Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Am J Physiol Cell Physiol ; 307(12): C1102-12, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25298423

ABSTRACT

Iodide is captured by thyrocytes through the Na(+)/I(-) symporter (NIS) before being released into the follicular lumen, where it is oxidized and incorporated into thyroglobulin for the production of thyroid hormones. Several reports point to pendrin as a candidate protein for iodide export from thyroid cells into the follicular lumen. Here, we show that a recently discovered Ca(2+)-activated anion channel, TMEM16A or anoctamin-1 (ANO1), also exports iodide from rat thyroid cell lines and from HEK 293T cells expressing human NIS and ANO1. The Ano1 mRNA is expressed in PCCl3 and FRTL-5 rat thyroid cell lines, and this expression is stimulated by thyrotropin (TSH) in rat in vivo, leading to the accumulation of the ANO1 protein at the apical membrane of thyroid follicles. Moreover, ANO1 properties, i.e., activation by intracellular calcium (i.e., by ionomycin or by ATP), low but positive affinity for pertechnetate, and nonrequirement for chloride, better fit with the iodide release characteristics of PCCl3 and FRTL-5 rat thyroid cell lines than the dissimilar properties of pendrin. Most importantly, iodide release by PCCl3 and FRTL-5 cells is efficiently blocked by T16Ainh-A01, an ANO1-specific inhibitor, and upon ANO1 knockdown by RNA interference. Finally, we show that the T16Ainh-A01 inhibitor efficiently blocks ATP-induced iodide efflux from in vitro-cultured human thyrocytes. In conclusion, our data strongly suggest that ANO1 is responsible for most of the iodide efflux across the apical membrane of thyroid cells.


Subject(s)
Cell Polarity , Chloride Channels/metabolism , Iodides/metabolism , Neoplasm Proteins/metabolism , Thyroid Gland/metabolism , Adenosine Triphosphate/metabolism , Animals , Anoctamin-1 , Biological Transport , Calcium/metabolism , Chloride Channels/antagonists & inhibitors , Chloride Channels/genetics , HEK293 Cells , Humans , Membrane Transport Modulators/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , RNA Interference , Rats , Thyroid Gland/cytology , Thyroid Gland/drug effects , Thyrotropin/metabolism , Time Factors , Transfection
2.
Cell Mol Life Sci ; 62(4): 446-60, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15719171

ABSTRACT

The use of substances that could activate the defective chloride channels of the mutant cystic fibrosis transmembrane conductance regulator (CFTR) has been suggested as possible therapy for cystic fibrosis. Using epithelia formed by cells stably transfected with wildtype or mutant (G551D, G1349D) CFTR, we estimated the apparent dissociation constant, K(D), of a series of CFTR activators by measuring the increase in the apical membrane current. Modification of apparent K(D) of CFTR activators by mutations of the nucleotide-binding domains (NBDs) suggests that the binding site might be in these regions. The human NBD structure was predicted by homology with murine NBD1. An NBD1-NBD2 complex was constructed by overlying monomers to a bacterial ABC transporter NBD dimer in the "head-to-tail" conformation. Binding sites for CFTR activators were predicted by molecular docking. Comparison of theoretical binding free energy estimated in the model to free energy estimated from the apparent dissociation constants, K(D), resulted in a remarkably good correlation coefficient for one of the putative binding sites, located in the interface between NBD1 and NBD2.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/agonists , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , ATP-Binding Cassette Transporters/agonists , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/physiology , Adenine Nucleotides/metabolism , Amino Acid Sequence , Animals , Binding Sites/drug effects , Binding Sites/genetics , Cell Line , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/genetics , Cell Membrane Permeability/physiology , Cystic Fibrosis/drug therapy , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Dimerization , Electric Conductivity , Electrophysiology , Genistein/chemistry , Genistein/pharmacology , Humans , Mice , Molecular Sequence Data , Mutation/genetics , Protein Structure, Tertiary , Rats , Sequence Alignment , Thermodynamics
3.
J Cell Sci ; 114(Pt 22): 4073-81, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11739639

ABSTRACT

A number of genetic diseases, including cystic fibrosis, have been identified as disorders of protein trafficking associated with retention of mutant protein within the endoplasmic reticulum. In the presence of the benzo(c)quinolizinium drugs, MPB-07 and its congener MPB-91, we show the activation of cystic fibrosis transmembrane conductance regulator (CFTR) delF508 channels in IB3-1 human cells, which express endogenous levels of delF508-CFTR. These drugs were without effect on the Ca(2+)-activated Cl- transport, whereas the swelling-activated Cl- transport was found altered in MPB-treated cells. Immunoprecipitation and in vitro phosphorylation shows a 20% increase of the band C form of delF508 after MPB treatment. We then investigated the effect of these drugs on the extent of mislocalisation of delF508-CFTR in native airway cells from cystic fibrosis patients. We first showed that delF508 CFTR was characteristically restricted to an endoplasmic reticulum location in approximately 80% of untreated cells from CF patients homozygous for the delF508-CFTR mutation. By contrast, 60-70% of cells from non-CF patients showed wild-type CFTR in an apical location. MPB-07 treatment caused dramatic relocation of delF508-CFTR to the apical region such that the majority of delF508/delF508 CF cells showed a similar CFTR location to that of wild-type. MPB-07 had no apparent effect on the distribution of wild-type CFTR, the apical membrane protein CD59 or the ER membrane Ca(2+),Mg-ATPase. We also showed a similar pharmacological effect in nasal cells freshly isolated from a delF508/G551D CF patient. The results demonstrate selective redirection of a mutant membrane protein using cell-permeant small molecules of the benzo(c)quinolizinium family and provide a major advance towards development of a targetted drug treatment for cystic fibrosis and other disorders of protein trafficking.


Subject(s)
Chlorides/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/metabolism , Quinolizines/pharmacology , Respiratory Mucosa/drug effects , Calcium/metabolism , Cell Polarity , Cells, Cultured , Cyclic AMP/agonists , Cyclic AMP/metabolism , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Enzyme Inhibitors/pharmacology , Humans , Immunohistochemistry , Iodides/metabolism , Quinolizines/chemistry , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism
4.
Am J Physiol Cell Physiol ; 281(5): C1657-66, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11600430

ABSTRACT

We have designed and synthesized benzo[c]quinolizinium derivatives and evaluated their effects on the activity of G551D cystic fibrosis transmembrane conductance regulator (CFTR) expressed in Chinese hamster ovary and Fisher rat thyroid cells. We demonstrated, using iodide efflux, whole cell patch clamp, and short-circuit recordings, that 5-butyl-6-hydroxy-10-chlorobenzo[c]quinolizinium chloride (MPB-91) restored the activity of G551D CFTR (EC(50) = 85 microM) and activated CFTR in Calu-3 cells (EC(50) = 47 microM). MPB-91 has no effect on the ATPase activity of wild-type and G551D NBD1/R/GST fusion proteins or on the ATPase, GTPase, and adenylate kinase activities of purified NBD2. The activation of CFTR by MPB-91 is independent of phosphorylation because 1) kinase inhibitors have no effect and 2) the compound still activated CFTR having 10 mutated protein kinase A sites (10SA-CFTR). The new pharmacological agent MPB-91 may be an important candidate drug to ameliorate the ion transport defect associated with CF and to point out a new pathway to modulate CFTR activity.


Subject(s)
Adenosine Triphosphatases/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Enzyme Activators/pharmacology , Quinolizines/pharmacology , Adenosine Triphosphatases/metabolism , Animals , CHO Cells , Chloride Channels/drug effects , Chloride Channels/metabolism , Cricetinae , Electrophysiology , Iodides/metabolism , Patch-Clamp Techniques , Phosphorylation , Rats , Rats, Inbred F344 , Thyroid Gland/cytology , Thyroid Gland/drug effects , Thyroid Gland/metabolism
5.
FEBS Lett ; 499(3): 220-4, 2001 Jun 22.
Article in English | MEDLINE | ID: mdl-11423120

ABSTRACT

The green fluorescent protein YFP-H148Q is sensitive to halides by a mechanism involving halide binding and a shift in pK(a). However, a limitation of YFP-H148Q is its low halide sensitivity, with K(d)>100 mM for Cl(-). Indicators with improved sensitivities are needed for cell transport studies, particularly in drug discovery by high-throughput screening, and for measurement of Cl(-) concentration in subcellular organelles. YFP-H148Q libraries were generated in which pairs of residues in the vicinity of the halide binding site were randomly mutated. An automated procedure was developed to screen bacterial colonies for improved halide sensitivity. Analysis of 1536 clones revealed improved anion sensitivities with K(d) down to 2 mM for I(-) (I152L), 40 mM for Cl(-) (V163S), and 10 mM for NO(3)(-) (I152L). The anion-sensitive mechanism of these indicators was established and their utility in cells was demonstrated using transfected cells expressing the cystic fibrosis transmembrane conductance regulator chloride channel.


Subject(s)
Chlorides/metabolism , Iodides/metabolism , Luminescent Proteins/metabolism , 3T3 Cells , Animals , Escherichia coli/genetics , Green Fluorescent Proteins , Kinetics , Luminescent Proteins/genetics , Mice , Mutation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transformation, Bacterial
6.
J Biol Chem ; 276(23): 19723-8, 2001 Jun 08.
Article in English | MEDLINE | ID: mdl-11262417

ABSTRACT

The flavonoid genistein and the benzo[c]quinolizinium MPB-07 have been shown to activate the cystic fibrosis transmembrane conductance regulator (CFTR), the protein that is defective in cystic fibrosis. Lead-based combinatorial and parallel synthesis yielded 223 flavonoid, quinolizinium, and related heterocyclic compounds. The compounds were screened for their ability to activate CFTR at 50 microm concentration by measurement of the kinetics of iodide influx in Fisher rat thyroid cells expressing wild-type or G551D CFTR together with the green fluorescent protein-based halide indicator YFP-H148Q. Duplicate screenings revealed that 204 compounds did not significantly affect CFTR function. Compounds of the 7,8-benzoflavone class, which are structurally intermediate between flavones and benzo[c]quinoliziniums, were effective CFTR activators with the most potent being 2-(4-pyridinium)benzo[h]4H-chromen-4-one bisulfate (UCcf-029). Compounds of the novel structural class of fused pyrazolo heterocycles were also strong CFTR activators with the most potent being 3-(3-butynyl)-5-methoxy-1-phenylpyrazole-4-carbaldehyde (UCcf-180). A CFTR inhibitor was also identified. The active compounds did not induce iodide influx in null cells deficient in CFTR. Short-circuit current measurements showed that the CFTR activators identified by screening induced strong anion currents in the transfected cell monolayers grown on porous supports. Compared with genistein, the most active compounds had up to 10 times greater potency in activating wild-type and/or G551D-CFTR. The activators had low cellular toxicity and did not elevate cellular cAMP concentration or inhibit phosphatase activity, suggesting that CFTR activation may involve a direct interaction. These results establish an efficient screening procedure to identify CFTR activators and inhibitors and have identified 7,8-benzoflavones and pyrazolo derivatives as novel classes of CFTR activators.


Subject(s)
Combinatorial Chemistry Techniques , Cystic Fibrosis Transmembrane Conductance Regulator/agonists , Flavonoids/chemistry , Quinazolines/chemistry , Animals , Cells, Cultured , Rats , Rats, Inbred F344
7.
Am J Physiol Cell Physiol ; 279(6): C1925-37, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11078708

ABSTRACT

The pharmacological activation of the cystic fibrosis gene protein cystic fibrosis transmembrane conductance regulator (CFTR) was studied in human airway epithelial Calu-3 cells, which express a high level of CFTR protein as assessed by Western blot and in vitro phosphorylation. Immunolocalization shows that CFTR is located in the apical membrane. We performed iodide efflux, whole cell patch-clamp, and short-circuit recordings to demonstrate that the novel synthesized xanthine derivative 3, 7-dimethyl-1-isobutylxanthine (X-33) is an activator of the CFTR channel in Calu-3 cells. Whole cell current activated by X-33 or IBMX is linear, inhibited by glibenclamide and diphenylamine-2-carboxylate but not by DIDS or TS-TM calix[4]arene. Intracellular cAMP was not affected by X-33. An outwardly rectifying Cl(-) current was recorded in the absence of cAMP and X-33 stimulation, inhibited by DIDS and TS-TM calix[4]arene. With the use of short-circuit recordings, X-33 and IBMX were able to stimulate a large concentration-dependent CFTR transport that was blocked by glibenclamide but not by DIDS. Our results show that manipulating the chemical structure of xanthine derivatives offers an opportunity to identify further specific activators of CFTR in airway cells.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/drug effects , Theophylline/analogs & derivatives , Xanthines/pharmacology , 1-Methyl-3-isobutylxanthine/pharmacology , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Animals , Biological Transport/drug effects , Biological Transport/physiology , CHO Cells , Caffeine/pharmacology , Calcium Channel Blockers/pharmacology , Chlorides/metabolism , Colforsin/pharmacology , Cricetinae , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Glyburide/pharmacology , Hypoglycemic Agents/pharmacology , In Vitro Techniques , Iodides/pharmacokinetics , Iodine Radioisotopes , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Proteins/metabolism , Patch-Clamp Techniques , Phosphodiesterase Inhibitors/pharmacology , Phosphorylation , Respiratory Mucosa/physiology , Theophylline/pharmacology , Xanthines/chemical synthesis , ortho-Aminobenzoates/pharmacology
8.
Am J Physiol Lung Cell Mol Physiol ; 278(6): L1186-94, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10835324

ABSTRACT

Human bronchial epithelial cells were treated in vitro with interferon-gamma or tumor necrosis factor-alpha to assess their effect on transepithelial ion transport. Short-circuit current measurements revealed that Na(+) absorption was markedly inhibited by interferon-gamma (10-1,000 U/ml). The cystic fibrosis transmembrane conductance regulator was also downregulated by interferon-gamma as evident at the protein level and by the decrease in the cAMP-dependent current. On the other hand, interferon-gamma caused an increase of the current elicited by apical UTP application, which is due to the activity of Ca(2+)-dependent Cl(-) channels. Tumor necrosis factor-alpha caused few changes in ion transport. Transepithelial fluid transport was measured in normal and cystic fibrosis cells. At rest, both types of cells showed an amiloride-sensitive fluid absorption that was inhibited by interferon-gamma but not by tumor necrosis factor-alpha. Our results show that interferon-gamma alters the transepithelial ion transport of cultured bronchial cells. This effect may change the ion composition and/or volume of periciliary fluid.


Subject(s)
Bronchi/metabolism , Interferon-gamma/pharmacology , Biological Transport/drug effects , Body Fluids/metabolism , Bronchi/cytology , Bronchi/drug effects , Calcium/metabolism , Cell Membrane/drug effects , Cells, Cultured , Chloride Channels/drug effects , Chloride Channels/physiology , Cyclic AMP/physiology , Cystic Fibrosis/metabolism , Cystic Fibrosis/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Down-Regulation , Electric Conductivity , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Ions , Reference Values , Sodium Channels/drug effects , Sodium Channels/physiology , Tumor Necrosis Factor-alpha/pharmacology , Uridine Triphosphate/pharmacology
9.
Genomics ; 60(3): 251-7, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10493825

ABSTRACT

We describe the identification and characterization of a new gene deleted in the AMME contiguous gene syndrome. This gene is predominantly expressed in heart, skeletal muscle, spinal cord, and brain. Screening of placenta and NT2 cDNA libraries enabled us to obtain the 1.5-kb full-length transcript, which shows a 426-bp open reading frame. Since the resulting 142-amino-acid peptide has a single putative transmembrane domain and a weak but suggestive homology with KCNE1 (minK), a protein associated with the KCNQ1 potassium channel (KVLQT1), we named this new gene KCNE1-like (KCNE1L). To obtain greater insight into this new member of an apparently distinct protein family, we have identified and characterized the homologous mouse gene (Kcne1l), which encodes a peptide of 143 amino acids with 91% homology and 80% identity. The expression pattern of mouse Kcne1l in the developing embryo revealed strong signal in ganglia, in the migrating neural crest cells of cranial nerves, in the somites, and in the myoepicardial layer of the heart. The specific distribution in adult tissues, the putative channel function, and the expression pp6tern in the developing mouse embryo suggest that KCNE1L could be involved in the development of the cardiac abnormalities as well as of some neurological signs observed in patients with AMME contiguous gene syndrome.


Subject(s)
Gene Deletion , Intellectual Disability/genetics , Nephritis, Hereditary/genetics , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , X Chromosome/genetics , Animals , Chromosome Mapping , Clone Cells , Databases, Factual , Electric Conductivity , Electrocardiography , Gene Expression , Heart Defects, Congenital/genetics , Humans , In Situ Hybridization , Male , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Potassium Channels/chemistry , Sequence Homology, Nucleic Acid , Syndrome
10.
J Biol Chem ; 274(39): 27415-25, 1999 Sep 24.
Article in English | MEDLINE | ID: mdl-10488073

ABSTRACT

Chloride channels play an important role in the physiology and pathophysiology of epithelia, but their pharmacology is still poorly developed. We have chemically synthesized a series of substituted benzo[c]quinolizinium (MPB) compounds. Among them, 6-hydroxy-7-chlorobenzo[c]quinolizinium (MPB-27) and 6-hydroxy-10-chlorobenzo[c]quinolizinium (MPB-07), which we show to be potent and selective activators of the cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel. We examined the effect of MPB compounds on the activity of CFTR channels in a variety of established epithelial and nonepithelial cell systems. Using the iodide efflux technique, we show that MPB compounds activate CFTR chloride channels in Chinese hamster ovary (CHO) cells stably expressing CFTR but not in CHO cells lacking CFTR. Single and whole cell patch clamp recordings from CHO cells confirm that CFTR is the only channel activated by the drugs. Ussing chamber experiments reveal that the apical addition of MPB to human nasal epithelial cells produces a large increase of the short circuit current. This current can be totally inhibited by glibenclamide. Whole cell experiments performed on native respiratory cells isolated from wild type and CF null mice also show that MPB compounds specifically activate CFTR channels. The activation of CFTR by MPB compounds was glibenclamide-sensitive and 4, 4'-diisothiocyanostilbene-2,2'-disulfonic acid-insensitive. In the human tracheal gland cell line MM39, MPB drugs activate CFTR channels and stimulate the secretion of the antibacterial secretory leukoproteinase inhibitor. In submandibular acinar cells, MPB compounds slightly stimulate CFTR-mediated submandibular mucin secretion without changing intracellular cAMP and ATP levels. Similarly, in CHO cells MPB compounds have no effect on the intracellular levels of cAMP and ATP or on the activity of various protein phosphatases (PP1, PP2A, PP2C, or alkaline phosphatase). Our results provide evidence that substituted benzo[c]quinolizinium compounds are a novel family of activators of CFTR and of CFTR-mediated protein secretion and therefore represent a new tool to study CFTR-mediated chloride and secretory functions in epithelial tissues.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Quinolizines/pharmacology , Animals , CHO Cells , Cilia/drug effects , Cilia/physiology , Colforsin/pharmacology , Cricetinae , Cystic Fibrosis Transmembrane Conductance Regulator/deficiency , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Drug Design , Female , Glyburide/pharmacology , Humans , Male , Membrane Potentials/drug effects , Mice , Mice, Inbred BALB C , Mice, Knockout , Molecular Structure , Nasal Mucosa/drug effects , Nasal Mucosa/physiology , Patch-Clamp Techniques , Quinolines/chemical synthesis , Quinolines/chemistry , Quinolines/pharmacology , Quinolizines/chemical synthesis , Quinolizines/chemistry , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Structure-Activity Relationship , Transfection
11.
Ann Allergy Asthma Immunol ; 83(1): 49-54, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10437816

ABSTRACT

OBJECTIVE: To test in vitro and in vivo the hypothesis that sodium nedocromil could modulate the expression of surface molecules on airway epithelial cells. METHODS: Human bronchial epithelial cells, obtained from surgically resected bronchi, were cultured and stimulated with recombinant IFN-gamma in the presence of sodium nedocromil. The intensity of the expression of surface molecules HLA-DR and ICAM-1 molecules on bronchial epithelial cells in vitro, was quantified by specific antibody staining and flow-cytometry analysis. Furthermore, we studied the effect of the drug on airway inflammation in vivo and on allergic rhinitis patients sensitized to house dust mites. Nasal epithelial cells were collected by brushing, at baseline and 2 to 3 weeks after treatment with sodium nedocromil. The expression of HLA-DR and ICAM-1 molecules was measured by flow-cytometry, and the proportions of neutrophils and eosinophils "contaminating" the epithelial cells evaluated by light microscopy examination of nasal brushings. RESULTS: The enhanced HLA-DR and ICAM-1 expression, induced by IFN-gamma, was effectively downregulated, in a dose-dependent manner, by sodium nedocromil. At all the concentrations tested (10(-9) to 10(-4) M), the inhibitory activity of the drug was stronger on HLA-DR than on ICAM-1 expression (P<.05, all comparisons). As compared with healthy subjects, patients with allergic rhinitis had a higher expression of HLA-DR (P<.05) but not of ICAM-1 molecules (P>.05) on nasal epithelial cells, and higher proportions of nasal eosinophils (P<.05). Treatment with sodium nedocromil downregulated the expression of HLA-DR (P<.05), but not of ICAM-1 (P>.05), and induced a mild, but not statistically significant, decrease of nasal eosinophilia (P>.05). CONCLUSION: These data demonstrate that the antiinflammatory activity of sodium nedocromil may include modulation of surface molecule expression on airway epithelial cells.


Subject(s)
Bronchi/cytology , Epithelial Cells/metabolism , HLA-DR Antigens/biosynthesis , Intercellular Adhesion Molecule-1/biosynthesis , Nedocromil/pharmacology , Adolescent , Anti-Allergic Agents/therapeutic use , Cell Communication/drug effects , Cells, Cultured , Child , Down-Regulation/drug effects , Female , Humans , Hypersensitivity, Immediate/drug therapy , Hypersensitivity, Immediate/pathology , Interferon-gamma/pharmacology , Male , Middle Aged , Nasal Mucosa/immunology , Nedocromil/therapeutic use
12.
J Immunol ; 162(9): 5359-66, 1999 May 01.
Article in English | MEDLINE | ID: mdl-10228012

ABSTRACT

The HIV-1 Nef protein plays an important role in the development of the pathology associated with AIDS. Despite various studies that have dealt with different aspects of Nef function, the complete mechanism by which it alters the physiology of infected cells remains to be established. Nef can associate with cell membranes, therefore supporting the hypothesis that it might interact with membrane proteins as ionic channels and modify their electrical properties. By using the patch-clamp technique, we found that Nef expression determines a 25-mV depolarization of lymphoblastoid CEM cells. Both charybdotoxin (CTX) and the membrane-permeable Ca2+ chelator BAPTA/AM depolarized the membrane of native cells without modifying that of Nef-transfected cells. These data suggested that the resting potential in native CEM cells is settled by a CTX- and Ca2+-sensitive K+ channel (KCa,CTX), whose activity is absent in Nef-expressing cells. This was confirmed by direct measurements of whole-cell KCa,CTX currents. Single-channel recordings on excised patches showed that a KCa,CTX channel of 35 pS with a half-activation near 400 nM Ca2+ was present in both native and Nef-transfected cells. The measurements of free intracellular Ca2+ were not different in the two cell lines, but Nef-transfected cells displayed an increased Ca2+ content in ionomycin-sensitive stores. Taken together, these results indicate that Nef expression alters the resting membrane potential of the T lymphocyte cell line by inhibiting a KCa,CTX channel, possibly by intervening in the regulation of intracellular Ca2+ homeostasis.


Subject(s)
Calcium/physiology , Gene Products, nef/biosynthesis , HIV-1/physiology , Potassium Channel Blockers , T-Lymphocytes/physiology , T-Lymphocytes/virology , Alkaline Phosphatase/antagonists & inhibitors , Calcium/metabolism , Cell Line, Transformed , Charybdotoxin/pharmacology , Enzyme Inhibitors/pharmacology , Gene Products, nef/genetics , Genistein/pharmacology , HIV-1/genetics , Humans , Hydrogen-Ion Concentration , Intracellular Fluid/physiology , Ion Channel Gating/drug effects , Ion Channel Gating/genetics , Membrane Potentials/drug effects , Membrane Potentials/genetics , Patch-Clamp Techniques , Potassium Channels/genetics , Potassium Channels/physiology , Protein-Tyrosine Kinases/antagonists & inhibitors , T-Lymphocytes/enzymology , Transfection , Tumor Cells, Cultured , Vanadates/pharmacology , nef Gene Products, Human Immunodeficiency Virus
13.
J Biol Chem ; 274(17): 11701-7, 1999 Apr 23.
Article in English | MEDLINE | ID: mdl-10206984

ABSTRACT

The activity of volume-sensitive Cl- channels was studied in human tracheal epithelial cells (9HTEo-) by taurine efflux experiments. The efflux elicited by a hypotonic shock was partially inhibited by adenosine receptor antagonists, by alpha,beta-methyleneadenosine 5'-diphosphate (alphabetaMeADP), an inhibitor of the 5'-ectonucleotidase, and by adenosine deaminase. On the other hand, dipyridamole, a nucleoside transporter inhibitor, increased the swelling-induced taurine efflux. Extracellular ATP and adenosine increased taurine efflux by potentiating the effect of hypotonic shock. alphabetaMeADP strongly inhibited the effect of extracellular ATP but not that of adenosine. These results suggest that anion channel activation involves the release of intracellular ATP, which is then degraded to adenosine by specific ectoenzymes. Adenosine then binds to purinergic receptors, causing the activation of the channels. To directly demonstrate ATP efflux, cells were loaded with [3H]AMP, and the release of radiolabeled molecules was analyzed by high performance liquid chromatography. During hypotonic shock, cell supernatants showed the presence of ATP, ADP, and adenosine. alphabetaMeADP inhibited adenosine formation and caused the appearance of AMP. Under hypotonic conditions, elevation of intracellular Ca2+ by ionomycin caused an increase of ATP and adenosine in the extracellular solution. Our results demonstrate that volume-sensitive anion channels are regulated with an autocrine mechanism involving swelling-induced ATP release and then hydrolysis to adenosine.


Subject(s)
Adenosine/pharmacology , Chloride Channels/drug effects , Trachea/drug effects , Adenine Nucleotides/metabolism , Cell Line, Transformed , Chloride Channels/metabolism , Chromatography, High Pressure Liquid , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Osmotic Pressure , Trachea/cytology , Trachea/metabolism , Type C Phospholipases/metabolism
14.
Gene ; 228(1-2): 181-8, 1999 Mar 04.
Article in English | MEDLINE | ID: mdl-10072771

ABSTRACT

The bovine CaCC protein is a putative Ca2+-dependent Cl- channel of airway epithelial cells. Therefore, CaCC proteins could contribute to transepithelial Cl- transport and accordingly modify the phenotype of cystic fibrosis (CF) patients. We have identified a murine EST containing a full-length cDNA coding for a 902-amino-acid protein highly homologous to bovine CaCC. The murine gene (mCaCC) maps to chromosome 3 at the H2-H3 band and is expressed, as indicated by Northern blot analysis, in mouse skin and kidney but not in brain, heart, lung or testis. RT-PCR indicates a low expression in tracheal epithelial cells. Heterologous expression of mCaCC in Xenopus oocytes elicits membrane currents that are anion-selective and inhibited by DIDS and by niflumic acid, a blocker of the endogenous chloride current in oocytes. The identification of genes belonging to the CaCC family will help to evaluate their role as ion channels or channel regulators and their actual contribution to epithelial chloride transport.


Subject(s)
Calcium/physiology , Chloride Channels/genetics , Genes/genetics , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Amino Acid Sequence , Animals , Blotting, Northern , Cattle , Chloride Channels/administration & dosage , Chromosome Mapping , Expressed Sequence Tags , Female , Gene Expression , In Situ Hybridization, Fluorescence , Ionomycin/pharmacology , Ionophores/pharmacology , Male , Membrane Potentials/drug effects , Mice , Microinjections , Molecular Sequence Data , Niflumic Acid/pharmacology , Oocytes/cytology , Oocytes/drug effects , Oocytes/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Sodium Chloride/pharmacology , Tissue Distribution , Xenopus
15.
Br J Pharmacol ; 125(6): 1382-6, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9863671

ABSTRACT

Chromones (sodium cromoglycate and sodium nedocromil) block cell swelling-activated Cl- channels in NIH-3T3 fibroblasts and endothelial cells. This has led to hypothesize that cell volume regulation might be involved in asthma pathogenesis. Using whole-cell patch-clamp experiments, we studied the effect of chromones on volume-sensitive Cl- currents in transformed human tracheal epithelial cells (9HTEo-) and in primary cultures of human bronchial epithelial cells (BE). Cl- currents activated by hypotonic shock were poorly blocked by extracellular nedocromil or cromoglycate. The block was voltage-dependent since it was observed only at positive membrane potentials. At the concentration of 5 mM, the current inhibition by both chromones at +80 mV was about 40% for 9HTEo- and only 20% for BE. Intracellular application of chromones elicited a voltage-independent inhibition in 9HTEo- cells. Under this condition, volume-sensitive Cl- currents were reduced at all membrane potentials (60 and 45% inhibition by 2 mM nedocromil and cromoglycate respectively). In contrast intracellular chromones were ineffective in BE cells. The relative refractoriness to chromones, in contrast with the high sensitivity shown by other Cl- channels, suggests that the epithelial volume-sensitive Cl- channel is not involved in asthma.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Chloride Channels/antagonists & inhibitors , Chlorides/physiology , Cromolyn Sodium/pharmacology , Nedocromil/pharmacology , Trachea/drug effects , 3T3 Cells/drug effects , 3T3 Cells/physiology , Animals , Cells, Cultured , Chloride Channels/physiology , Epithelial Cells/drug effects , Epithelial Cells/physiology , Humans , Kinetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Sensitivity and Specificity , Trachea/physiology
16.
Am J Physiol ; 275(5): L917-23, 1998 11.
Article in English | MEDLINE | ID: mdl-9815109

ABSTRACT

We performed Ussing chamber experiments on cultured human bronchial epithelial cells to look for the presence of electrogenic dibasic amino acid transport. Apical but not basolateral L-arginine (10-1, 000 microM) increased the short-circuit current. Maximal effect and EC50 were approximately 3.5 microA/cm2 and 80 microM, respectively, in cells from normal subjects and cystic fibrosis patients. The involvement of nitric oxide was ruled out because a nitric oxide synthase inhibitor (NG-nitro-L-arginine methyl ester) did not decrease the arginine-dependent current. Apical L-lysine, L-alanine, and L-proline, but not aspartic acid, were also effective in increasing the short-circuit current, with EC50 values ranging from 26 to 971 microM. Experiments performed with radiolabeled arginine demonstrated the presence of an Na+-dependent concentrative transporter on the apical membrane of bronchial cells. This transporter could be important in vivo to maintain a low amino acid concentration in the fluid covering the airway surface.


Subject(s)
Amino Acids/pharmacology , Bronchi/physiology , Carrier Proteins/metabolism , Cell Membrane/physiology , Epithelial Cells/physiology , Alanine/pharmacology , Amiloride/pharmacology , Amino Acid Transport Systems , Arginine/pharmacology , Cell Membrane/drug effects , Cells, Cultured , Epithelial Cells/drug effects , Humans , Kinetics , Lysine/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Nitric Oxide Donors/pharmacology , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Proline/pharmacology , S-Nitroso-N-Acetylpenicillamine
17.
J Cell Physiol ; 177(2): 214-23, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9766518

ABSTRACT

Transforming growth factor beta (TGFbeta) determines a nearly complete inhibition of cystogenesis by MDCK cells grown in collagen I-enriched matrices in vitro. In order to elucidate the mechanism implicated in this phenomenon, we performed a series of experiments aimed at discovering a relevant role of extracellular matrix. TGFbeta (2 ng/ml) played a marked stimulatory effect on the expression of extracellular matrix by MDCK with a selective effect on collagen V (three to fourfold increase of protein and mRNA) and in parallel inhibited cystogenesis by 95%. Cotreatment with TGFbeta and anti-collagen V antibodies restored a normal cystogenesis. In analogy, when MDCK cells were grown in three-dimensional matrices containing collagen I and minor (10%) amounts of collagen V, cystogenesis was once again inhibited by 95%. To characterize the molecular mechanism activated by TGFbeta and collagen V, we looked at the electrophysiological characteristics of MDCK monolayers and found a drastic fall of transepithelial electrical resistance (TER) in both conditions. In parallel with the decrease in TER, TGFbeta and collagen V also induced the leakage of two high molecular weight tracers, i.e., [3H]-inulin and 150 kD FITC-Dextran, suggesting a perturbation of the paracellular permeability. Finally, TGFbeta at the relevant concentration did not stimulate apoptosis in our cellular model, as judged by propidium iodide staining and by in situ end labeling of DNA fragments. These observations suggest that TGFbeta inhibits cystogenesis by MDCK cells in vitro by altering the collagenic composition of the three-dimensional milieu where MDCK cells grow and form cysts. The molecular mechanism responsible for inhibition of cystogenesis is the increase of paracellular flux which overcomes the active transport of solutes and water inside cysts.


Subject(s)
Collagen/physiology , Epithelial Cells/physiology , Extracellular Matrix/physiology , Kidney Diseases, Cystic/etiology , Transforming Growth Factor beta/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Biological Transport/drug effects , Cell Line , Cell Membrane/ultrastructure , Dogs , Drug Synergism , Extracellular Matrix/drug effects , Inulin/metabolism , Kidney/cytology , Kidney Diseases, Cystic/metabolism , Membrane Potentials/drug effects , Transforming Growth Factor beta/physiology , Tritium
18.
J Membr Biol ; 165(3): 255-64, 1998 Oct 01.
Article in English | MEDLINE | ID: mdl-9767679

ABSTRACT

The question of whether a single molecule can account for every observed swelling-activated Cl- current deserves to be addressed and biophysical description seems to be an adequate criterion to classify these channels. We studied the biophysical properties of swelling-activated Cl- currents in 9HTEo-cells using whole-cell and outside-out patch clamp recordings. Hypotonic shock activated outwardly rectifying currents that inactivated at potentials higher than 20 mV. The decay phase of the current was well fitted by two exponential functions and both time constants were voltage-dependent. Two voltage-dependent time constants were also necessary to describe reactivation. The midpoint of current inactivation was 54 mV. The voltage dependence of kinetics did not significantly change by modifying the extracellular NaCl concentration while the inactivation midpoint slightly shifted. In conclusion, our results indicate that the voltage-dependent properties of the swelling-activated Cl- currents in 9HTEo- cells are largely independent from the extracellular ionic strength and the extracellular Cl- concentration. Excised patches from cells exposed to hypotonic shock showed single channel currents that inactivated at positive membrane potentials and displayed chord conductance of approximately 60 pS at 100 mV and of approximately 20 pS at -80 mV. The permeability sequence for the single channel was I- > Br- > Cl- > gluconate and currents were blocked by Reactive blue 2. These properties indicate that intermediate conductance outwardly rectifying channels are responsible for the macroscopic swelling-activated current.


Subject(s)
Chloride Channels/physiology , Epithelial Cells/physiology , Cell Line, Transformed , Cell Membrane/physiology , Humans , Membrane Potentials/physiology , Osmolar Concentration , Patch-Clamp Techniques , Simian virus 40/genetics , Sodium Chloride/pharmacology , Trachea
19.
In Vitro Cell Dev Biol Anim ; 34(6): 478-81, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9661051

ABSTRACT

Electrophysiological studies of human bronchial epithelial cells in vitro are limited by the scarcity of biological material available for primary culture. To overcome this problem, we set up a protocol in which the cell number is first enlarged in LHC9/RPMI 1640 serum-free medium for up to six passages, each passage giving a four- to eightfold amplification. The cells are then plated at high density on permeable supports. Cell differentiation, monitored by measuring transepithelial potential difference (PD) and electrical resistance (R), is induced with a medium containing serum and a cocktail of different supplements and hormones. Maximal values of PD and R, obtained after 4-7 d of culture on permeable supports, are around -50 mV and 3000-4000 omega/cm2, respectively. Ussing chamber experiments show that basal short-circuit current (Isc) is partially inhibited by the epithelial Na+ channel blocker amiloride. Stimulation with a cAMP-elevating agent induces a Isc increase that is inhibited by the cystic fibrosis transmembrane conductance regulator (CFTR) blocker glibenclamide. Our culture protocol provides a large number of differentiated bronchial epithelial cell monolayers starting from a low amount of material. This characteristic is useful for in vitro studies of ion transport in airway epithelium.


Subject(s)
Cell Culture Techniques/methods , Epithelial Cells/cytology , Bronchi/cytology , Cell Differentiation , Culture Media , Culture Media, Serum-Free , Epithelial Cells/physiology , Humans
20.
Gene ; 209(1-2): 59-63, 1998 Mar 16.
Article in English | MEDLINE | ID: mdl-9524223

ABSTRACT

Expression cloning revealed a chloride channel (ICln) that we found to be fundamental for the regulatory volume decrease in a variety of cells. The chromosomal localization of the human ICln-gene showed two loci, one at chromosome 11 in position q13.5-q14.1, termed CLNS1A, and a second one at chromosome 6 at position p12.1-q13, termed CLNS1B. In this study, we offer a detailed characterization of the CLNS1A gene and provide the exact position (6p12) and sequence data of CLNS1B, an intronless gene 91.3% homologous to the coding region of CLNS1A.


Subject(s)
Chloride Channels/genetics , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 6 , Ion Channels , Amino Acid Sequence , Base Sequence , Chloride Channels/biosynthesis , Chloride Channels/chemistry , Chromosome Mapping , DNA Primers , Exons , Genomic Library , Humans , In Situ Hybridization, Fluorescence , Introns , Molecular Sequence Data , Open Reading Frames , Polymerase Chain Reaction , Sequence Alignment , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...