Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
FASEB J ; 30(9): 3011-25, 2016 09.
Article in English | MEDLINE | ID: mdl-27178322

ABSTRACT

Chronic Pseudomonas aeruginosa lung infections are associated with progressive epithelial damage and lung function decline. In addition to its role in tissue injury, the persistent presence of P. aeruginosa-secreted products may also affect epithelial repair ability, raising the need for new antivirulence therapies. The purpose of our study was to better understand the outcomes of P. aeruginosa exoproducts exposure on airway epithelial repair processes to identify a strategy to counteract their deleterious effect. We found that P. aeruginosa exoproducts significantly decreased wound healing, migration, and proliferation rates, and impaired the ability of directional migration of primary non-cystic fibrosis (CF) human airway epithelial cells. Impact of exoproducts was inhibited after mutations in P. aeruginosa genes that encoded for the quorum-sensing (QS) transcriptional regulator, LasR, and the elastase, LasB, whereas impact was restored by LasB induction in ΔlasR mutants. P. aeruginosa purified elastase also induced a significant decrease in non-CF epithelial repair, whereas protease inhibition with phosphoramidon prevented the effect of P. aeruginosa exoproducts. Furthermore, treatment of P. aeruginosa cultures with 4-hydroxy-2,5-dimethyl-3(2H)-furanone, a QS inhibitor, abrogated the negative impact of P. aeruginosa exoproducts on airway epithelial repair. Finally, we confirmed our findings in human airway epithelial cells from patients with CF, a disease featuring P. aeruginosa chronic respiratory infection. These data demonstrate that secreted proteases under the control of the LasR QS system impair airway epithelial repair and that QS inhibitors could be of benefit to counteract the deleterious effect of P. aeruginosa in infected patients.-Ruffin, M., Bilodeau, C., Maillé, É., LaFayette, S. L., McKay, G. A., Trinh, N. T. N., Beaudoin, T., Desrosiers, M.-Y., Rousseau, S., Nguyen, D., Brochiero, E. Quorum-sensing inhibition abrogates the deleterious impact of Pseudomonas aeruginosa on airway epithelial repair.


Subject(s)
Bacterial Proteins/metabolism , Epithelial Cells/microbiology , Pseudomonas aeruginosa/physiology , Cell Movement , Cell Proliferation , Cells, Cultured , Gene Expression Regulation, Bacterial/physiology , Humans , Mutation , Respiratory Mucosa/cytology , Respiratory System
2.
PLoS One ; 11(4): e0153665, 2016.
Article in English | MEDLINE | ID: mdl-27092946

ABSTRACT

In epithelia, Cl- channels play a prominent role in fluid and electrolyte transport. Of particular importance is the cAMP-dependent cystic fibrosis transmembrane conductance regulator Cl- channel (CFTR) with mutations of the CFTR encoding gene causing cystic fibrosis. The bulk transepithelial transport of Cl- ions and electrolytes needs however to be coupled to an increase in K+ conductance in order to recycle K+ and maintain an electrical driving force for anion exit across the apical membrane. In several epithelia, this K+ efflux is ensured by K+ channels, including KCa3.1, which is expressed at both the apical and basolateral membranes. We show here for the first time that CFTR and KCa3.1 can physically interact. We first performed a two-hybrid screen to identify which KCa3.1 cytosolic domains might mediate an interaction with CFTR. Our results showed that both the N-terminal fragment M1-M40 of KCa3.1 and part of the KCa3.1 calmodulin binding domain (residues L345-A400) interact with the NBD2 segment (G1237-Y1420) and C- region of CFTR (residues T1387-L1480), respectively. An association of CFTR and F508del-CFTR with KCa3.1 was further confirmed in co-immunoprecipitation experiments demonstrating the formation of immunoprecipitable CFTR/KCa3.1 complexes in CFBE cells. Co-expression of KCa3.1 and CFTR in HEK cells did not impact CFTR expression at the cell surface, and KCa3.1 trafficking appeared independent of CFTR stimulation. Finally, evidence is presented through cross-correlation spectroscopy measurements that KCa3.1 and CFTR colocalize at the plasma membrane and that KCa3.1 channels tend to aggregate consequent to an enhanced interaction with CFTR channels at the plasma membrane following an increase in intracellular Ca2+ concentration. Altogether, these results suggest 1) that the physical interaction KCa3.1/CFTR can occur early during the biogenesis of both proteins and 2) that KCa3.1 and CFTR form a dynamic complex, the formation of which depends on internal Ca2+.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Protein Interaction Maps/physiology , Calcium/metabolism , Cell Line , Cell Membrane/metabolism , Chloride Channels/metabolism , Cystic Fibrosis/metabolism , Epithelial Cells/metabolism , Epithelium/metabolism , Humans , Ion Transport/physiology , Mutation/genetics , Potassium/metabolism , Protein Binding/physiology
3.
Respir Res ; 16: 100, 2015 Sep 04.
Article in English | MEDLINE | ID: mdl-26335442

ABSTRACT

BACKGROUND: Extensive alveolar epithelial injury and remodelling is a common feature of acute lung injury and acute respiratory distress syndrome (ARDS) and it has been established that epithelial regeneration, and secondary lung oedema resorption, is crucial for ARDS resolution. Much evidence indicates that K(+) channels are regulating epithelial repair processes; however, involvement of the KCa3.1 channels in alveolar repair has never been investigated before. RESULTS: Wound-healing assays demonstrated that the repair rates were increased in primary rat alveolar cell monolayers grown on a fibronectin matrix compared to non-coated supports, whereas an anti-ß1-integrin antibody reduced it. KCa3.1 inhibition/silencing impaired the fibronectin-stimulated wound-healing rates, as well as cell migration and proliferation, but had no effect in the absence of coating. We then evaluated a putative relationship between KCa3.1 channel and the migratory machinery protein ß1-integrin, which is activated by fibronectin. Co-immunoprecipitation and immunofluorescence experiments indicated a link between the two proteins and revealed their cellular co-distribution. In addition, we demonstrated that KCa3.1 channel and ß1-integrin membrane expressions were increased on a fibronectin matrix. We also showed increased intracellular calcium concentrations as well as enhanced expression of TRPC4, a voltage-independent calcium channel belonging to the large TRP channel family, on a fibronectin matrix. Finally, wound-healing assays showed additive effects of KCa3.1 and TRPC4 inhibitors on alveolar epithelial repair. CONCLUSION: Taken together, our data demonstrate for the first time complementary roles of KCa3.1 and TRPC4 channels with extracellular matrix and ß1-integrin in the regulation of alveolar repair processes.


Subject(s)
Alveolar Epithelial Cells/metabolism , Integrin beta1/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Pulmonary Alveoli/metabolism , Wound Healing , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/pathology , Animals , Cell Movement , Cell Proliferation , Cells, Cultured , Dose-Response Relationship, Drug , Fibronectins/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Male , Potassium Channel Blockers/pharmacology , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , RNA Interference , Rats, Sprague-Dawley , Signal Transduction , TRPC Cation Channels/antagonists & inhibitors , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism , Time Factors , Transfection , Wound Healing/drug effects
4.
Eur Respir J ; 45(6): 1590-602, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25792634

ABSTRACT

The epithelial response to bacterial airway infection, a common feature of lung diseases such as chronic obstructive pulmonary disease and cystic fibrosis, has been extensively studied. However, its impact on cystic fibrosis transmembrane conductance regulator (CFTR) channel function is not clearly defined. Our aims were, therefore, to evaluate the effect of Pseudomonas aeruginosa on CFTR function and expression in non-cystic fibrosis airway epithelial cells, and to investigate its impact on ΔF508-CFTR rescue by the VRT-325 corrector in cystic fibrosis cells. CFTR expression/maturation was evaluated by immunoblotting and its function by short-circuit current measurements. A 24-h exposure to P. aeruginosa diffusible material (PsaDM) reduced CFTR currents as well as total and membrane protein expression of the wildtype (wt) CFTR protein in CFBE-wt cells. In CFBE-ΔF508 cells, PsaDM severely reduced CFTR maturation and current rescue induced by VRT-325. We also confirmed a deleterious impact of PsaDM on wt-CFTR currents in non-cystic fibrosis primary airway cells as well as on the rescue of ΔF508-CFTR function induced by VRT-325 in primary cystic fibrosis cells. These findings show that CFTR function could be impaired in non-cystic fibrosis patients infected by P. aeruginosa. Our data also suggest that CFTR corrector efficiency may be affected by infectious components, which should be taken into account in screening assays of correctors.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/metabolism , Epithelial Cells/metabolism , Pseudomonas Infections/metabolism , RNA, Messenger/metabolism , Respiratory Mucosa/metabolism , Cells, Cultured , Cystic Fibrosis/complications , Cystic Fibrosis/microbiology , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Epithelial Cells/microbiology , Humans , Piperazines/pharmacology , Pseudomonas Infections/complications , Pseudomonas aeruginosa , Quinazolines/pharmacology , Respiratory Mucosa/cytology , Respiratory Mucosa/microbiology , Young Adult
5.
Int J Oncol ; 44(3): 838-48, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24366043

ABSTRACT

K+ channels, which are overexpressed in several cancers, have been identified as regulators of cell proliferation and migration, key processes in cancer development/propagation. Their role in lung cancer has not been studied extensively; but we showed previously that KvLQT1 channels are involved in cell migration, proliferation and repair of normal lung epithelial cells. We now investigated the role of these channels in lung cancer cell lines and their expression levels in human lung adenocarcinoma (AD) tissues. First, we observed that the wound-healing rates of A549 lung adenocarcinoma cell monolayers were reduced by clofilium and chromanol or after silencing with KvLQT1-specific siRNA. Dose-dependent decrease of A549 cell growth and cell accumulation in G0/G1 phase were seen after KvLQT1 inhibition. Clofilium also affected 2D and 3D migration of A549 cells. Similarly, H460 cell growth, migration and wound healing were diminished by this drug. Because K+ channel overexpression has been encountered in some cancers, we assessed KvLQT1 expression levels in tumor tissues from patients with lung AD. KvLQT1 protein expression in tumor samples was increased by 1.5- to 7-fold, compared to paired non-neoplastic tissues, in 17 of 26 patients. In summary, our data reveal that KvLQT1 channel blockade efficiently reduces A549 and H460 cell proliferation and migration. Moreover, KvLQT1 overexpression in AD samples suggests that it could be a potential therapeutic target in lung cancer.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Non-Small-Cell Lung/genetics , KCNQ Potassium Channels/genetics , Lung Neoplasms/genetics , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Carcinogenesis , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , KCNQ Potassium Channels/biosynthesis , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Quaternary Ammonium Compounds/administration & dosage , RNA, Small Interfering/genetics , Wound Healing/genetics
6.
Blood ; 120(13): 2745-56, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-22898603

ABSTRACT

DEP-1/CD148 is a receptor-like protein tyrosine phosphatase with antiproliferative and tumor-suppressive functions. Interestingly, it also positively regulates Src family kinases in hematopoietic and endothelial cells, where we showed it promotes VE-cadherin-associated Src activation and endothelial cell survival upon VEGF stimulation. However, the molecular mechanism involved and its biologic functions in endothelial cells remain ill-defined. We demonstrate here that DEP-1 is phosphorylated in a Src- and Fyn-dependent manner on Y1311 and Y1320, which bind the Src SH2 domain. This allows DEP-1-catalyzed dephosphorylation of Src inhibitory Y529 and favors the VEGF-induced phosphorylation of Src substrates VE-cadherin and Cortactin. Accordingly, RNA interference (RNAi)-mediated knockdown of DEP-1 or expression of DEP-1 Y1311F/Y1320F impairs Src-dependent biologic responses mediated by VEGF including permeability, invasion, and branching capillary formation. In addition, our work further reveals that above a threshold expression level, DEP-1 can also dephosphorylate Src Y418 and attenuate downstream signaling and biologic responses, consistent with the quiescent behavior of confluent endothelial cells that express the highest levels of endogenous DEP-1. Collectively, our findings identify the VEGF-dependent phosphorylation of DEP-1 as a novel mechanism controlling Src activation, and show this is essential for the proper regulation of permeability and the promotion of the angiogenic response.


Subject(s)
Capillaries/metabolism , Cell Membrane Permeability , Endothelium, Vascular/cytology , Neovascularization, Pathologic , Tyrosine/metabolism , src-Family Kinases/metabolism , Antigens, CD/metabolism , Blotting, Western , Cadherins/metabolism , Cell Adhesion , Cell Movement , Cell Proliferation , Cells, Cultured , Cortactin/metabolism , Endothelium, Vascular/metabolism , Fluorescent Antibody Technique , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immunoprecipitation , Mutation/genetics , Neoplasm Invasiveness , Phosphorylation , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
7.
Eur Respir J ; 40(6): 1390-400, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22496330

ABSTRACT

Airway damage and remodelling are important components of lung pathology progression in cystic fibrosis (CF). Although repair mechanisms are engaged to restore the epithelial integrity, these processes are obviously insufficient to maintain lung function in CF airways. Our aims were therefore to study how the basic cystic fibrosis transmembrane conductance regulator (CFTR) defect could impact epithelial wound healing and to determine if CFTR correction could improve it. Wound-healing experiments, as well as cell migration and proliferation assays, were performed to study the early phases of epithelial repair in human CF and non-CF airway cells. CFTR function was evaluated using CFTR small interferring (si)RNA and inhibitor GlyH101 in non-CF cells, and conversely after CFTR rescue with the CFTR corrector VRT-325 in CF cells. Wound-healing experiments first showed that airway cells from CF patients repaired slower than non-CF cells. CFTR inhibition or silencing in non-CF primary airway cells significantly inhibited wound closure. GlyH101 also decreased cell migration and proliferation. Interestingly, wild-type CFTR transduction in CF airway cell lines or CFTR correction with VRT-325 in CFBE-ΔF508 and primary CF bronchial monolayers significantly improved wound healing. Altogether our results demonstrated that functional CFTR plays a critical role in wound repair, and CFTR correction may represent a novel strategy to promote the airway repair processes in CF.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/metabolism , Adult , Bronchi/metabolism , Cell Line , Cell Movement , Cell Proliferation , Cells, Cultured , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelium/metabolism , Humans , Lung/microbiology , Lung Diseases/microbiology , Lung Transplantation/methods , Mutation , RNA, Small Interfering/metabolism , Regeneration , Respiratory Mucosa/metabolism , Time Factors , Wound Healing
8.
Am J Physiol Lung Cell Mol Physiol ; 301(6): L945-55, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21908587

ABSTRACT

Chronic infection and inflammation have been associated with progressive airway epithelial damage in patients with cystic fibrosis (CF). However, the effect of inflammatory products on the repair capacity of respiratory epithelia is unclear. Our objective was to study the regulation of repair mechanisms by tumor necrosis factor-α (TNF-α), a major component of inflammation in CF, in a model of mechanical wounding, in two bronchial cell lines, non-CF NuLi and CF CuFi. We observed that TNF-α enhanced the NuLi and CuFi repair rates. Chronic exposure (24-48 h) to TNF-α augmented this stimulation as well as the migration rate during repair. The cellular mechanisms involved in this stimulation were then evaluated. First, we discerned that TNF-α induced metalloproteinase-9 release, epidermal growth factor (EGF) shedding, and subsequent EGF receptor transactivation. Second, TNF-α-induced stimulation of the NuLi and CuFi wound-closure rates was prevented by GM6001 (metalloproteinase inhibitor), EGF antibody (to titrate secreted EGF), and EGF receptor tyrosine kinase inhibitors. Furthermore, we recently reported a relationship between the EGF response and K(+) channel function, both controlling bronchial repair. We now show that TNF-α enhances KvLQT1 and K(ATP) currents, while their inhibition abolishes TNF-α-induced repair stimulation. These results indicate that the effect of TNF-α is mediated, at least in part, through EGF receptor transactivation and K(+) channel stimulation. In contrast, cell proliferation during repair was slowed by TNF-α, suggesting that TNF-α could exert contrasting actions on repair mechanisms of CF airway epithelia. Finally, the stimulatory effect of TNF-α on airway wound repair was confirmed on primary airway epithelial cells, from non-CF and CF patients.


Subject(s)
Cystic Fibrosis/pathology , Epithelial Cells/pathology , Tumor Necrosis Factor-alpha/pharmacology , Bronchioles/metabolism , Bronchioles/pathology , Cell Movement , Cell Proliferation/drug effects , Cells, Cultured , Cystic Fibrosis/metabolism , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Epidermal Growth Factor/physiology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , ErbB Receptors/agonists , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Membrane Potentials , Phosphorylation , Potassium Channel Blockers/pharmacology , Potassium Channels/metabolism , Primary Cell Culture , Quaternary Ammonium Compounds/pharmacology , Transcriptional Activation , Tumor Necrosis Factor-alpha/physiology
9.
Med Sci (Paris) ; 25(4): 391-7, 2009 Apr.
Article in French | MEDLINE | ID: mdl-19409192

ABSTRACT

Transcripts of more than 30 different K(+) channels have been detected in the respiratory epithelium lining airways and alveoli. These channels belong to the 3 main classes of K(+) channels, i.e. i) voltage-dependent or calcium-activated, 6 transmembrane segments (TM), ii) 2-pores 4-TM and iii) inward-rectified 2-TM channels. The physiological and functional significance of this high molecular diversity of lung epithelial K(+) channels is not well understood. Surprisingly, relatively few studies are focused on K(+) channel function in lung epithelial physiology. Nevertheless, several studies have shown that KvLQT1, KCa and K(ATP) K(+) channels play a crucial role in ion and fluid transport, contributing to the control of airway and alveolar surface liquid composition and volume. K(+) channels are involved in other key functions, such as O(2) sensing or the capacity of the respiratory epithelia to repair after injury. This mini-review aims to discuss potential functions of lung K(+) channels.


Subject(s)
Lung/cytology , Potassium Channels/physiology , Potassium/physiology , Animals , Biological Transport , Body Fluids/metabolism , Epithelial Cells/metabolism , Epithelial Cells/physiology , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Ion Transport/physiology , Lung/metabolism , Lung Diseases/metabolism , Pneumonia/metabolism , Potassium/metabolism , Potassium Channels/classification , Potassium Channels/genetics , Pulmonary Gas Exchange/physiology , Pulmonary Surfactants/metabolism , Regeneration/physiology , Signal Transduction/physiology
10.
Am J Physiol Lung Cell Mol Physiol ; 296(2): L145-55, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19060226

ABSTRACT

Multiple K(+) channels are expressed in the respiratory epithelium lining airways and alveoli. Of the three main classes [1) voltage-dependent or Ca(2+)-activated, 6-transmembrane domains (TMD), 2) 2-pores 4-TMD, and 3) inward-rectified 2-TMD K(+) channels], almost 40 different transcripts have already been detected in the lung. The physiological and functional significance of this high molecular diversity of lung epithelial K(+) channels is intriguing. As detailed in the present review, K(+) channels are located at both the apical and basolateral membranes in the respiratory epithelium, where they mediate K(+) currents of diverse electrophysiological and regulatory properties. The main recognized function of K(+) channels is to control membrane potential and to maintain the driving force for transepithelial ion and liquid transport. In this manner, KvLQT1, KCa and K(ATP) channels, for example, contribute to the control of airway and alveolar surface liquid composition and volume. Thus, K(+) channel activation has been identified as a potential therapeutic strategy for the resolution of pathologies characterized by ion transport dysfunction. K(+) channels are also involved in other key functions in lung physiology, such as oxygen-sensing, inflammatory responses and respiratory epithelia repair after injury. The purpose of this review is to summarize and discuss what is presently known about the molecular identity of lung K(+) channels with emphasis on their role in lung epithelial physiology.


Subject(s)
Potassium Channels/physiology , Pulmonary Alveoli/physiology , Respiratory Mucosa/physiology , Animals , Humans
11.
Am J Physiol Cell Physiol ; 296(2): C285-95, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19052260

ABSTRACT

The vectorial transport of ions and water across epithelial cells depends to a large extent on the coordination of the apical and basolateral ion fluxes with energy supply. In this work we provide the first evidence for a regulation by the 5'-AMP-activated protein kinase (AMPK) of the calcium-activated potassium channel KCa3.1 expressed at the basolateral membrane of a large variety of epithelial cells. Inside-out patch-clamp experiments performed on human embryonic kidney (HEK) cells stably transfected with KCa3.1 first revealed a decrease in KCa3.1 activity following the internal addition of AMP at a fixed ATP concentration. This effect was dose dependent with half inhibition at 140 muM AMP in 1 mM ATP. Evidence for an interaction between the COOH-terminal region of KCa3.1 and the gamma1-subunit of AMPK was next obtained by two-hybrid screening and pull-down experiments. Our two-hybrid analysis confirmed in addition that the amino acids extending from Asp(380) to Ala(400) in COOH-terminal were essential for the interaction AMPK-gamma1/KCa3.1. Inside-out experiments on cells coexpressing KCa3.1 with the dominant negative AMPK-gamma1-R299G mutant showed a reduced sensitivity of KCa3.1 to AMP, arguing for a functional link between KCa3.1 and the gamma1-subunit of AMPK. More importantly, coimmunoprecipitation experiments carried out on bronchial epithelial NuLi cells provided direct evidence for the formation of a KCa3.1/AMPK-gamma1 complex at endogenous AMPK and KCa3.1 expression levels. Finally, treating NuLi monolayers with the membrane permeant AMPK activator 5-aminoimidazole-4-carboxamide-1-beta-d-ribofuranoside (AICAR) caused a significant decrease of the KCa3.1-mediated short-circuit currents, an effect reversible by coincubation with the AMPK inhibitor Compound C. These observations argue for a regulation of KCa3.1 by AMPK in a functional epithelium through protein/protein interactions involving the gamma1-subunit of AMPK.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Epithelial Cells/enzymology , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Ion Channel Gating , Respiratory Mucosa/enzymology , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Cell Polarity , Cells, Cultured , Enzyme Activation , Enzyme Activators/pharmacology , Epithelial Cells/drug effects , Humans , Immunoprecipitation , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Ion Channel Gating/drug effects , Ion Transport , Membrane Potentials , Mutation , Patch-Clamp Techniques , Protein Binding , Recombinant Proteins/metabolism , Respiratory Mucosa/drug effects , Ribonucleotides/pharmacology , Transfection , Two-Hybrid System Techniques
12.
Am J Physiol Lung Cell Mol Physiol ; 295(5): L866-80, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18757521

ABSTRACT

Severe lesions of airway epithelia are observed in cystic fibrosis (CF) patients. The regulatory mechanisms of cell migration and proliferation processes, involved in the repair of injured epithelia, then need to be better understood. A model of mechanical wounding of non-CF (NuLi) and CF (CuFi) bronchial monolayers was employed to study the repair mechanisms. We first observed that wound repair, under paracrine and autocrine EGF control, was slower (up to 33%) in CuFi than in NuLi. Furthermore, EGF receptor (EGFR) activation, following wounding, was lower in CuFi than in NuLi monolayers. Cell proliferation and migration assays indicated a similar rate of proliferation in both cell lines but with reduced (by 25%) CuFi cell migration. In addition, cell migration experiments performed in the presence of conditioned medium, collected from NuLi and CuFi wounded bronchial monolayers, suggested a defect in EGF/EGFR signaling in CF cells. We (49) recently demonstrated coupling between the EGF response and K(+) channel function, which is crucial for EGF-stimulated alveolar repair. In CuFi cells, lower EGF/EGFR signaling was accompanied by a 40-70% reduction in K(+) currents and KvLQT1, ATP-sensitive potassium (K(ATP)), and Ca(2+)-activated K(+) (KCa3.1) channel expression. In addition, EGF-stimulated bronchial wound healing, cell migration, and proliferation were severely decreased by K(+) channel inhibitors. Finally, acute CFTR inhibition failed to reduce wound healing, EGF secretion, and K(+) channel expression in NuLi. In summary, the delay in CuFi wound healing could be due to diminished EGFR signaling coupled with lower K(+) channel function, which play a crucial role in bronchial repair.


Subject(s)
Bronchi/pathology , Cystic Fibrosis/pathology , Epidermal Growth Factor/metabolism , Epithelium/pathology , Potassium Channels/metabolism , Wound Healing , Bronchi/metabolism , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Epidermal Growth Factor/pharmacology , Epithelium/drug effects , ErbB Receptors/metabolism , Humans , Ion Channel Gating/drug effects , Wound Healing/drug effects
13.
Am J Physiol Lung Cell Mol Physiol ; 293(4): L870-82, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17631610

ABSTRACT

Several respiratory diseases are associated with extensive damage of lung epithelia, and the regulatory mechanisms involved in their regeneration are not clearly defined. Growth factors released by epithelial cells or fibroblasts from injured lungs are important regulators of alveolar repair by stimulating cell motility, proliferation, and differentiation. In addition, K(+) channels regulate cell proliferation/migration and are coupled with growth factor signaling in several tissues. We decided to explore the hypothesis, never investigated before, that K(+) could play a prominent role in alveolar repair. We employed a model of mechanical wounding of rat alveolar type II epithelia, in primary culture, to study their response to injury. Wound healing was suppressed by one-half upon epidermal growth factor (EGF) titration with EGF-antibody (Ab) or erbB1/erbB2 tyrosine-kinase inhibition with AG-1478/AG-825. The addition of exogenous EGF slightly stimulated the alveolar wound healing and enhanced, by up to five times, alveolar cell migration measured in a Boyden-type chamber. Conditioned medium collected from injured alveolar monolayers also stimulated cell migration; this effect was abolished in the presence of EGF-Ab. The impact of K(+) channel modulators was examined in basal and EGF-stimulated conditions. Wound healing was stimulated by pinacidil, an ATP-dependent K(+) channel (K(ATP)) activator, which also increased cell migration, by twofold, in basal conditions and potentiated the stimulatory effect of EGF. K(ATP) or KvLQT1 inhibitors (glibenclamide, clofilium) reduced EGF-stimulated wound healing, cell migration, and proliferation. Finally, EGF stimulated K(ATP) and KvLQT1 currents and channel expression. In summary, stimulation of K(+) channels through autocrine activation of EGF receptors could play a crucial role in lung epithelia repair processes.


Subject(s)
Adenosine Triphosphate/metabolism , Epidermal Growth Factor/metabolism , KCNQ1 Potassium Channel/metabolism , Potassium Channels/metabolism , Pulmonary Alveoli/injuries , Wound Healing , Animals , Antibodies/pharmacology , Autocrine Communication , Benzothiazoles/pharmacology , Biological Transport/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Drug Synergism , Electric Conductivity , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/immunology , Epidermal Growth Factor/pharmacology , Epithelial Cells/pathology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , KCNQ1 Potassium Channel/drug effects , Male , Pinacidil/pharmacology , Potassium/metabolism , Potassium Channels/drug effects , Protein Kinase Inhibitors/pharmacology , Pulmonary Alveoli/pathology , Pulmonary Alveoli/physiopathology , Quinazolines , Rats , Rats, Sprague-Dawley , Receptor, ErbB-2/antagonists & inhibitors , Tyrphostins/pharmacology , Wound Healing/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...