribution of two RGS2 SNPs (A638G, rs2746071 and C395G, rs2746072) between patients with asthma and nonasthmatic subjects. These two SNPs are always associated with each other and have the same higher prevalence in patients with asthma (65%) as compared with nonasthmatic subjects (35%). Point mutations corresponding to these SNPs decrease RGS2 promoter activity by 44%. The importance of RGS2 down-regulation was then determined in an acute IL-13 mouse model of asthma. Intranasal administration of IL-13 in mice also decreased RGS2 expression in lungs by ∼50% and caused AHR. Although naive RGS2 knockout (KO) mice exhibit spontaneous AHR, acute IL-13 exposure further increased AHR in RGS2 KO mice. Loss of RGS2 also significantly enhanced IL-13-induced mouse airway remodeling, including peribronchial smooth muscle thickening and fibrosis, without effects on goblet cell hyperplasia or airway inflammation in mice. Thus, genetic variations and increased inflammatory cytokines can lead to RGS2 repression, which exacerbates AHR and airway remodeling in asthma.


Subject(s)
Asthma/genetics , Asthma/metabolism , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , RGS Proteins , Airway Remodeling , Animals , Asthma/chemically induced , Asthma/pathology , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 1/metabolism , Disease Models, Animal , Female , Humans , Interleukin-13/toxicity , Male , Mice , Mice, Knockout , Muscle, Smooth/metabolism , Muscle, Smooth/pathology , RGS Proteins/genetics , RGS Proteins/metabolism
7.
Am J Physiol Renal Physiol ; 308(2): F131-9, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25377915

ABSTRACT

Treating chronic kidney disease (CKD) has been challenging because of its pathogenic complexity. Epoxyeicosatrienoic acids (EETs) are cytochrome P-450-dependent derivatives of arachidonic acid with antihypertensive, anti-inflammatory, and profibrinolytic functions. We recently reported that genetic ablation of soluble epoxide hydrolase (sEH), an enzyme that converts EETs to less active dihydroxyeicosatrienoic acids, prevents renal tubulointerstitial fibrosis and inflammation in experimental mouse models of CKD. Here, we tested the hypothesis that pharmacological inhibition of sEH after unilateral ureteral obstruction (UUO) would attenuate tubulointerstitial fibrosis and inflammation in mouse kidneys and may provide a novel approach to manage the progression of CKD. Inhibition of sEH enhanced levels of EET regioisomers and abolished tubulointerstitial fibrosis, as demonstrated by reduced collagen deposition and myofibroblast formation after UUO. The inflammatory response was also attenuated, as demonstrated by decreased influx of neutrophils and macrophages and decreased expression of inflammatory cytokines keratinocyte chemoattractant, macrophage inflammatory protein-2, monocyte chemotactic protein-1, TNF-α, and ICAM-1 in kidneys after UUO. UUO upregulated transforming growth factor-ß1/Smad3 signaling and induced NF-κB activation, oxidative stress, tubular injury, and apoptosis; in contrast, it downregulated antifibrotic factors, including peroxisome proliferator-activated receptor (PPAR) isoforms, especially PPAR-γ. sEH inhibition mitigated the aforementioned malevolent effects in UUO kidneys. These data demonstrate that pharmacological inhibition of sEH promotes anti-inflammatory and fibroprotective effects in UUO kidneys by preventing tubular injury, downregulation of NF-κB, transforming growth factor-ß1/Smad3, and inflammatory signaling pathways, and activation of PPAR isoforms. Our data suggest the potential use of sEH inhibitors in treating fibrogenesis in the UUO model of CKD.


Subject(s)
Arachidonic Acids/metabolism , Benzoates/therapeutic use , Epoxide Hydrolases/antagonists & inhibitors , Nephrosclerosis/prevention & control , Phenylurea Compounds/therapeutic use , Renal Insufficiency, Chronic/prevention & control , Animals , Benzoates/pharmacology , Blood Pressure/drug effects , Cell Death/drug effects , Drug Evaluation, Preclinical , Male , Mice, Inbred C57BL , Nephrosclerosis/etiology , Nephrosclerosis/metabolism , Oxidative Stress/drug effects , Peroxisome Proliferator-Activated Receptors/metabolism , Phenylurea Compounds/pharmacology , Renal Circulation/drug effects , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/metabolism , Ureteral Obstruction/complications
8.
Am J Physiol Lung Cell Mol Physiol ; 309(4): L388-99, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26092994

ABSTRACT

Workers exposed to aerosolized dust present in concentrated animal feeding operations (CAFOs) are susceptible to inflammatory lung diseases, such as chronic obstructive pulmonary disease. Extracts of dust collected from hog CAFOs [hog dust extract (HDE)] are potent stimulators of lung inflammatory responses in several model systems. The observation that HDE contains active proteases prompted the present study, which evaluated the role of CAFO dust proteases in lung inflammatory processes and tested whether protease-activated receptors (PARs) are involved in the signaling pathway for these events. We hypothesized that the damaging proinflammatory effect of HDE is due, in part, to the proteolytic activation of PARs, and inhibiting the proteases in HDE or disrupting PAR activation would attenuate HDE-mediated inflammatory indexes in bronchial epithelial cells (BECs), in mouse lung slices in vitro, and in a murine in vivo exposure model. Human BECs and mouse lung slice cultures stimulated with 5% HDE released significantly more of each of the cytokines measured (IL-6, IL-8, TNF-α, keratinocyte-derived chemokine/CXC chemokine ligand 1, and macrophage inflammatory protein-2/CXC chemokine ligand 2) than controls, and these effects were markedly diminished by protease inhibition. Inhibition of PARs also blunted the HDE-induced cytokine release from BECs. In addition, protease depletion inhibited HDE-induced BEC intracellular PKCα and PKCε activation. C57BL/6J mice administered 12.5% HDE intranasally, either once or daily for 3 wk, exhibited increased total cellular and neutrophil influx, bronchial alveolar fluid inflammatory cytokines, lung histopathology, and inflammatory scores compared with mice receiving protease-depleted HDE. These data suggest that proteases in dust from CAFOs are important mediators of lung inflammation, and these proteases and their receptors may provide novel targets for therapeutic intervention in CAFO dust-induced airways disease.


Subject(s)
Agricultural Workers' Diseases/immunology , Peptide Hydrolases/immunology , Pneumonia/immunology , Receptor, PAR-1/metabolism , Receptor, PAR-2/metabolism , Air Pollutants, Occupational/immunology , Animal Feed , Animals , Bronchi/pathology , Cell Line , Cytokines/metabolism , Dust/immunology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Humans , Male , Mice, Inbred C57BL , Occupational Exposure , Protein Kinase C/metabolism , Swine
9.
Article in English | MEDLINE | ID: mdl-25460827

ABSTRACT

Epithelial-mesenchymal transition (EMT) is critical for embryonic development, and this process is recapitulated in adults during wound healing, tissue regeneration, fibrosis and cancer progression. Cell migration is believed to play a key role in both normal wound repair and in abnormal tissue remodeling. Prostaglandin E2 (PGE2) inhibits fibroblast chemotaxis, but stimulates chemotaxis in airway epithelial cells. The current study was designed to explore the role of PGE2 and its four receptors on airway epithelial cell migration following EMT using both the Boyden blindwell chamber chemotaxis assay and the wound closure assay. EMT in human bronchial epithelial cells (HBECs) was induced by TGF-ß1 and a mixture of cytokines (IL-1ß, TNF-α, and IFN-γ). PGE2 and selective agonists for all four EP receptors stimulated chemotaxis and wound closure in HBECs. Following EMT, the EP1 and EP3 agonists were without effect, while the EP2 and EP4 agonists inhibited chemotaxis as did PGE2. The effects of the EP2 and EP4 receptors on HBEC and EMT cell migration were further confirmed by blocking the expected signaling pathways. Taken together, these results demonstrate that PGE2 switches from a stimulator to an inhibitor of cell migration following EMT of airway epithelial cells and that this inhibition is mediated by an altered effect of EP2 and EP4 signaling and an apparent loss of the stimulatory effects of EP1 and EP3. Change in the PGE2 modulation of chemotaxis may play a role in repair following injury.


Subject(s)
Cell Movement/drug effects , Dinoprostone/pharmacology , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/drug effects , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Cell Line , Cytokines/metabolism , Epithelial Cells/cytology , Humans
10.
Am J Respir Cell Mol Biol ; 49(4): 571-81, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23656623

ABSTRACT

Lung fibroblasts are believed to be a major source of vascular endothelial growth factor (VEGF), which supports the survival of lung endothelial cells and modulates the maintenance of the pulmonary microvasculature. VEGF has been related to the pathogenesis of lung diseases, including chronic obstructive pulmonary disease (COPD). Prostaglandin E2 (PGE2) stimulates VEGF production from lung fibroblasts via the E-prostanoid (EP)-2 receptor. The EP2 signaling pathway uses cyclic adenosine monophosphate (cAMP) as a second messenger, and cAMP is degraded by phosphodiesterases (PDEs). This study investigates whether phosphodiesterase inhibition modulates the human lung fibroblast VEGF production induced by PGE2. Human fetal lung fibroblasts were cultured with PGE2 and PDE inhibitors. The PDE4 inhibitors roflumilast, roflumilast N-oxide, and rolipram with PGE2 increased VEGF release, as quantified in supernatant media by ELISA. In contrast, PDE3, PDE5, and PDE7 inhibitors did not affect VEGF release. Roflumilast increased VEGF release with either an EP2 or an EP4 agonist. Roflumilast augmented the cytosolic cAMP levels induced by PGE2 and VEGF release with other agents that use the cAMP signaling pathway. Roflumilast-augmented VEGF release was completely inhibited by a protein kinase A (PKA) inhibitor. Roflumilast with PGE2 increased VEGF mRNA levels, and the blockade of mRNA synthesis inhibited the augmented VEGF release. The stimulatory effect of roflumilast on VEGF release was replicated using primary healthy and COPD lung fibroblasts. These findings demonstrate that PDE4 inhibition can modulate human lung fibroblast VEGF release by PGE2 acting through the EP2 and EP4 receptor-cAMP/PKA signaling pathway. Through this action, PDE4 inhibitors such as roflumilast could contribute to the survival of lung endothelial cells.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Dinoprostone/pharmacology , Lung/drug effects , Lung/metabolism , Phosphodiesterase 4 Inhibitors/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Aminopyridines/pharmacology , Benzamides/pharmacology , Cells, Cultured , Cyclic AMP/genetics , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclopropanes/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Lung/cytology , Pulmonary Disease, Chronic Obstructive/metabolism , RNA, Messenger/genetics , Receptors, Prostaglandin E, EP2 Subtype/genetics , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/genetics
11.
Am J Respir Cell Mol Biol ; 47(6): 729-37, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23043089

ABSTRACT

The etiology of chronic obstructive pulmonary disease (COPD) is complex and involves an aberrant inflammatory response. Prostaglandin (PG)E2 is elevated in COPD, is a key modulator of lung fibroblast functions, and may influence COPD progression. Most studies evaluating the effects of PGE2 on lung fibroblasts have used acute exposures. The current study evaluated whether longer-term exposure would induce attenuation of PGE2 signaling as part of an autoregulatory pathway. Human fetal lung fibroblasts were pretreated with PGE2 for 24 hours, and migration and cAMP accumulation in response to acute stimulation with PGE2 were assessed. Fibroblasts from adults with and without COPD were pretreated, and migration was assessed. PGE2 pretreatment attenuated subsequent PGE2-mediated inhibition of chemotaxis and cAMP stimulation. This attenuation was predominantly due to an increase in phosphodiesterase (PDE)4-mediated degradation of cAMP rather than to decreased activation of PGE2 receptors (receptor desensitization). Albuterol- and iloprost-mediated signaling were also attenuated after PGE2 pretreatment, suggesting that activation of PDE4 was able to broadly modulate multiple cAMP-coupled pathways. Lung fibroblasts from adult control subjects pretreated with PGE2 also developed attenuation of PGE2-mediated inhibition of chemotaxis. In contrast, fibroblasts obtained from patients with COPD maintained inhibitory PGE2 signaling after PGE2 pretreatment. These data identify a PDE4-mediated attenuation of PGE2 inhibitory signaling in normal fibroblasts that appears to be altered in COPD fibroblasts. These alterations may contribute to COPD pathogenesis and could provide novel therapeutic targets.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Dinoprostone/metabolism , Fibroblasts/metabolism , Lung/pathology , Adrenergic beta-2 Receptor Agonists/pharmacology , Albuterol/pharmacology , Aminopyridines/pharmacology , Benzamides/pharmacology , Cells, Cultured , Chemotaxis , Colforsin/pharmacology , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Cyclopropanes/pharmacology , Dinoprostone/physiology , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibronectins/physiology , Gene Expression , Humans , Iloprost/pharmacology , Isoenzymes/genetics , Isoenzymes/metabolism , Phosphodiesterase 4 Inhibitors/pharmacology , Pulmonary Disease, Chronic Obstructive/enzymology , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Receptors, Epoprostenol , Receptors, Prostaglandin/agonists , Receptors, Prostaglandin E, EP2 Subtype/agonists , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Rolipram/pharmacology , Second Messenger Systems
12.
J Pharmacol Exp Ther ; 342(2): 305-11, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22543031

ABSTRACT

We recently reported that phosphoinositide 3-kinase γ (PI3Kγ) directly regulates airway smooth muscle (ASM) contraction by modulating Ca(2+) oscillations. Because ASM contraction plays a critical role in airway hyperresponsiveness (AHR) of asthma, the aim of the present study was to determine whether targeting PI3Kγ in ASM cells could suppress AHR in vitro and in vivo. Intranasal administration into mice of interleukin-13 (IL-13; 10 µg per mouse), a key pathophysiologic cytokine in asthma, induced AHR after 48 h, as assessed by invasive tracheostomy. Intranasal administration of a broad-spectrum PI3K inhibitor or a PI3Kγ-specific inhibitor 1 h before AHR assessment attenuated IL-13 effects. Airway responsiveness to bronchoconstrictor agonists was also examined in precision-cut mouse lung slices pretreated without or with IL-13 for 24 h. Acetylcholine and serotonin dose-response curves indicated that IL-13-treated lung slices had a 40 to 50% larger maximal airway constriction compared with controls. Furthermore, acetylcholine induced a larger initial Ca(2+) transient and increased Ca(2+) oscillations in IL-13-treated primary mouse ASM cells compared with control cells, correlating with increased cell contraction. As expected, PI3Kγ inhibitor treatment attenuated IL-13-augmented airway contractility of lung slices and ASM cell contraction. In both control and IL-13-treated ASM cells, small interfering RNA-mediated knockdown of PI3Kγ by 70% only reduced the initial Ca(2+) transient by 20 to 30% but markedly attenuated Ca(2+) oscillations and contractility of ASM cells by 50 to 60%. This report is the first to demonstrate that PI3Kγ in ASM cells is important for IL-13-induced AHR and that acute treatment with a PI3Kγ inhibitor can ameliorate AHR in a murine model of asthma.


Subject(s)
Bronchial Hyperreactivity/metabolism , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Interleukin-13/immunology , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/drug effects , Phosphoinositide-3 Kinase Inhibitors , Acetylcholine/pharmacology , Animals , Asthma/genetics , Asthma/immunology , Asthma/metabolism , Bronchial Hyperreactivity/genetics , Bronchial Hyperreactivity/immunology , Calcium/metabolism , Calcium Signaling/drug effects , Calcium Signaling/genetics , Cells, Cultured , Class Ib Phosphatidylinositol 3-Kinase/genetics , Interleukin-13/metabolism , Lung/drug effects , Lung/immunology , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle Contraction/drug effects , Muscle Contraction/genetics , Muscle Contraction/immunology , Muscle, Smooth/immunology , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism , Serotonin/pharmacology , Trachea/drug effects , Trachea/immunology , Trachea/metabolism
13.
Am J Respir Cell Mol Biol ; 45(4): 882-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21441380

ABSTRACT

A subset of workers in swine confinement facilities develops chronic respiratory disease. An aqueous extract of dust from these facilities (hogbarn dust extract [HDE]) induces IL-6 and IL-8 release and several other responses in isolated airway epithelial cells. The cell membrane receptors by which HDE initiates these responses have not been identified. Because several other inhaled agents induce airway epithelial cell responses through epidermal growth factor receptor (EGFR) activation, we hypothesized that HDE would activate EGFRs and that EGFRs would be required for some of the responses to HDE. Exposure of Beas-2B cells to HDE caused EGFR phosphorylation and downstream ERK activation, and both responses were blocked by the EGFR-selective kinase inhibitor AG1478. AG1478 and EGFR-neutralizing antibody reduced HDE-stimulated IL-6 and IL-8 release by about half. Similar EGFR phosphorylation and requirement of EGFRs for maximal IL-6 and IL-8 release were found with primary isolates of human bronchial epithelial cells. Because HDE-stimulated IL-6 and IL-8 release involve the Ca(2+)-dependent protein kinase Cα, we hypothesized that HDE would induce intracellular Ca(2+) mobilization. HDE exposure induced intracellular Ca(2+) mobilization in Beas-2B cells and in primary cell isolates, but this response was neither mimicked by EGF nor inhibited by AG1478. Thus, HDE activates EGFRs and their downstream signaling, and EGFR activation is required for some but not all airway epithelial cell responses to HDE.


Subject(s)
Air Pollutants, Occupational/toxicity , Animal Husbandry , Bronchi/drug effects , Calcium/metabolism , Cytokines/metabolism , Dust , Epithelial Cells/drug effects , ErbB Receptors/metabolism , Respiratory Mucosa/drug effects , Animals , Antibodies, Neutralizing/pharmacology , Bronchi/immunology , Bronchi/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Epithelial Cells/immunology , Epithelial Cells/metabolism , ErbB Receptors/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Interleukin-6/metabolism , Interleukin-8/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Quinazolines , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Swine , Tyrphostins/pharmacology
14.
Am J Respir Cell Mol Biol ; 44(1): 99-107, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20203295

ABSTRACT

The migration of fibroblasts is believed to play a key role in both normal wound repair and abnormal tissue remodeling. Prostaglandin E (PGE)(2), a mediator that can inhibit many fibroblast functions including chemotaxis, was reported to be mediated by the E-prostanoid (EP) receptor EP2. PGE(2), however, can act on four receptors. This study was designed to determine if EP receptors, in addition to EP2, can modulate fibroblast chemotaxis. Using human fetal lung fibroblasts, the expression of all four EP receptors was demonstrated by Western blotting. EP2-selective and EP4-selective agonists inhibited both chemotaxis toward fibronectin in the blindwell assay and migration in a wound-closure assay. In contrast, EP1-selective and EP3-selective agonists stimulated cell migration in both assay systems. These results were confirmed using EP-selective antagonists. The role of both EP2 and EP4 receptors in mediating the PGE(2) inhibition of chemotaxis was also confirmed by small interfering RNA suppression. Furthermore, the role of EP receptors was confirmed by blocking the expected signaling pathways. Taken together, these results demonstrate that PGE(2) can act on multiple EP receptors in human lung fibroblasts, to exert disparate effects. Alterations in EP receptor expression may have the potential to alter PGE(2) action. Targeting specific EP receptors may offer therapeutic opportunities in conditions characterized by abnormal tissue repair and remodeling.


Subject(s)
Bronchi/metabolism , Chemotaxis , Dinoprostone/metabolism , Fibroblasts/metabolism , Receptors, Prostaglandin E/metabolism , Signal Transduction , Wound Healing , Blotting, Western , Bronchi/drug effects , Cell Proliferation , Cells, Cultured , Chemotaxis/drug effects , Fibroblasts/drug effects , Fibronectins/metabolism , Humans , RNA Interference , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP1 Subtype/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Signal Transduction/drug effects , Time Factors , Wound Healing/drug effects
15.
J Pharmacol Exp Ther ; 334(3): 703-9, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20501633

ABSTRACT

Phosphoinositide 3-kinase gamma (PI3Kgamma) has been implicated in the pathogenesis of asthma, but its mechanism has been considered indirect, through release of inflammatory cell mediators. Because airway smooth muscle (ASM) contractile hyper-responsiveness plays a critical role in asthma, the aim of the present study was to determine whether PI3Kgamma can directly regulate contractility of ASM. Immunohistochemistry staining indicated expression of PI3Kgamma protein in ASM cells of mouse trachea and lung, which was confirmed by Western blot analysis in isolated mouse tracheal ASM cells. PI3Kgamma inhibitor II inhibited acetylcholine (ACh)-stimulated airway contraction of cultured precision-cut mouse lung slices in a dose-dependent manner with 75% inhibition at 10 muM. In contrast, inhibitors of PI3Kalpha, PI3Kbeta, or PI3Kdelta, at concentrations 40-fold higher than their reported IC(50) values for their primary targets, had no effect. It is noteworthy that airways in lung slices pretreated with PI3Kgamma inhibitor II still exhibited an ACh-induced initial contraction, but the sustained contraction was significantly reduced. Furthermore, the PI3Kgamma-selective inhibitor had a small inhibitory effect on the ACh-stimulated initial Ca(2+) transient in ASM cells of mouse lung slices or isolated mouse ASM cells but significantly attenuated the sustained Ca(2+) oscillations that are critical for sustained airway contraction. This report is the first to show that PI3Kgamma directly controls contractility of airways through regulation of Ca(2+) oscillations in ASM cells. Thus, in addition to effects on airway inflammation, PI3Kgamma inhibitors may also exert direct effects on the airway contraction that contribute to pathologic airway hyper-responsiveness.


Subject(s)
Calcium Signaling/physiology , Muscle, Smooth/physiology , Phosphatidylinositol 3-Kinases/metabolism , Respiratory System/drug effects , Respiratory System/enzymology , Acetylcholine/pharmacology , Animals , Bronchial Hyperreactivity/physiopathology , Calcium Signaling/drug effects , Cell Separation , Class Ib Phosphatidylinositol 3-Kinase , Enzyme Inhibitors/pharmacology , Immunohistochemistry , In Vitro Techniques , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Isoenzymes/physiology , Lung/drug effects , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Smooth/drug effects , Phosphoinositide-3 Kinase Inhibitors , Trachea/cytology , Trachea/drug effects
16.
Biochem Pharmacol ; 180: 114172, 2020 10.
Article in English | MEDLINE | ID: mdl-32712053

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a devastating interstitial lung disease with irreversible loss of lung tissue and function. Myofibroblasts in the lung are key cellular mediators of IPF progression. Transforming growth factor (TGF)-ß1, a major profibrogenic cytokine, induces pulmonary myofibroblast differentiation, and emerging evidence has established the importance of microRNAs (miRs) in the development of IPF. The objective of this study was to define the pro-fibrotic roles and mechanisms of miRs in TGF-ß1-induced pulmonary myofibroblast differentiation. Using RNA sequencing, we identified miR-424 as an important TGF-ß1-induced miR in human lung fibroblasts (HLFs). Quantitative RT-PCR confirmed that miR-424 expression was increased by 2.6-fold in HLFs in response to TGF-ß1 and was 1.7-fold higher in human fibrotic lung tissues as compared to non-fibrotic lung tissues. TGF-ß1-induced upregulation of miR-424 was blocked by the Smad3 inhibitor SIS3, suggesting the involvement of this canonical TGF-ß1 signaling pathway. Transfection of a miR-424 hairpin inhibitor into HLFs reduced TGF-ß1-induced expression of classic myofibroblast differentiation markers including ɑ-smooth muscle actin (ɑ-SMA) and connective tissue growth factor (CTGF), whereas a miR-424 mimic significantly enhanced TGF-ß1-induced myofibroblast differentiation. In addition, TGF-ß1 induced Smad3 phosphorylation in HLFs, and this response was reduced by the miR-424 inhibitor. In silico analysis identified Slit2, a protein that inhibits TGF-ß1 profibrogenic signaling, as a putative target of regulation by miR-424. Slit2 is less highly expressed in human fibrotic lung tissues than in non-fibrotic lung tissues, and knockdown of Slit2 by its siRNA enhanced TGF-ß1-induced HLF differentiation. Overexpression of a miR-424 mimic down-regulated expression of Slit2 but not the Slit2 major receptor ROBO1 in HLFs. Luciferase reporter assays showed that the miR-424 mimic represses Slit2 3' untranslated region (3'-UTR) reporter activity, and mutations at the seeding regions in the 3'-UTR of Slit2 abolish this inhibition. Together, these data demonstrate a pro-fibrotic role of miR-424 in TGF-ß1-induced HLF differentiation. It functions as a positive feed-back regulator of the TGF-ß1 signaling pathway by reducing expression of the negative regulator Slit2. Thus, targeting miR-424 may provide a new therapeutic strategy to prevent myofibroblast differentiation and IPF progression.


Subject(s)
Cell Differentiation/physiology , Intercellular Signaling Peptides and Proteins/biosynthesis , Lung/metabolism , MicroRNAs/biosynthesis , Myofibroblasts/metabolism , Nerve Tissue Proteins/biosynthesis , Transforming Growth Factor beta1/pharmacology , Cell Differentiation/drug effects , Dose-Response Relationship, Drug , Gene Expression , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Lung/cytology , Lung/drug effects , MicroRNAs/genetics , Myofibroblasts/drug effects , Nerve Tissue Proteins/genetics
17.
Naunyn Schmiedebergs Arch Pharmacol ; 393(5): 843-856, 2020 05.
Article in English | MEDLINE | ID: mdl-31884570

ABSTRACT

Pulmonary fibrosis is characterized by fibroblasts persisting in an activated form, producing excessive fibrous material that destroys alveolar structure. The second messenger molecule cyclic 3',5'-adenosine monophosphate (cAMP) has antifibrotic properties, and prostaglandin E2 (PGE2) can stimulate cAMP production through prostaglandin E (EP)2 and EP4 receptors. Although EP receptors are attractive therapeutic targets, the effects of long-term exposure to PGE2 have not been characterized. To determine the effects of long-term exposure of lung fibroblasts to PGE2, human fetal lung (HFL)-1 cells were treated for 24 h with 100 nM PGE2 or other cAMP-elevating agents. cAMP levels stimulated by acute exposure to PGE2 were measured using a fluorescent biosensor. Pretreatment for 24 h with PGE2 shifted the concentration-response curve to PGE2 rightward by approximately 22-fold but did not affect responses to the beta-adrenoceptor agonist isoproterenol. Neither isoproterenol nor forskolin pretreatment altered PGE2 responses, implying that other cAMP-elevating agents do not induce desensitization. Use of EP2- and EP4-selective agonists and antagonists suggested that PGE2-stimulated cAMP responses in HFL-1 cells are mediated by EP2 receptors. EP2 receptors are resistant to classical mechanisms of agonist-specific receptor desensitization, so we hypothesized that increased PDE activity mediates the loss of signaling after PGE2 pretreatment. PGE2 treatment upregulated messenger RNA for PDE3A, PDE3B, PDE4B, and PDE4D and increased overall PDE activity. The PDE4 inhibitor rolipram partially reversed PGE2-mediated desensitization and PDE4 activity was increased, but rolipram did not alter responses to isoproterenol. The PDE3 inhibitor cilostazol had minimal effect. These results show that long-term exposure to PGE2 causes agonist-specific desensitization of EP2 receptor-stimulated cAMP signaling through the increased expression of PDE isozymes, most likely of the PDE4 family.


Subject(s)
Cyclic AMP/metabolism , Dinoprostone/pharmacology , Fibroblasts/drug effects , Lung/drug effects , Phosphoric Diester Hydrolases/metabolism , Pulmonary Fibrosis/drug therapy , Receptors, Prostaglandin E, EP2 Subtype/agonists , Cells, Cultured , Dose-Response Relationship, Drug , Fibroblasts/enzymology , Fibroblasts/pathology , Humans , Isoenzymes , Lung/enzymology , Lung/pathology , Phosphoric Diester Hydrolases/genetics , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/pathology , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Second Messenger Systems , Up-Regulation
18.
Sci Rep ; 9(1): 15480, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31664130

ABSTRACT

Oxytocin (OXT) is an important neuromodulator of social behaviors via activation of both oxytocin receptors (OXTR) and vasopressin (AVP) 1a receptors (AVPR1a). Marmosets are neotropical primates with a modified OXT ligand (Pro8-OXT), and this ligand shows significant coevolution with traits including social monogamy and litter size. Pro8-OXT produces more potent and efficacious responses at primate OXTR and stronger behavioral effects than the consensus mammalian OXT ligand (Leu8-OXT). Here, we tested whether OXT/AVP ligands show differential levels of crosstalk at primate AVPR1a. We measured binding affinities and Ca2+ signaling responses of AVP, Pro8-OXT and Leu8-OXT at human, macaque, and marmoset AVPR1a. We found that AVP binds with higher affinity than OXT across AVPR1a, and marmoset AVPR1a show a 10-fold lower OXT binding affinity compared to human and macaque AVPR1a. Both Leu8-OXT and Pro8-OXT produce a less efficacious response than AVP at human AVPR1a and higher efficacious response than AVP at marmoset AVPR1a. These data suggest that OXT might partially antagonize endogenous human AVPR1a signaling and enhance marmoset AVPR1a signaling. These findings aid in further understanding inconsistencies observed following systemic intranasal administration of OXT and provide important insights into taxon-specific differences in nonapeptide ligand/receptor coevolution and behavior.


Subject(s)
Arginine Vasopressin/pharmacology , Leucine/chemistry , Oxytocin/pharmacology , Proline/chemistry , Receptors, Oxytocin/agonists , Receptors, Vasopressin/agonists , Animals , Arginine Vasopressin/chemistry , CHO Cells , Calcium/metabolism , Callithrix , Cricetulus , Humans , Macaca , Oxytocin/chemistry , Receptors, Oxytocin/metabolism , Signal Transduction , Species Specificity
19.
Am J Respir Cell Mol Biol ; 39(3): 356-63, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18367729

ABSTRACT

Migration of fibroblasts plays an essential role in tissue repair after injury. Sphingosine 1-phosphate (S1P) is a multifunctional mediator released by many cells that can be released in inflammation and after injury. This study evaluated the effect of S1P on fibroblast chemotaxis toward fibronectin. S1P alone did not affect fibroblast migration, but S1P enhanced fibronectin-directed chemotaxis in a concentration-dependent manner. The effect of S1P was not mimicked by dihydro (dh) S1P or the S1P(1) receptor agonist SEW2871. S1P augmentation of fibroblast chemotaxis, however, was completely blocked by JTE-013, an S1P(2) antagonist, but not by suramin, an S1P(3) antagonist. Suppression of the S1P(2) receptor by small interfering (si)RNA also completely blocked S1P augmentation of fibroblast chemotaxis to fibronectin. S1P stimulated Rho activation and focal adhesion kinase (FAK) phosphorylation, and these were also significantly inhibited by the S1P(2) receptor antagonist (JTE-013) or by S1P(2) siRNA. Further, the potentiation of S1P signaling was blocked by the Rho-kinase inhibitor Y-27632 in a concentration-dependent manner. Inhibition of FAK with siRNA reduced basal chemotaxis toward fibronectin slightly but significantly, and almost completely blocked S1P augmented chemotaxis. These results suggest that S1P-augmented fibroblast chemotaxis toward fibronectin depends on the S1P(2) receptor and requires Rho and Rho-kinase, and FAK phosphorylation. By augmenting fibroblast recruitment, S1P has the potential to modulate tissue repair after injury. The pathways by which S1P mediates this effect, therefore, represent a potential therapeutic target to affect tissue repair and remodeling.


Subject(s)
Chemotaxis , Fibroblasts/physiology , Lung/physiology , Lysophospholipids/physiology , Receptors, Lysosphingolipid/physiology , Sphingosine/analogs & derivatives , Base Sequence , Blotting, Western , Cells, Cultured , DNA Primers , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibronectins/pharmacology , Humans , Immunoprecipitation , Lung/cytology , Lung/drug effects , Lung/metabolism , Lysophospholipids/pharmacology , RNA Interference , Regeneration/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Sphingosine/pharmacology , Sphingosine/physiology
20.
J Pharmacol Exp Ther ; 325(3): 809-17, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18309089

ABSTRACT

Lysophosphatidic acid (LPA) and epidermal growth factor (EGF) are important mediators of lung cell function and lung diseases. We showed previously that LPA decreases epidermal growth factor receptor (EGFR) binding rapidly in BEAS-2B airway epithelial cells, and this decrease is sustained to at least 18 h. The current studies investigate which LPA signaling pathways mediate the rapid versus sustained decreases in EGFR binding in BEAS-2B cells. The G(i/o) inhibitor pertussis toxin and the Rho kinase inhibitor Y-27632 [(R)-(+)-trans-N-(4-pyridyl)-4-(1-aminoethyl)-cyclohexanecarboxamide] had no effect on the rapid or sustained decreases. However, the mitogen-activated protein kinase kinase (MEK) inhibitor U0126 [1,4-diamino-2,3-dicyano-1,4-bis(o-aminophenylmercapto)-butadiene ethanolate] decreased extracellular signal-regulated kinase (ERK) 1/2 phosphorylation, completely inhibited the rapid decrease in binding, and partially inhibited the sustained decrease. The direct Ca2+- and phospholipid-dependent protein kinase (PKC) activator phorbol-12-myristate-13-acetate stimulated ERK1/2 phosphorylation and decreased EGFR binding at both 15 min and 18 h. Furthermore, inhibitors of PKC partially inhibited ERK1/2 phosphorylation and the 15-min decrease but completely inhibited the 18-h decrease. Inhibitor time course studies showed that PKC induction of the 18-h decrease occurred during the first 3 h of treatment. We showed previously that LPA-stimulated EGFR transactivation contributes to the rapid decrease. Two transactivation inhibitors partially inhibited ERK1/2 phosphorylation, and U0126 partially inhibited EGFR transactivation, indicating that MEK may be involved both upstream and downstream of EGFR activation. Together, the data presented here indicate that LPA mediates the rapid decrease in EGFR binding via EGFR transactivation, MEK/ERK, and PKC, whereas the sustained decrease is regulated primarily by PKC.


Subject(s)
Epithelial Cells/drug effects , ErbB Receptors/metabolism , Lysophospholipids/pharmacology , Protein Serine-Threonine Kinases/metabolism , Cell Line , Epithelial Cells/metabolism , Humans , Protein Serine-Threonine Kinases/antagonists & inhibitors , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL