Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
JCI Insight ; 5(16)2020 08 20.
Article in English | MEDLINE | ID: mdl-32814712

ABSTRACT

Airway mucociliary clearance (MCC) is the main mechanism of lung defense keeping airways free of infection and mucus obstruction. Airway surface liquid volume, ciliary beating, and mucus are central for proper MCC and critically regulated by sodium absorption and anion secretion. Impaired MCC is a key feature of muco-obstructive diseases. The calcium-activated potassium channel KCa.3.1, encoded by Kcnn4, participates in ion secretion, and studies showed that its activation increases Na+ absorption in airway epithelia, suggesting that KCa3.1-induced hyperpolarization was sufficient to drive Na+ absorption. However, its role in airway epithelium is not fully understood. We aimed to elucidate the role of KCa3.1 in MCC using a genetically engineered mouse. KCa3.1 inhibition reduced Na+ absorption in mouse and human airway epithelium. Furthermore, the genetic deletion of Kcnn4 enhanced cilia beating frequency and MCC ex vivo and in vivo. Kcnn4 silencing in the Scnn1b-transgenic mouse (Scnn1btg/+), a model of muco-obstructive lung disease triggered by increased epithelial Na+ absorption, improved MCC, reduced Na+ absorption, and did not change the amount of mucus but did reduce mucus adhesion, neutrophil infiltration, and emphysema. Our data support that KCa3.1 inhibition attenuated muco-obstructive disease in the Scnn1btg/+ mice. K+ channel modulation may be a therapeutic strategy to treat muco-obstructive lung diseases.


Subject(s)
Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Lung Diseases, Obstructive/etiology , Mucociliary Clearance/physiology , Animals , Calcium/metabolism , Cells, Cultured , Cilia/drug effects , Cilia/metabolism , Disease Models, Animal , Epithelium/metabolism , Female , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Lung/physiopathology , Lung Diseases, Obstructive/genetics , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Mucociliary Clearance/drug effects , Sodium/metabolism
2.
Sci Rep ; 8(1): 2608, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29422673

ABSTRACT

Anion selective ionophores, anionophores, are small molecules capable of facilitating the transmembrane transport of anions. Inspired in the structure of natural product prodigiosin, four novel anionophores 1a-d, including a 1,2,3-triazole group, were prepared. These compounds proved highly efficient anion exchangers in model phospholipid liposomes. The changes in the hydrogen bond cleft modified the anion transport selectivity exhibited by these compounds compared to prodigiosin and suppressed the characteristic high toxicity of the natural product. Their activity as anionophores in living cells was studied and chloride efflux and iodine influx from living cells mediated by these derivatives was demonstrated. These compounds were shown to permeabilize cellular membranes to halides with efficiencies close to the natural anion channel CFTR at doses that do not compromise cellular viability. Remarkably, optimal transport efficiency was measured in the presence of pH gradients mimicking those found in the airway epithelia of Cystic Fibrosis patients. These results support the viability of developing small molecule anionophores as anion channel protein surrogates with potential applications in the treatment of conditions such as Cystic Fibrosis derived from the malfunction of natural anion transport mechanisms.


Subject(s)
Cell Membrane Permeability/drug effects , Cell Membrane/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/metabolism , Ionophores/pharmacology , Animals , Anions/metabolism , Cell Membrane/metabolism , Humans , Ion Transport/drug effects , Ionophores/chemical synthesis , Ionophores/chemistry , Prodigiosin/chemistry , Tumor Cells, Cultured
3.
Hum Mutat ; 38(7): 849-862, 2017 07.
Article in English | MEDLINE | ID: mdl-28477385

ABSTRACT

The transfer of genomic information into the primary RNA sequence can be altered by RNA editing. We have previously shown that genomic variants can be RNA-edited to wild-type. The presence of distinct "edited" iduronate 2-sulfatase (IDS) mRNA transcripts ex vivo evidenced the correction of a nonsense and frameshift variant, respectively, in three unrelated Hunter syndrome patients. This phenomenon was confirmed in various patient samples by a variety of techniques, and was quantified by single-nucleotide primer extension. Western blotting also confirmed the presence of IDS protein similar in size to the wild-type. Since preliminary experimental evidence suggested that the "corrected" IDS proteins produced by the patients were similar in molecular weight and net charge to their wild-type counterparts, an in vitro system employing different cell types was established to recapitulate the site-specific editing of IDS RNA (uridine to cytidine conversion and uridine deletion), and to confirm the findings previously observed ex vivo in the three patients. In addition, confocal microscopy and flow cytometry analyses demonstrated the expression and lysosomal localization in HEK293 cells of GFP-labeled proteins translated from edited IDS mRNAs. Confocal high-content analysis of the two patients' cells expressing wild-type or mutated IDS confirmed lysosomal localization and showed no accumulation in the Golgi or early endosomes.


Subject(s)
Glycoproteins/genetics , Mucopolysaccharidosis II/genetics , Mutation , RNA, Messenger/genetics , Base Sequence , Codon, Nonsense , Computational Biology , Exons , Frameshift Mutation , Genetic Variation , Genetic Vectors , Genome, Human , Golgi Apparatus/metabolism , HEK293 Cells , HeLa Cells , Hemizygote , Humans , Lysosomes/metabolism , Male , Protein Biosynthesis , RNA Editing
4.
Cell Mol Life Sci ; 74(1): 117-128, 2017 01.
Article in English | MEDLINE | ID: mdl-27704174

ABSTRACT

CFTR protein is an ion channel regulated by cAMP-dependent phosphorylation and expressed in many types of epithelial cells. CFTR-mediated chloride and bicarbonate secretion play an important role in the respiratory and gastrointestinal systems. Pharmacological modulators of CFTR represent promising drugs for a variety of diseases. In particular, correctors and potentiators may restore the activity of CFTR in cystic fibrosis patients. Potentiators are also potentially useful to improve mucociliary clearance in patients with chronic obstructive pulmonary disease. On the other hand, CFTR inhibitors may be useful to block fluid and electrolyte loss in secretory diarrhea and slow down the progression of polycystic kidney disease.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/agonists , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Cystic Fibrosis/drug therapy , Drug Discovery/methods , Animals , Bicarbonates/metabolism , Chlorides/metabolism , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Cells/metabolism , Humans , Mucociliary Clearance/drug effects , Mutation
5.
Eur J Pharmacol ; 781: 100-8, 2016 Jun 15.
Article in English | MEDLINE | ID: mdl-27063443

ABSTRACT

Defective epithelial chloride secretion occurs in humans with cystic fibrosis (CF), a genetic defect due to loss of function of CFTR, a cAMP-activated chloride channel. In the airways, absence of an active CFTR causes a severe lung disease. In mice, genetic ablation of CFTR function does not result in similar lung pathology. This may be due to the expression of an alternative chloride channel which is activated by calcium. The most probable protein performing this function is TMEM16A, a calcium-activated chloride channel (CaCC). Our aim was to assess the relative contribution of CFTR and TMEM16A to chloride secretion in adult mouse trachea. For this purpose we tested pharmacological inhibitors of chloride channels in normal and CF mice. The amplitude of the cAMP-activated current was similar in both types of animals and was not affected by a selective CFTR inhibitor. In contrast, a CaCC inhibitor (CaCCinh-A01) strongly blocked the cAMP-activated current as well as the calcium-activated chloride secretion triggered by apical UTP. Although control experiments revealed that CaCCinh-A01 also shows inhibitory activity on CFTR, our results indicate that transepithelial chloride secretion in adult mouse trachea is independent of CFTR and that another channel, possibly TMEM16A, performs both cAMP- and calcium-activated chloride transport. The prevalent function of a non-CFTR channel may explain the absence of a defect in chloride transport in CF mice.


Subject(s)
Bronchi/cytology , Chlorides/metabolism , Animals , Anoctamin-1 , Biological Transport/drug effects , Calcium/metabolism , Chloride Channels/antagonists & inhibitors , Cyclic AMP/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelium/drug effects , Epithelium/metabolism , Humans , Male , Mice
6.
J Cyst Fibros ; 15(4): 425-35, 2016 07.
Article in English | MEDLINE | ID: mdl-26971626

ABSTRACT

BACKGROUND: Mistrafficking of CFTR protein caused by F508del, the most frequent mutation in cystic fibrosis (CF), can be corrected by cell incubation at low temperature, an effect that may be mediated by altered expression of proteostasis genes. METHODS: To identify small molecules mimicking low temperature, we compared gene expression profiles of cells kept at 27°C with those previously generated from more than 1300 compounds. The resulting candidates were tested with a functional assay on a bronchial epithelial cell line. RESULTS: We found that anti-inflammatory glucocorticoids, such as mometasone, budesonide, and fluticasone, increased mutant CFTR function. However, this activity was not confirmed in primary bronchial epithelial cells. Actually, glucocorticoids enhanced Na(+) absorption, an effect that could further impair mucociliary clearance in CF airways. CONCLUSIONS: Our results suggest that rescue of F508del-CFTR by low temperature cannot be easily mimicked by small molecules and that compounds with closer transcriptional and functional effects need to be found.


Subject(s)
Chloride Channel Agonists/pharmacology , Chloride Channels/physiology , Cold Temperature , Cystic Fibrosis Transmembrane Conductance Regulator , Cystic Fibrosis , Epithelial Cells/metabolism , Bronchi/pathology , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis/therapy , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Drug Repositioning/methods , Humans , Mucociliary Clearance/physiology , Mutant Proteins/metabolism , Mutation , Systems Biology/methods , Transcriptome/physiology
7.
J Struct Biol ; 194(1): 102-11, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26850167

ABSTRACT

CFTR is an anionic channel expressed in epithelia whose mutations cause cystic fibrosis. Wild (WT) and mutated (F508del) types were over-expressed in yeast, solubilised in the detergent LPG-14 and purified. The detergent-CFTR complexes were studied by SAXS techniques using a solvent of variable density. The final result of the study is the numerical value of a set of parameters: molecular mass, volume and radius of gyration, average electron density and second moment of the electron density fluctuations inside the particles. It is also shown that in the complex the centres of gravity of CFTR and of the detergent are displaced relative to each other. The analysis of these parameters led to the determination of the size and shape of the volumes occupied by protein and by detergent in the complex. WT-CFTR to be an elongated molecule (maximum diameter ∼12.4nm) which spans a flat detergent micelle. The distance distribution function, P(r) confirms that the WT-CFTR is elongated and with an inhomogeneous electronic density. The F508del-CFTR molecule is also elongated (maximum diameter ∼13.2nm), but the associated detergent micelle hides a larger surface, plausibly related to an increased exposure of hydrophobic portions of the mutated protein. The corresponding P(r) is consistent with the presence of well defined domains, probably linked by flexible regions. These differences suggest that the full-length mutant F508del-CFTR has a detectably different conformation, in contrast to the minor differences observed for the isolated F508-containing domain. We interpret the data in terms of an incomplete post-translational assembly of the protein domains.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Detergents/chemistry , Scattering, Small Angle , X-Ray Diffraction/methods , Algorithms , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Humans , Kinetics , Mutation , Protein Conformation
8.
J Aerosol Med Pulm Drug Deliv ; 29(4): 337-45, 2016 08.
Article in English | MEDLINE | ID: mdl-26741302

ABSTRACT

BACKGROUND: Cystic fibrosis (CF) is characterized by a thick, sticky mucus responsible for both airway obstruction and resistance to drug diffusion, reducing the effectiveness of drug delivery to the lung. Studies of drug-mucus interaction may be a crucial step in therapeutic management of CF. In the present research, the effect of a saline solution of sodium bicarbonate (100 mM) on sputum viscosity and the permeation properties of ketoprofen lysinate (Klys) from a previously developed dry powder inhaler were evaluated. METHODS: Rheological measurements were performed using an ARES rotational rheometer (Rheometrics, Inc.) with a parallel plate geometry. The gel fraction, separated from the liquid phase of various sputum samples from CF patients was loaded onto the plate. The elastic (G') and the viscous (G") moduli, tan δ (ratio of G" to G') and η* (complex viscosity) were evaluated as frequency-dependent parameters. Drug permeation across CF sputum from dry powders was studied by means of Franz-type vertical diffusion cells. The experiments were conducted on untreated sputum and on sputum treated with bicarbonate. RESULTS: Rheological studies showed that the elastic modulus (G') was always greater than the viscous modulus (G") and the viscosity decreased with increasing frequency, as for pseudo-plastic fluids. Bicarbonate caused a downward shift of both the elastic and viscous moduli, with a reduction in complex viscosity. As to drug permeation, the untreated sputum slowed down drug dissolution and permeation compared to buffer permeability (control). Permeation studies across CF sputum treated with bicarbonate showed higher Klys dissolution/permeation than untreated sputum. CONCLUSIONS: The interesting results confirm the previously reported bicarbonate. effectiveness in CF; this weak base seems to act by decreasing high viscosity of the CF bronchial secretion and, potentially, resulting in better mucus clearance and in fighting pulmonary infections.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/metabolism , Bronchi/metabolism , Cystic Fibrosis/physiopathology , Ketoprofen/analogs & derivatives , Lysine/analogs & derivatives , Sodium Bicarbonate/pharmacology , Sputum/drug effects , Administration, Inhalation , Adolescent , Adult , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Dry Powder Inhalers , Elastic Modulus , Female , Humans , Ketoprofen/administration & dosage , Ketoprofen/metabolism , Lysine/administration & dosage , Lysine/metabolism , Male , Middle Aged , Models, Biological , Permeability , Rheology , Sputum/metabolism , Viscosity , Young Adult
9.
J Cyst Fibros ; 15(3): 295-301, 2016 05.
Article in English | MEDLINE | ID: mdl-26677762

ABSTRACT

BACKGROUND: In CF patients, the defective ion transport causes a simultaneous reduction of fluid, Cl(-) and HCO3(-) secretion. We aimed to demonstrate that the resulting altered properties of mucus can be recovered using lumacaftor, a CFTR corrector. METHODS: The micro-rheology of non-CF and CF mucus was analysed using Multiple Particle Tracking. RESULTS: The diffusion coefficient of nano-beads imbedded in mucus from CF human bronchial epithelium was lower than in non-CF mucus, and the elastic and viscous moduli were higher. We found that 25% correction of F508del-CFTR mutation with lumacaftor was enough to improve significantly CF mucus properties. Surprisingly, also incubation with amiloride, a compound that reduces fluid absorption but might not change the secretion of HCO3(-) towards the airway surface fluid, improved CF mucus properties. CONCLUSION: CF mucus properties can be recovered by either improving the hydration of the airways or recovering Cl(-) and HCO3(-) secretion across the mutated protein treated with a corrector compound.


Subject(s)
Amiloride , Aminopyridines , Benzodioxoles , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis , Mucus , Respiratory Mucosa , Amiloride/administration & dosage , Amiloride/pharmacokinetics , Aminopyridines/administration & dosage , Aminopyridines/pharmacokinetics , Benzodioxoles/administration & dosage , Benzodioxoles/pharmacokinetics , Biological Availability , Cell Culture Techniques , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Epithelial Sodium Channel Blockers/administration & dosage , Epithelial Sodium Channel Blockers/pharmacokinetics , Humans , Microfluidics/methods , Models, Theoretical , Mucus/drug effects , Mucus/metabolism , Mutant Proteins/genetics , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism
10.
Eur J Med Chem ; 99: 14-35, 2015 Jun 24.
Article in English | MEDLINE | ID: mdl-26041577

ABSTRACT

The cystic fibrosis transmembrane conductance regulator (CFTR) is a chloride channel present in the membrane of epithelial cells. Mutations affecting the CFTR gene cause cystic fibrosis (CF), a multi-organ severe disease. The most common CF mutation, F508del, impairs the processing and activity (gating) of CFTR protein. Other mutations, like G551D, only cause a gating defect. Processing and gating defects can be targeted by small molecules called generically correctors and potentiators, respectively. Aminoarylthiazoles (AATs) represent an interesting class of compounds that includes molecules with dual activity, as correctors and potentiators. With the aim to improve the activity profile of AATs, we have now designed and synthesized a library of novel compounds in order to establish an initial SAR that may provide indications about the chemical groups that are beneficial or detrimental for rescue activity. The new compounds were tested as correctors and potentiators in CFBE41o-expressing F508del-CFTR using a functional assay. A dual active compound, AAT-4a, characterized by improved efficacy and marked synergy when combined with the corrector VX-809 has been identified. Moreover, by computational methods, a possible binding site for AATs in nucleotide binding domain NBD1 has been detected. These results will direct the synthesis of new analogues with possibly improved activity.


Subject(s)
Chlorides/metabolism , Cystic Fibrosis/metabolism , Drug Design , Thiazoles/chemical synthesis , Thiazoles/pharmacology , Biological Transport/drug effects , Biological Transport/genetics , Cell Line , Chemistry Techniques, Synthetic , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Humans , Ion Channel Gating/drug effects , Models, Molecular , Mutation , Protein Structure, Tertiary , Structure-Activity Relationship , Thiazoles/chemistry , Thiazoles/therapeutic use
11.
Cell Mol Life Sci ; 72(7): 1363-75, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25274064

ABSTRACT

The cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel is a membrane-integral protein that belongs to the ATP-binding cassette superfamily. Mutations in the CFTR gene cause cystic fibrosis in which salt, water, and protein transports are defective in various tissues. To investigate the conformation of the CFTR in the membrane, we applied the small-angle x-ray scattering (SAXS) technique on microsomal membranes extracted from NIH/3T3 cells permanentely transfected with wild-type (WT) CFTR and with CFTR carrying the ΔF508 mutation. The electronic density profile of the membranes was calculated from the SAXS data, assuming the lipid bilayer electronic density to be composed by a series of Gaussian shells. The data indicate that membranes in the microsome vesicles, that contain mostly endoplasmic reticulum membranes, are oriented in the outside-out conformation. Phosphorylation does not change significantly the electronic density profile, while dephosphorylation produces a significant modification in the inner side of the profile. Thus, we conclude that the CFTR and its associated protein complex in microsomes are mostly phosphorylated. The electronic density profile of the ΔF508-CFTR microsomes is completely different from WT, suggesting a different assemblage of the proteins in the membranes. Low-temperature treatment of cells rescues the ΔF508-CFTR protein, resulting in a conformation that resembles the WT. Differently, treatment with the corrector VX-809 modifies the electronic profile of ΔF508-CFTR membrane, but does not recover completely the WT conformation. To our knowledge, this is the first report of a direct physical measurement of the structure of membranes containing CFTR in its native environment and in different functional and pharmacological conditions.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Mutant Proteins/chemistry , Scattering, Small Angle , X-Ray Diffraction/methods , Algorithms , Aminopyridines/pharmacology , Animals , Benzodioxoles/pharmacology , Blotting, Western , Cell Membrane/chemistry , Cell Membrane/drug effects , Cell Membrane/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Humans , Intracellular Membranes/chemistry , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Mice , Microscopy, Electron , Microsomes/chemistry , Microsomes/metabolism , Microsomes/ultrastructure , Models, Molecular , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation , NIH 3T3 Cells , Phosphorylation , Protein Conformation/drug effects , Transfection
12.
Biochim Biophys Acta ; 1848(1 Pt A): 105-14, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25306966

ABSTRACT

Cl⁻ channels activated by acidic extracellular pH have been observed in various mammalian cells but their molecular identity and mechanisms of regulation are unknown. The aim of this study was to analyse the acid-activated Cl- current (ICl(H)) by elucidating its functional properties and mechanisms of regulation in three different cell types: primary human bronchial epithelial (HBE) cells, neuroblastoma SK-N-MC cells and HEK-293 cells. We found that outward rectification, sensitivity to acidic pH (50% activation at pH5.15), permeability sequence (SCN⁻>I⁻>Br⁻>Cl⁻>gluconate), voltage dependence and sensitivity to blockers of ICl(H) were identical in all cells. These findings suggest a common molecular basis for ICl(H). We analysed the possible relationship of ICl(H) with members of ClC and TMEM16 protein families. By gene silencing, validated using RT-PCR, we found that ICl(H) is unrelated to ClC-3, ClC-7, TMEM16A, TMEM16D, TMEM16F, TMEM16H and TMEM16K. Analysis of possible mechanisms of regulation indicate that Ca²âº, ATP and phosphorylation by PKA or PKC do not seem to be implicated in channel activation. Instead, the inhibition of ICl(H) by genistein and wortmannin suggest regulation by other kinases, possibly a tyrosine kinase and a phosphatidylinositol-3-kinase. Moreover, by using dynasore, the dynamin inhibitor, we found indications that exo/endocytosis is a mechanism responsible for ICl(H) regulation. Our results provide the first evidence about acid-activated Cl⁻ channel regulation and, thus, could open the way for a better understanding of the channel function and for the molecular identification of the underlying protein.


Subject(s)
Acids/metabolism , Chloride Channels/metabolism , Ion Channel Gating/physiology , Androstadienes/pharmacology , Animals , CHO Cells , Cell Line , Cell Line, Tumor , Cells, Cultured , Chloride Channels/genetics , Cricetinae , Cricetulus , Genistein/pharmacology , HEK293 Cells , Humans , Hydrazones/pharmacology , Hydrogen-Ion Concentration , Ion Channel Gating/genetics , Membrane Potentials/drug effects , Membrane Potentials/genetics , Membrane Potentials/physiology , Patch-Clamp Techniques , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , RNA Interference , Wortmannin
13.
Eur Biophys J ; 43(6-7): 341-6, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24771136

ABSTRACT

Cystic fibrosis transmembrane conductance regulator (CFTR) potentiators and correctors are new drugs that target the basic CFTR protein defect and are expected to benefit cystic fibrosis patients. To optimize the substances so far proposed for human use, and to minimise unwanted side effects, it is essential to investigate possible interactions between the drugs and cell components. We used small-angle X-ray scattering with synchrotron radiation to analyse the effects of two representative drugs, the potentiator VX-770 (Ivacaftor), approved for human use, and the corrector VX-809 (Lumacaftor), on a model phospholipid membrane. By reconstruction of the electron density profile of unilamellar vesicles treated with VX-770 or VX-809 we found that these drugs penetrate the phospholipid bilayer. VX-809 becomes homogeneously distributed throughout the bilayer whereas VX-770 accumulates predominantly in the internal leaflet, behaviour probably favoured by the asymmetry of the bilayer, because of vesicle curvature. Penetration of the bilayer by these drugs, probably as part of the mechanisms of permeation, causes destabilization of the membrane; this must be taken into account during future drug development.


Subject(s)
Aminophenols/pharmacology , Aminopyridines/pharmacology , Benzodioxoles/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Phospholipids/metabolism , Quinolones/pharmacology , Protein Binding , Temperature , Unilamellar Liposomes/chemistry , Unilamellar Liposomes/metabolism
14.
Am J Respir Cell Mol Biol ; 49(3): 445-52, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23600628

ABSTRACT

In the respiratory system, Na(+) absorption and Cl(-) secretion are balanced to maintain an appropriate airway surface fluid (ASF) volume and ensure efficient mucociliary clearance. In cystic fibrosis (CF), this equilibrium is disrupted by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene, resulting in the absence of functional CFTR-dependent Cl(-) secretion. The consequences of defective Cl(-) transport are worsened by the persistence of Na(+) absorption, which contributes to airway surface dehydration. We asked whether normal ASF can be restored to an equal extent by recovering Cl(-) secretion from mutated CFTR or by reducing Na(+) absorption. This is highly relevant in the selection of the best strategy for the treatment of patients with CF. We analyzed the ASF thickness of primary cultured bronchial CF and non-CF epithelia after silencing the epithelial Na(+) channel (ENaC) with specific short, interfering RNAs (siRNAs) and after the pharmacological stimulation of CFTR. Our results indicate that (1) single siRNAs complementary to ENaC subunits are sufficient to reduce ENaC transcripts, Na(+) channel activity, and fluid transport, but only silencing both the α and ß ENaC subunits at the same time leads to an increase of ASF (from nearly 7 µm to more than 9 µm); (2) the ASF thickness obtained in this way is about half that measured after maximal CFTR stimulation in non-CF epithelia (10-14 µm); and (3) the pharmacological rescue of mutant CFTR increases the ASF to the same extent as ENaC silencing. Our results indicate that CFTR rescue and ENaC silencing both produce a significant and long-lasting increase of airway hydration in vitro.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/metabolism , Epithelial Cells/metabolism , Epithelial Sodium Channel Blockers/metabolism , Epithelial Sodium Channels/genetics , RNA, Small Interfering/metabolism , Body Fluids , Bronchi/metabolism , Bronchi/pathology , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Cells/pathology , Epithelial Sodium Channels/metabolism , Gene Expression Regulation , Gene Silencing , Humans , Ion Transport , Mutation , Primary Cell Culture , RNA, Small Interfering/genetics , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology
15.
J Physiol ; 590(23): 6141-55, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22988141

ABSTRACT

The TMEM16A protein has a potential role as a Ca(2+)-activated Cl(-) channel (CaCC) in airway epithelia where it may be important in the homeostasis of the airway surface fluid. We investigated the function and expression of TMEM16A in primary human bronchial epithelial cells and in a bronchial cell line (CFBE41o-). Under resting conditions, TMEM16A protein expression was relatively low. However, TMEM16A silencing with short-interfering RNAs caused a marked inhibition of CaCC activity, thus demonstrating that a low TMEM16A expression is sufficient to support Ca(2+)-dependent Cl(-) transport. Following treatment for 24-72 h with interleukin-4 (IL-4), a cytokine that induces mucous cell metaplasia, TMEM16A protein expression was strongly increased in approximately 50% of primary bronchial epithelial cells, with a specific localization in the apical membrane. IL-4 treatment also increased the percentage of cells expressing MUC5AC, a marker of goblet cells. Interestingly, MUC5AC was detected specifically in cells expressing TMEM16A. In particular, MUC5AC was found in 15 and 60% of TMEM16A-positive cells when epithelia were treated with IL-4 for 24 or 72 h, respectively. In contrast, ciliated cells showed expression of the cystic fibrosis transmembrane conductance regulator Cl(-) channel but not of TMEM16A. Our results indicate that TMEM16A protein is responsible for CaCC activity in airway epithelial cells, particularly in cells treated with IL-4, and that TMEM16A upregulation by IL-4 appears as an early event of goblet cell differentiation. These findings suggest that TMEM16A expression is particularly required under conditions of mucus hypersecretion to ensure adequate secretion of electrolytes and water.


Subject(s)
Chloride Channels/physiology , Goblet Cells/physiology , Metaplasia/physiopathology , Neoplasm Proteins/physiology , Anoctamin-1 , Bronchi/cytology , Cell Line , Cells, Cultured , Epithelial Cells , HEK293 Cells , Humans , Interleukin-4/pharmacology , RNA, Small Interfering/administration & dosage
16.
Biochim Biophys Acta ; 1808(9): 2214-23, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21645494

ABSTRACT

TMEM16A protein, also known as anoctamin-1, has been recently identified as an essential component of Ca(2+)-activated Cl(-) channels. We previously reported the existence of different TMEM16A isoforms generated by alternative splicing. In the present study, we have determined the functional properties of a minimal TMEM16A protein. This isoform, called TMEM16A(0), has a significantly shortened amino-terminus and lacks three alternative segments localized in the intracellular regions of the protein (total length: 840 amino acids). TMEM16A(0) expression is associated with Ca(2+)-activated Cl(-) channel activity as measured by three different functional assays based on the halide-sensitive yellow fluorescent protein, short-circuit current recordings, and patch-clamp technique. However, compared to a longer isoform, TMEM16(abc) (total length: 982 amino acids), TMEM16A(0) completely lacks voltage-dependent activation. Furthermore, TMEM16A(0) and TMEM16A(abc) have similar but not identical responses to extracellular anion replacement, thus suggesting a difference in ion selectivity and conductance. Our results indicate that TMEM16A(0) has the basic domains required for anion transport and Ca(2+)-sensitivity. However, the absence of alternative segments, which are present in more complex isoforms of TMEM16A, modifies the channel gating and ion transport ability.


Subject(s)
Chloride Channels/chemistry , Membrane Proteins/chemistry , Neoplasm Proteins/chemistry , Alternative Splicing , Anoctamin-1 , Bacterial Proteins/metabolism , Calcium/chemistry , Chlorides/pharmacology , HEK293 Cells , Humans , Ions , Luminescent Proteins/metabolism , Membrane Potentials , Microscopy, Fluorescence/methods , Patch-Clamp Techniques , Protein Conformation , Protein Isoforms , Transfection
17.
J Cyst Fibros ; 10 Suppl 2: S129-45, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21658632

ABSTRACT

With knowledge of the molecular behaviour of the cystic fibrosis transmembrane conductance regulator (CFTR), its physiological role and dysfunction in cystic fibrosis (CF), therapeutic strategies are now being developed that target the root cause of CF rather than disease symptoms. Here, we review progress towards the development of rational new therapies for CF. We highlight the discovery of small molecules that rescue the cell surface expression and defective channel gating of CF mutants, termed CFTR correctors and CFTR potentiators, respectively. We draw attention to alternative approaches to restore epithelial ion transport to CF epithelia, including inhibitors of the epithelial Na(+) channel (ENaC) and activators of the Ca(2+)-activated Cl(-) channel TMEM16A. The expertise required to translate small molecules identified in the laboratory to drugs for CF patients depends on our ability to coordinate drug development at an international level and our ability to provide pertinent biological information using suitable disease models.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/agonists , Cystic Fibrosis/drug therapy , Drug Design , Epithelial Sodium Channel Blockers , Membrane Proteins/agonists , Neoplasm Proteins/agonists , Animals , Anoctamin-1 , Chloride Channels , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Drug Industry/organization & administration , Drug Industry/trends , Humans
18.
Methods Mol Biol ; 741: 13-21, 2011.
Article in English | MEDLINE | ID: mdl-21594775

ABSTRACT

Small molecules acting as selective activators (potentiators), inhibitors, or "correctors" of the CFTR chloride channel represent candidate drugs for various pathological conditions including cystic fibrosis and secretory diarrhea. The identification of CFTR pharmacological modulators may be achieved by screening highly diverse synthetic or natural compound libraries using high-throughput methods. A convenient assay for CFTR function is based on the halide sensitivity of the yellow fluorescent protein (YFP). CFTR activity can be simply assessed by measuring the rate of YFP signal decrease caused by iodide influx. This assay can be automated to test thousands of compounds per day.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/methods , Small Molecule Libraries/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/agonists , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , HEK293 Cells , Humans , Luminescent Proteins/genetics , Microscopy, Fluorescence , Mutation , Transfection
19.
J Biol Chem ; 286(17): 15215-26, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21383017

ABSTRACT

A large fraction of mutations causing cystic fibrosis impair the function of the cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel by causing reduced channel activity (gating defect) and/or impaired exit from the endoplasmic reticulum (trafficking defect). Such defects need to be treated with separate pharmacological compounds termed potentiators and correctors, respectively. Here, we report the characterization of aminoarylthiazoles (AATs) as compounds having dual activity. Cells expressing mutant CFTR were studied with functional assays (fluorescence-based halide transport and short circuit current measurements) to assess the effect of acute and chronic treatment with compounds. We found that AATs are effective on F508del, the most frequent cystic fibrosis mutation, which is associated with both a gating and a trafficking defect. AATs are also effective on mutations like G1349D and G551D, which cause only a gating defect. Evaluation of a panel of AAT analogs identified EN277I as the most effective compound. Incubation of cells expressing mutant CFTR with EN277I caused a strong stimulation of channel activity as demonstrated by single channel recordings. Compounds with dual activity such as AATs may be useful for the development of effective drugs for the treatment of cystic fibrosis.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Cystic Fibrosis/genetics , Ion Channel Gating , Mutation , Thiazoles/pharmacology , Biological Transport/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Humans , Ion Channel Gating/genetics , Patch-Clamp Techniques , Structure-Activity Relationship , Thiazoles/therapeutic use
20.
Compr Physiol ; 1(4): 2155-74, 2011 Oct.
Article in English | MEDLINE | ID: mdl-23733701

ABSTRACT

Ca(2+)-activated Cl(-) channels (CaCCs) are plasma membrane proteins involved in various important physiological processes. In epithelial cells, CaCC activity mediates the secretion of Cl(-) and of other anions, such as bicarbonate and thiocyanate. In smooth muscle and excitable cells of the nervous system, CaCCs have an excitatory role coupling intracellular Ca(2+) elevation to membrane depolarization. Recent studies indicate that TMEM16A (transmembrane protein 16 A or anoctamin 1) and TMEM16B (transmembrane protein 16 B or anoctamin 2) are CaCC-forming proteins. Induced expression of TMEM16A and B in null cells by transfection causes the appearance of Ca(2+)-activated Cl(-) currents similar to those described in native tissues. Furthermore, silencing of TMEM16A by RNAi causes disappearance of CaCC activity in cells from airway epithelium, biliary ducts, salivary glands, and blood vessel smooth muscle. Mice devoid of TMEM16A expression have impaired Ca(2+)-dependent Cl(-) secretion in the epithelial cells of the airways, intestine, and salivary glands. These animals also show a loss of gastrointestinal motility, a finding consistent with an important function of TMEM16A in the electrical activity of gut pacemaker cells, that is, the interstitial cells of Cajal. Identification of TMEM16 proteins will help to elucidate the molecular basis of Cl(-) transport.


Subject(s)
Chloride Channels/metabolism , Chlorides/metabolism , Animals , Chloride Channels/classification , Chloride Channels/genetics , Endothelial Cells/metabolism , Epithelial Cells/metabolism , Gene Expression , Humans , Ion Transport , Muscle Cells/metabolism , Neurons/metabolism , Organ Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...