Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
Front Cell Dev Biol ; 8: 165, 2020.
Article in English | MEDLINE | ID: mdl-32328490

ABSTRACT

With the ability to switch between proliferative and contractile phenotype, airway smooth muscle (ASM) cells can contribute to the progression of airway diseases such as asthma and chronic obstructive pulmonary disease (COPD), in which airway obstruction is associated with ASM hypertrophy and hypercontractility. A-kinase anchoring proteins (AKAPs) have emerged as important regulatory molecules in various tissues, including ASM cells. AKAPs can anchor the regulatory subunits of protein kinase A (PKA), and guide cellular localization via various targeting domains. Here we investigated whether disruption of the AKAP-PKA interaction, by the cell permeable peptide stearated (st)-Ht31, alters human ASM proliferation and contractility. Treatment of human ASM with st-Ht31 enhanced the expression of protein markers associated with cell proliferation in both cultured cells and intact tissue, although this was not accompanied by an increase in cell viability or cell-cycle progression, suggesting that disruption of AKAP-PKA interaction on its own is not sufficient to drive ASM cell proliferation. Strikingly, st-Ht31 enhanced contractile force generation in human ASM tissue with concomitant upregulation of the contractile protein α-sm-actin. This upregulation of α-sm-actin was independent of mRNA stability, transcription or translation, but was dependent on proteasome function, as the proteasome inhibitor MG-132 prevented the st-Ht31 effect. Collectively, the AKAP-PKA interaction appears to regulate markers of the multi-functional capabilities of ASM, and this alter the physiological function, such as contractility, suggesting potential to contribute to the pathophysiology of airway diseases.

2.
Cells ; 9(2)2020 02 03.
Article in English | MEDLINE | ID: mdl-32028718

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) plays a role in chronic obstructive pulmonary diseases (COPD). Cyclic adenosine monophosphate (cAMP) can inhibit transforming growth factor-ß1 (TGF-ß1) mediated EMT. Although compartmentalization via A-kinase anchoring proteins (AKAPs) is central to cAMP signaling, functional studies regarding their therapeutic value in the lung EMT process are lacking. The human bronchial epithelial cell line (BEAS-2B) and primary human airway epithelial (pHAE) cells were exposed to TGF-ß1. Epithelial (E-cadherin, ZO-1) and mesenchymal markers (collagen Ӏ, α-SMA, fibronectin) were analyzed (mRNA, protein). ELISA measured TGF-ß1 release. TGF-ß1-sensitive AKAPs Ezrin, AKAP95 and Yotiao were silenced while using siRNA. Cell migration was analyzed by wound healing assay, xCELLigence, Incucyte. Prior to TGF-ß1, dibutyryl-cAMP (dbcAMP), fenoterol, rolipram, cilostamide, and forskolin were used to elevate intracellular cAMP. TGF-ß1 induced morphological changes, decreased E-cadherin, but increased collagen Ӏ and cell migration, a process that was reversed by the inhibitor of δ/epsilon casein kinase I, PF-670462. TGF-ß1 altered (mRNA, protein) expression of Ezrin, AKAP95, and Yotiao. St-Ht31, the AKAP antagonist, decreased E-cadherin (mRNA, protein), but counteracted TGF-ß1-induced collagen Ӏ upregulation. Cigarette smoke (CS) increased TGF-ß1 release, activated TGF signaling, augmented cell migration, and reduced E-cadherin expression, a process that was blocked by TGF-ß1 neutralizing antibody. The silencing of Ezrin, AKAP95, and Yotiao diminished TGF-ß1-induced collagen Ӏ expression, as well as TGF-ß1-induced cell migration. Fenoterol, rolipram, and cilostamide, in AKAP silenced cells, pointed to distinct cAMP compartments. We conclude that Ezrin, AKAP95, and Yotiao promote TGF-ß1-mediated EMT, linked to a TGF-ß1 release by CS. AKAP members might define the ability of fenoterol, rolipram, and cilostamide to modulate the EMT process, and they might represent potential relevant targets in the treatment of COPD.


Subject(s)
A Kinase Anchor Proteins/metabolism , Epithelial-Mesenchymal Transition , Smoking/adverse effects , Transforming Growth Factor beta1/pharmacology , A Kinase Anchor Proteins/genetics , Biomarkers/metabolism , Cadherins/metabolism , Cell Line , Cell Movement/drug effects , Cell Shape/drug effects , Collagen Type I/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Epithelial-Mesenchymal Transition/drug effects , Humans , Phenotype , Signal Transduction/drug effects , Up-Regulation/drug effects
3.
Br J Pharmacol ; 175(14): 2988-3006, 2018 07.
Article in English | MEDLINE | ID: mdl-29722436

ABSTRACT

BACKGROUND AND PURPOSE: cAMP is a central second messenger that broadly regulates cell function and can underpin pathophysiology. In chronic obstructive pulmonary disease, a lung disease primarily provoked by cigarette smoke (CS), the activation of cAMP-dependent pathways, via inhibition of hydrolyzing PDEs, is a major therapeutic strategy. Mechanisms that disrupt cAMP signalling in airway cells, in particular regulation of endogenous PDEs, are poorly understood. EXPERIMENTAL APPROACH: We used a novel Förster resonance energy transfer (FRET) based cAMP biosensor in mice in vivo, ex vivo precision cut lung slices (PCLS) and in human cell models, in vitro, to track the effects of CS exposure. KEY RESULTS: Under fenoterol stimulation, FRET responses to cilostamide were significantly increased in in vivo, ex vivo PCLS exposed to CS and in human airway smooth muscle cells exposed to CS extract. FRET signals to rolipram were only increased in the in vivo CS model. Under basal conditions, FRET responses to cilostamide and rolipram were significantly increased in in vivo, ex vivo PCLS exposed to CS. Elevated FRET signals to rolipram correlated with a protein up-regulation of PDE4 subtypes. In ex vivo PCLS exposed to CS extract, rolipram reversed down-regulation of ciliary beating frequency, whereas only cilostamide significantly increased airway relaxation of methacholine pre-contracted airways. CONCLUSION AND IMPLICATIONS: Exposure to CS, in vitro or in vivo, up-regulated expression and activity of both PDE3 and PDE4, which affected real-time cAMP dynamics. These mechanisms determine the availability of cAMP and can contribute to CS-induced pulmonary pathophysiology.


Subject(s)
Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Smoke , Tobacco Products , Animals , Biosensing Techniques , Cell Line , Epithelial Cells/metabolism , Fluorescence Resonance Energy Transfer , Humans , Mice, Transgenic , Myocytes, Smooth Muscle/metabolism , Phosphodiesterase 3 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/pharmacology , Quinolones/pharmacology , Respiratory System/cytology , Respiratory System/metabolism , Rolipram/pharmacology , Up-Regulation
4.
Oncotarget ; 7(29): 46354-46370, 2016 Jul 19.
Article in English | MEDLINE | ID: mdl-27344171

ABSTRACT

In epithelial cells, ß-catenin is localized at cell-cell junctions where it stabilizes adherens junctions. When these junctions are disrupted, ß-catenin can translocate to the nucleus where it functions as a transcriptional cofactor. Recent research has indicated that PGE2 enhances the nuclear function of ß-catenin through cyclic AMP. Here, we aim to study the role of the cyclic AMP effector Epac in ß-catenin activation by PGE2 in non-small cell lung carcinoma cells. We show that PGE2 induces a down-regulation of E-cadherin, promotes cell migration and enhances ß-catenin translocation to the nucleus. This results in ß-catenin-dependent gene transcription. We also observed increased expression of Epac1. Inhibition of Epac1 activity using the CE3F4 compound or Epac1 siRNA abolished the effects of PGE2 on ß-catenin. Further, we observed that Epac1 and ß-catenin associate together. Expression of an Epac1 mutant with a deletion in the nuclear pore localization sequence prevents this association. Furthermore, the scaffold protein Ezrin was shown to be required to link Epac1 to ß-catenin. This study indicates a novel role for Epac1 in PGE2-induced EMT and subsequent activation of ß-catenin.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Epithelial-Mesenchymal Transition/physiology , Guanine Nucleotide Exchange Factors/metabolism , Lung Neoplasms/pathology , beta Catenin/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Dinoprostone/metabolism , Gene Expression Regulation, Neoplastic/physiology , Humans , Lung Neoplasms/metabolism , Protein Transport , Transcription, Genetic
5.
Sci Rep ; 6: 26928, 2016 05 27.
Article in English | MEDLINE | ID: mdl-27229886

ABSTRACT

COPD is characterized by persistent airflow limitation, neutrophilia and oxidative stress from endogenous and exogenous insults. Current COPD therapy involving anticholinergics, ß2-adrenoceptor agonists and/or corticosteroids, do not specifically target oxidative stress, nor do they reduce chronic pulmonary inflammation and disease progression in all patients. Here, we explore the effects of Sul-121, a novel compound with anti-oxidative capacity, on hyperresponsiveness (AHR) and inflammation in experimental models of COPD. Using a guinea pig model of lipopolysaccharide (LPS)-induced neutrophilia, we demonstrated that Sul-121 inhalation dose-dependently prevented LPS-induced airway neutrophilia (up to ~60%) and AHR (up to ~90%). Non-cartilaginous airways neutrophilia was inversely correlated with blood H2S, and LPS-induced attenuation of blood H2S (~60%) was prevented by Sul-121. Concomitantly, Sul-121 prevented LPS-induced production of the oxidative stress marker, malondialdehyde by ~80%. In immortalized human airway smooth muscle (ASM) cells, Sul-121 dose-dependently prevented cigarette smoke extract-induced IL-8 release parallel with inhibition of nuclear translocation of the NF-κB subunit, p65 (each ~90%). Sul-121 also diminished cellular reactive oxygen species production in ASM cells, and inhibited nuclear translocation of the anti-oxidative response regulator, Nrf2. Our data show that Sul-121 effectively inhibits airway inflammation and AHR in experimental COPD models, prospectively through inhibition of oxidative stress.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/pharmacology , Chromans/pharmacology , Hypersensitivity/prevention & control , Piperazines/pharmacology , Pulmonary Disease, Chronic Obstructive/drug therapy , Reactive Oxygen Species/antagonists & inhibitors , Animals , Cell Line, Transformed , Chromans/chemistry , Complex Mixtures/antagonists & inhibitors , Complex Mixtures/pharmacology , Disease Models, Animal , Gene Expression Regulation , Guinea Pigs , Humans , Hydrogen Sulfide/agonists , Hydrogen Sulfide/blood , Hypersensitivity/etiology , Hypersensitivity/immunology , Hypersensitivity/metabolism , Inflammation , Interleukin-8/antagonists & inhibitors , Interleukin-8/genetics , Interleukin-8/immunology , Lipopolysaccharides/administration & dosage , Lung , Male , Malondialdehyde/antagonists & inhibitors , Malondialdehyde/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/pathology , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/immunology , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/pathology , Oxidative Stress , Piperazines/chemistry , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/physiopathology , Reactive Oxygen Species/metabolism , Tars/chemistry , Tars/toxicity , Transcription Factor RelA/antagonists & inhibitors , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology
6.
Am J Physiol Lung Cell Mol Physiol ; 308(8): L766-75, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25637608

ABSTRACT

ß2-Agonist inhibitors can relieve chronic obstructive pulmonary disease (COPD) symptoms by stimulating cyclic AMP (cAMP) signaling. A-kinase-anchoring proteins (AKAPs) compartmentalize cAMP signaling by establishing protein complexes. We previously reported that the ß2-agonist fenoterol, direct activation of protein kinase A (PKA), and exchange factor directly activated by cAMP decrease cigarette smoke extract (CSE)-induced release of neutrophil attractant interleukin-8 (IL-8) from human airway smooth muscle (ASM) cells. In the present study, we tested the role of AKAPs in CSE-induced IL-8 release from ASM cells and assessed the effect of CSE on the expression levels of different AKAPs. We also studied mRNA and protein expression of AKAPs in lung tissue from patients with COPD. Our data show that CSE exposure of ASM cells decreases AKAP5 and AKAP12, both capable of interacting with ß2-adrenoceptors. In lung tissue of patients with COPD, mRNA levels of AKAP5 and AKAP12 were decreased compared with lung tissue from controls. Using immunohistochemistry, we detected less AKAP5 protein in ASM of patients with COPD Global Initiative for Chronic Obstructive Lung Disease (GOLD) stage II compared with control subjects. St-Ht31, which disrupts AKAP-PKA interactions, augmented CSE-induced IL-8 release from ASM cells and diminished its suppression by fenoterol, an effect mediated by disturbed ERK signaling. The modulatory role of AKAP-PKA interactions in the anti-inflammatory effects of fenoterol in ASM cells and the decrease in expression of AKAP5 and AKAP12 in response to cigarette smoke and in lungs of patients with COPD suggest that cigarette smoke-induced changes in AKAP5 and AKAP12 in patients with COPD may affect efficacy of pharmacotherapy.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cell Cycle Proteins/metabolism , Myocytes, Smooth Muscle/metabolism , Smoking/adverse effects , A Kinase Anchor Proteins/genetics , Aged , Aged, 80 and over , Cell Cycle Proteins/genetics , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Gene Expression , Humans , Interleukin-8/metabolism , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/metabolism
7.
Am J Physiol Cell Physiol ; 306(6): C585-97, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24452374

ABSTRACT

Airway epithelium, which forms the first barrier towards environmental insults, is disturbed by cigarette smoking, a major risk factor for developing chronic obstructive pulmonary disease (COPD). A-kinase anchoring proteins (AKAP) maintain endothelial barrier function and coordinate subcellular localization of protein kinase A (PKA). However, the role of AKAPs in epithelial barrier function is unknown. We studied the role of AKAPs in regulating human bronchial epithelial (Hogg JC, Timens W. Annu Rev Pathol 4: 435-459, 2009; HBE) barrier. Cigarette smoke extract (CSE) reduced barrier function in 16HBE cells and the expression of the adhesion molecule E-cadherin specifically at the cell membrane. In addition, CSE reduced the protein expression of the AKAP family member AKAP9 at the cell membrane. The expression of AKAP5 and AKAP12 was unaffected by CSE. AKAP9 interacted and colocalized with E-cadherin at the cell membrane, suggesting that the reduction of both proteins may be related. Interestingly, disruption of AKAP-PKA interactions by st-Ht31 prevented the CSE-induced reduction of E-cadherin and AKAP9 protein expression and subsequent loss of barrier function. Silencing of AKAP9 reduced the functional epithelial barrier and prevented the ability of st-Ht31 to restore membrane localization of E-cadherin. Our data suggest the possibility of a specific role for AKAP9 in the maintenance of the epithelial barrier. E-cadherin, but not AKAP9, protein expression was reduced in lung tissue from COPD patients compared with controls. However, AKAP9 mRNA expression was decreased in primary bronchial epithelial cells from current smokers compared with non/ex-smokers. In conclusion, our results indicate that AKAP proteins, most likely AKAP9, maintain the bronchial epithelial barrier by regulating the E-cadherin expression at the cell membrane.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cadherins/metabolism , Cytoskeletal Proteins/metabolism , Nicotiana/adverse effects , Respiratory Mucosa/metabolism , Smoke/adverse effects , Smoking/metabolism , A Kinase Anchor Proteins/genetics , Adult , Aged , Aged, 80 and over , Bronchi/drug effects , Bronchi/metabolism , Cadherins/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Cell Membrane/metabolism , Cytoskeletal Proteins/genetics , Female , Humans , Male , Middle Aged , Minor Histocompatibility Antigens , Proto-Oncogene Proteins/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , RNA Interference , RNA, Small Interfering , Respiratory Mucosa/drug effects
8.
Neuropharmacology ; 72: 169-78, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23643754

ABSTRACT

Wistar-Kyoto (WKY) rats are sensitive to chronic stressors and exhibit depression-like behavior. Dorsal raphe nucleus (DRN) serotonin (5-HT) neurons projecting to the prefrontal cortex (PFC) comprise the important neurocircuitry underlying the pathophysiology of depression. To evaluate the DRN-PFC 5-HT system in WKY rats, we examined the effects of escitalopram (ESCIT) on the extracellular 5-HT level in comparison with Wistar rats using dual-probe microdialysis. The basal levels of 5-HT in the DRN, but not in the PFC, in WKY rats was reduced as low as 30% of Wistar rats. Responses of 5-HT in the DRN and PFC to ESCIT administered systemically and locally were attenuated in WKY rats. Feedback inhibition of DRN 5-HT release induced by ESCIT into the PFC was also attenuated in WKY rats. Chronic ESCIT induced upregulation of the DRN-PFC 5-HT system in WKY rats, with increases in basal 5-HT in the DRN, responsiveness to ESCIT in the DRN and PFC, and feedback inhibition, whereas downregulation of these effects was induced in Wistar rats. Thus, the WKY rat is an animal model of depression with low activity of the DRN-PFC 5HT system. The finding that chronic ESCIT upregulates the 5-HT system in hyposerotonergic WKY rats may contribute to improved understanding of mechanisms of action of antidepressants, especially in depression with 5-HT deficiency.


Subject(s)
Citalopram/pharmacology , Prefrontal Cortex/drug effects , Raphe Nuclei/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin/metabolism , Up-Regulation/drug effects , Animals , Anxiety/drug therapy , Anxiety/pathology , Disease Models, Animal , Exploratory Behavior/drug effects , Feeding Behavior/drug effects , Food Preferences/drug effects , Immobility Response, Tonic/drug effects , Male , Motor Activity/drug effects , Neural Pathways/drug effects , Neural Pathways/metabolism , Prefrontal Cortex/metabolism , Raphe Nuclei/metabolism , Rats , Rats, Inbred WKY , Rats, Wistar , Sucrose/administration & dosage , Swimming/psychology
9.
J Renin Angiotensin Aldosterone Syst ; 12(4): 420-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21788250

ABSTRACT

INTRODUCTION: Ace b/l polymorphism in rats is associated with differential tissue angiotensin-converting enzyme (ACE) expression and activity, and susceptibility to renal damage. Same polymorphism was recently found in outbred Wistar rat strain with b allele accounting for higher renal ACE, and provided a model for studying renin-angiotensin-aldosterone system (RAAS) response behind the innate high or low ACE conditions. METHODS: We investigated the reaction of these alleles on chronic angiotensin II (AngII) infusion. Wistar rats were selected to breed male homozygotes for the b (WU-B) or l allele (WU-L) (n = 12). For each allele, one group (n = 6) received AngII infusion via an osmotic minipump (435 ng/kg/min) for 3 weeks. The other group (n = 6) served as a control. RESULTS: WU-B had higher ACE activity at baseline then WU-L. Interestingly, baseline renal ACE2 expression and activity were higher in WU-L. AngII infusion induced the same increase in blood pressure in both genotypes, no proteinuria, but caused tubulo-interstitial renal damage with increased α-SMA and monocyte/macrophage influx only in WU-B (p < 0.05). Low ACE WU-L rats did not develop renal damage. CONCLUSION: AngII infusion causes proteinuria-independent renal damage only in rats with genetically predetermined high ACE while rats with low ACE seemed to be protected against the detrimental effect of AngII. Differences in renal ACE2, mirroring those in ACE, might be involved.


Subject(s)
Alleles , Genetic Predisposition to Disease , Kidney/pathology , Peptidyl-Dipeptidase A/genetics , Angiotensin II , Angiotensin-Converting Enzyme 2 , Animals , Biomarkers/metabolism , Blood Pressure/drug effects , Body Weight/drug effects , Creatinine/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Kidney/drug effects , Kidney/enzymology , Kidney/physiopathology , Kidney Function Tests , Male , Organ Size/drug effects , Peptidyl-Dipeptidase A/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL