Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters










Publication year range
1.
Acta Physiol (Oxf) ; 202(3): 523-31, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21251238

ABSTRACT

Anion transport drives fluid into the airways and is essential for humidifying inspired air and supplying surface liquid for mucociliary transport. Despite the importance of airway secretion in diseases such as cystic fibrosis, the cellular mechanisms remain poorly understood, in part due to the small size and complicated structure of the submucosal glands that produce most of the fluid. The Calu-3 human lung adenocarcinoma cell line has become a popular model for studying airway secretion because it can be cultured as a flat sheet, expresses the cystic fibrosis transmembrane conductance regulator and several acinar cell markers, forms polarized monolayers with tight junctions, has robust cAMP-stimulated anion transport, and responds to secretagogues that regulate the glands in vivo. However, some properties of Calu-3 cells are less consistent with those of native tissue. In particular, Calu-3 monolayers do not secrete chloride when stimulated by forskolin under short-circuit conditions. Bicarbonate ions are thought to carry the short-circuit current (I(sc)) and the drive secretion of alkaline fluid, in contrast to the neutral pH secretions that are produced by submucosal glands. Calu-3 cells also have abnormal chromosomes and characteristics of both serous and mucus cells. In this article, we discuss Calu-3 as a model in light of our ongoing studies, which suggest that Calu-3 monolayers resemble submucosal glands more closely than was previously thought. For example, we find that net HCO(3)(-) flux fully accounts for I(sc) as previously suggested but Cl(-) is the main anion transported under physiological conditions. A novel, HCO(3)(-) -dependent mechanism of Cl(-) transport is emerging which may explain secretion by Calu-3 and perhaps other epithelial cells.


Subject(s)
Anions/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelium/metabolism , Ion Transport/physiology , Cell Line, Tumor , Humans , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism
2.
J Physiol ; 548(Pt 1): 39-52, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12588899

ABSTRACT

Activation of the cystic fibrosis transmembrane conductance regulator (CFTR) channel by protein kinase A (PKA) is enhanced by protein kinase C (PKC). However, the mechanism of modulation is not known and it remains uncertain whether PKC acts directly on CFTR or through phosphorylation of an ancillary protein. Using excised patches that had been pre-treated with phosphatases, we found that PKC exposure results in much larger PKA-activated currents and shifts the PKA concentration dependence. To examine if these effects are mediated by direct PKC phosphorylation of CFTR, a mutant was constructed in which serines or threonines at nine PKC consensus sequences on CFTR were replaced by alanines (i.e. the '9CA' mutant T582A/T604A/S641A/T682A/S686A/S707A/S790A/T791A/S809A). In excised patches, 9CA channels had greatly reduced responses to PKA (i.e. 5-10 % that of wild-type), which were not enhanced by PKC pre-treatment, although the mutant channels were still functional according to iodide efflux assays. Stimulation of iodide efflux by chlorophenylthio-cAMP (cpt-cAMP) was delayed in cells expressing 9CA channels, and a similar delay was observed when cells expressing wild-type CFTR were treated with the PKC inhibitor chelerythrine. This suggests that weak activation by PKA in excised patches and slow stimulation of iodide efflux from intact cells are specifically due to the loss of PKC phosphorylation. Finally, PKC caused a slight activation of wild-type channels when added to excised patches after phosphatase pre-treatment but had no effect on the mutant. We conclude that direct phosphorylation of CFTR at one or more of the nine sites mutated in 9CA is required for both the partial activation by PKC and for its modulation of CFTR responses to PKA.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , Cyclic AMP/analogs & derivatives , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Protein Kinase C/metabolism , Alkaloids , Amino Acid Sequence , Animals , Benzophenanthridines , Binding Sites , Cattle , Cell Membrane/drug effects , Cell Membrane/metabolism , Cricetinae , Cyclic AMP/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Electrophysiology , Enzyme Inhibitors/pharmacology , Immunoblotting , Iodides/metabolism , Membrane Potentials/physiology , Molecular Sequence Data , Mutation/physiology , Patch-Clamp Techniques , Phenanthridines/pharmacology , Phosphorylation , Rats , Thionucleotides/pharmacology
3.
J Membr Biol ; 189(2): 131-41, 2002 Sep 15.
Article in English | MEDLINE | ID: mdl-12235488

ABSTRACT

Changes in amino-acid sequence of the unique pore-forming protein of H. influenzae (OmpP2; porin) have been associated with increased antimicrobial resistance in H. influenzae strains isolated from cystic fibrosis patients. From patients who were subjected to long-term antimicrobial therapy, H. influenzae strains 67d and 69a (patient 27) and strains 77a and 77f (patient 30) were isolated. Strains 67d and 77a were previously shown to have elevated values for minimal inhibitory concentrations of antibiotics compared to strains 69a and 77f. Porins were extracted from all four H. influenzae strains by detergent treatment and purified to homogeneity by ion exchange chromatography. By reconstitution of the clinical Hi porins into planar lipid bilayers, single-channel conductance, ionic selectivity, and voltage-gating characteristics were assessed. Porins 77a and 77f displayed similar single-channel conductance and ionic selectivity. Current-voltage relationships were determined for the different porins: porin 77f displayed substantial voltage gating at both positive and negative polarity; porin 77a gated at negative polarity only. Porins 67d and 69a showed substantial differences in their pore-forming properties: the single-channel conductance of porin 69a was significantly increased (1.05 nS) relative to porin 67d (0.73 nS). Porin 67d was twice as permeable to cations as porin 69a, and at both positive and negative polarities the extent of voltage gating was greater for porin 67d relative to porin 69a. Expression of the porins in an isogenic, porin-deleted H. influenzae background allowed for assessment of the contribution of each porin to the minimum inhibitory concentrations of various antimicrobial compounds. Porin 67d was found to have a decreased susceptibility to the antimicrobials novobiocin and streptomycin. This decreased susceptibility of porin 67d to novobiocin and streptomycin correlates with its decrease in single-channel conductance.


Subject(s)
Drug Resistance, Bacterial , Haemophilus influenzae/classification , Haemophilus influenzae/genetics , Ion Channels/physiology , Lipid Bilayers/chemistry , Porins/chemistry , Amino Acid Sequence , Cystic Fibrosis/genetics , Cystic Fibrosis/microbiology , Electric Conductivity , Escherichia coli/classification , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation , Haemophilus influenzae/drug effects , Haemophilus influenzae/metabolism , Humans , Ion Channel Gating/physiology , Lactams/classification , Lactams/pharmacology , Membrane Potentials , Microbial Sensitivity Tests , Molecular Sequence Data , Mutagenesis, Site-Directed , Porins/genetics , Porins/isolation & purification , Porins/metabolism , Proteolipids/chemistry , Reference Values , Sensitivity and Specificity , Species Specificity , Statistics as Topic
4.
Biochemistry ; 40(48): 14621-8, 2001 Dec 04.
Article in English | MEDLINE | ID: mdl-11724575

ABSTRACT

Porin (341 amino acids; M(r) 37 782) of Haemophilus influenzae type b mediates exchange of solutes between the external environment and the periplasm of this Gram-negative bacterium. Positively charged residues in the extracellular loops have been shown to be involved in the voltage gating of this protein. To further elucidate our observations on the functional properties of this channel, we mutated seven lysines (Lys(48), Lys(161), Lys(165), Lys(170), Lys(248), Lys(250), and Lys(253)) to glutamic acid. The selected residues were previously shown to be accessible to chemical modification, and they map to three locations: loop 4 and loop 6, and within the barrel lumen. The seven mutant proteins were purified, and each was reconstituted into planar lipid bilayers to characterize its channel forming properties. The single substitution mutant porins displayed increased single channel conductances in 1 M KCl ranging between 134 and 178% of the single channel conductance for wild-type Hib porin. Six of the seven mutant porins also displayed altered current-voltage relationships when compared to wild-type Hib porin. Whereas Lys(170)Glu had activity similar to wild-type Hib porin, Lys(48)Glu, Lys(248)Glu, and Lys(253)Glu showed substantial voltage gating at both positive and negative polarities. Lys(161)Glu and Lys(250)Glu gated only at negative potentials, and Lys(165)Glu gated only at positive potentials. Rather than ascribing one specific loop in gating, our analyses of these mutant Hib porins suggest that voltage gating can be attributed to contributions from loops 4 and 6 and a residue within the barrel lumen.


Subject(s)
Haemophilus influenzae type b/genetics , Ion Channel Gating/physiology , Porins/genetics , Amino Acid Sequence , Amino Acid Substitution , Cell Membrane Permeability/physiology , DNA Primers/chemistry , Electric Conductivity , Haemophilus influenzae type b/chemistry , Lipid Bilayers/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Porins/chemistry , Protein Structure, Secondary
5.
Biochemistry ; 40(22): 6620-7, 2001 Jun 05.
Article in English | MEDLINE | ID: mdl-11380256

ABSTRACT

The cystic fibrosis transmembrane conductance regulator (CFTR) Cl(-) channel contains 12 membrane-spanning regions which are presumed to form the transmembrane pore. Although a number of findings have suggested that the sixth transmembrane region plays a key role in forming the pore and determining its functional properties, the role of other transmembrane regions is currently not well established. Here we assess the functional importance of the twelfth transmembrane region, which occupies a homologous position in the carboxy terminal half of the CFTR molecule to that of the sixth transmembrane region in the amino terminal half. Five residues in potentially important regions of the twelfth transmembrane region were mutated individually to alanines, and the function of the mutant channels was examined using patch clamp recording following expression in mammalian cell lines. Three of the five mutations significantly weakened block of unitary Cl(-) currents by SCN(-), implying a partial disruption of anion binding within the pore. Two of these mutations also caused a large reduction in the steady-state channel mean open probability, suggesting a role for the twelfth transmembrane region in channel gating. However, in direct contrast to analogous mutations in the sixth transmembrane region, all mutants studied here had negligible effects on the anion selectivity and unitary Cl(-) conductance of the channel. The relatively minor effects of these five mutations on channel permeation properties suggests that, despite their symmetrical positions within the CFTR protein, the sixth and twelfth transmembrane regions make highly asymmetric contributions to the functional properties of the pore.


Subject(s)
Chlorides/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Peptide Fragments/genetics , Amino Acid Sequence , Animals , Anions , CHO Cells , Cell Line , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/genetics , Cricetinae , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Cystic Fibrosis Transmembrane Conductance Regulator/biosynthesis , Electric Conductivity , Humans , Membrane Potentials/drug effects , Membrane Potentials/genetics , Molecular Sequence Data , Patch-Clamp Techniques , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/physiology , Protein Structure, Tertiary/genetics , Thiocyanates/pharmacology , Transfection
6.
Pflugers Arch ; 443 Suppl 1: S92-6, 2001.
Article in English | MEDLINE | ID: mdl-11845311

ABSTRACT

Cystic fibrosis transmembrane conductance regulator (CFTR) chloride channels are regulated tightly by protein kinases and phosphatases. The regulatory domain of CFTR has about 20 potential sites for phosphorylation by protein kinases A (PKA) and C (PKC). The reason for this large number of sites is not known, however their conservation from fish to humans implies that they play important roles in vivo. PKA is an important activator, and its stimulation of CFTR is enhanced by PKC via mechanisms which are not fully understood. The physiological stimuli of CFTR are not known for some epithelia, and it appears likely that other serine/threonine and even tyrosine kinases also regulate CFTR in particular tissues. Phosphatases that deactivate CFTR have yet to be identified definitively at the molecular level, however CFTR is regulated by a membrane-bound form of protein phosphatase-2C (PP2C) in several cell types. Patch-clamp studies of channel rundown, co-immunoprecipitation, chemical cross-linking studies, and pull-down assays all indicate that CFTR and PP2C are closely associated within a stable regulatory complex. Understanding the regulation of CFTR by PP2C is a priority due to its potential as a target for pharmacotherapies in the treatment of cystic fibrosis.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Saccharomyces cerevisiae Proteins , Cystic Fibrosis/metabolism , Humans , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Protein Kinases/metabolism , Protein Phosphatase 2 , Protein Phosphatase 2C
7.
Am J Physiol Cell Physiol ; 279(1): C108-19, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10898722

ABSTRACT

Genistein and bromotetramisole (Br-t) strongly activate cystic fibrosis transmembrane conductance regulator (CFTR; ABCC7) chloride channels on Chinese hamster ovary cells and human airway epithelial cells. We have examined the possible role of phosphatases in stimulation by these drugs using patch-clamp and biochemical methods. Genistein inhibited the spontaneous rundown of channel activity that occurs after membrane patches are excised from cAMP-stimulated cells but had no effect on purified protein phosphatase type 1 (PP1), PP2A, PP2B, PP2C, or endogenous phosphatases when assayed as [(32)P]PO(4) release from prelabeled casein, recombinant GST-R domain fusion protein, or immunoprecipitated full-length CFTR. Br-t also slowed rundown of CFTR channels, but, in marked contrast to genistein, it did inhibit all four protein phosphatases tested. Half-maximal inhibition of PP2A and PP2C was observed with 0.5 and 1.5 mM Br-t, respectively. Protein phosphatases were also sensitive to (+)-p-Br-t, a stereoisomer of Br-t that does not inhibit alkaline phosphatases. Br-t appeared to act exclusively through phosphatases since it did not affect CFTR channels in patches that had low apparent endogenous phosphatase activity (i.e., those lacking spontaneous rundown). We conclude that genistein and Br-t act through different mechanisms. Genistein stimulates CFTR without inhibiting phosphatases, whereas Br-t acts by inhibiting a membrane-associated protein phosphatase (probably PP2C) that presumably allows basal phosphorylation to accumulate.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Enzyme Inhibitors/pharmacology , Genistein/pharmacology , Phosphoprotein Phosphatases/physiology , Tetramisole/analogs & derivatives , Animals , CHO Cells , Cricetinae , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Isoenzymes/metabolism , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism , Phosphorylation/drug effects , Precipitin Tests , Subcellular Fractions/enzymology , Tetramisole/pharmacology
8.
Biophys J ; 78(6): 2973-82, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10827976

ABSTRACT

Ionic selectivity in many cation channels is achieved over a short region of the pore known as the selectivity filter, the molecular determinants of which have been identified in Ca(2+), Na(+), and K(+) channels. However, a filter controlling selectivity among different anions has not previously been identified in any Cl(-) channel. In fact, because Cl(-) channels are only weakly selective among small anions, and because their selectivity has proved so resistant to site-directed mutagenesis, the very existence of a discrete anion selectivity filter has been called into question. Here we show that mutation of a putative pore-lining phenylalanine residue, F337, in the sixth membrane-spanning region of the cystic fibrosis transmembrane conductance regulator (CFTR) Cl(-) channel, dramatically alters the relative permeabilities of different anions in the channel. Specifically, mutations that reduce the size of the amino acid side chain present at this position virtually abolish the relationship between anion permeability and hydration energy, a relationship that characterizes the anion selectivity not only of wild-type CFTR, but of most classes of Cl(-) channels. These results suggest that the pore of CFTR may indeed contain a specialized region, analogous to the selectivity filter of cation channels, at which discrimination between different permeant anions takes place. Because F337 is adjacent to another amino acid residue, T338, which also affects anion selectivity in CFTR, we suggest that selectivity is predominantly determined over a physically discrete region of the pore located near these important residues.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Amino Acid Substitution , Animals , Anions , Cell Line , Cell Membrane Permeability , Cricetinae , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Phenylalanine , Point Mutation , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Substrate Specificity , Transfection
10.
J Membr Biol ; 178(3): 185-93, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11140274

ABSTRACT

Porin of Haemophilus influenzae type b (341 amino acids; M(r) 37782) determines the permeability of the outer membrane to low molecular mass compounds. Purified Hib porin was subjected to chemical modification of lysine residues by succinic anhydride. Electrospray ionization mass spectrometry identified up to 12 modifications per porin molecule. Tryptic digestion of modified Hib porin followed by reverse phase chromatography and matrix assisted laser desorption ionization time-of-flight mass spectrometry mapped the succinylation sites. Most modified lysines are positioned in surface-located loops, numbers 1 and 4 to 7. Succinylated porin was reconstituted into planar lipid bilayers, and biophysical properties were analyzed and compared to Hib porin: there was an increased average single channel conductance compared to Hib porin (1.24 +/- 0.41 vs. 0.85 +/- 0.40 nanosiemens). The voltage-gating activity of succinylated porin differed considerably from that of Hib porin. The threshold voltage for gating was decreased from 75 to 40 mV. At 80 mV, steady-state conductance for succinylated porin was 50-55% of the instantaneous conductance. Hib porin at 80 mV showed a decrease to 89-91% of the instantaneous current levels. We propose that surface-located lysine residues are determinants of voltage gating for porin of Haemophilus influenzae type b.


Subject(s)
Haemophilus influenzae/chemistry , Ion Channel Gating/physiology , Porins/chemistry , Amino Acid Sequence , Cell Membrane Permeability/physiology , Electric Conductivity , Lipid Bilayers , Lysine/chemistry , Models, Molecular , Molecular Sequence Data , Molecular Weight , Peptide Mapping , Protein Conformation , Protein Structure, Tertiary , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Structure-Activity Relationship , Succinic Acid/chemistry
11.
Biochim Biophys Acta ; 1461(2): 275-83, 1999 Dec 06.
Article in English | MEDLINE | ID: mdl-10581361

ABSTRACT

CFTR possesses a large cluster of strict dibasic consensus sites for phosphorylation by protein kinase A (PKA) in the R-domain and an obligatory dependence on phosphorylation is a hallmark of CFTR Cl(-) channel function. Removal of as many as 11 of these sites reduces the conformational change in the R-domain and the degree of channel activation in response to PKA. However, until recently a completely PKA-unresponsive CFTR variant has not been reported, leaving open the possibility that the residual response may be mediated by associating ancillary phosphoproteins. We traced the residual PKA-catalyzed (32)P-labelling of the variant with 11 sites mutagenized (11SA) to distinct CNBr phosphopeptides within the R-domain. Mutagenesis of 4 additional monobasic sites in these segments produced a 15SA variant in which Cl(-) channel response to PKA was abolished. Therefore, it can be concluded that ancillary phosphoproteins do not contribute to CFTR activation by PKA. Notably, however, the 15SA protein did exhibit a low level of constitutive channel activity not dependent on PKA, which might have reflected a down-regulating effect of phosphorylation of one or two of the 15 sites as suggested by others. However, this did not prove to be the case.Since immature CFTR has been claimed to be active in the endoplasmic reticulum (ER), we also examined whether it can be phosphorylated in cells and what influence if any this might have on its susceptibility to degradation. Teleologically, activation by phosphorylation of CFTR Cl(-) channels in the ER might be undesirable to the cell. Using various phosphorylation site mutants and kinase and phosphatase inhibitors in pulse-chase experiments, we have found that although nascent CFTR can be phosphorylated at the ER, this is without effect on its ability to mature and avoid proteolysis. Furthermore, we found that microsomes from cells expressing CFTR processing mutants such as DeltaF508 do not generate Cl(-) active channels when fused with planar bilayers unless maturation is promoted, e.g. by growth of cells at reduced temperature or other means. We conclude that the ER-retained mutant nascent chains which are incapable of maturation may be phosphorylated but do not form active channels. Stimulation by PKA of the insertion of CFTR containing vesicles into the plasma membrane as part of the mechanism of stimulation of chloride secretion has been reported, as has an influence of CFTR on the balance between endocytosis and exocytosis but these findings have not been universally confirmed.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Membrane/metabolism , Cyclic AMP-Dependent Protein Kinases/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Endoplasmic Reticulum/metabolism , Enzyme Activation , Humans , Ion Channel Gating , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Protein Conformation
12.
J Biol Chem ; 274(41): 29102-7, 1999 Oct 08.
Article in English | MEDLINE | ID: mdl-10506164

ABSTRACT

Cystic fibrosis transmembrane conductance regulator (CFTR) chloride channels are rapidly deactivated by a membrane-bound phosphatase activity. The efficiency of this regulation suggests CFTR and protein phosphatases may be associated within a regulatory complex. In this paper we test that possibility using co-immunoprecipitation and cross-linking experiments. A monoclonal anti-CFTR antibody co-precipitated type 2C protein phosphatase (PP2C) from baby hamster kidney cells stably expressing CFTR but did not co-precipitate PP1, PP2A, or PP2B. Conversely, a polyclonal anti-PP2C antibody co-precipitated CFTR from baby hamster kidney membrane extracts. Exposing baby hamster kidney cell lysates to dithiobis (sulfosuccinimidyl propionate) caused the cross-linking of histidine-tagged CFTR (CFTR(His10)) and PP2C into high molecular weight complexes that were isolated by chromatography on Ni(2+)-nitrilotriacetic acid-agarose. Chemical cross-linking was specific for PP2C, because PP1, PP2A, and PP2B did not co-purify with CFTR(His10) after dithiobis (sulfosuccinimidyl propionate) exposure. These results suggest CFTR and PP2C exist in a stable complex that facilitates regulation of the channel.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Phosphoprotein Phosphatases/metabolism , Saccharomyces cerevisiae Proteins , Animals , Antibodies, Monoclonal/metabolism , CHO Cells , Cell Fractionation , Cells, Cultured , Cricetinae , Cross-Linking Reagents , Phosphoprotein Phosphatases/analysis , Phosphorylation , Precipitin Tests , Protein Phosphatase 2 , Protein Phosphatase 2C , Succinimides
13.
Gastroenterology ; 116(6): 1342-7, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10348817

ABSTRACT

BACKGROUND & AIMS: When rice is incorporated into oral rehydration therapy for patients with secretory diarrhea, clinical outcomes improve. We have shown that a factor purified from boiled rice (RF) blocks the secretory response of intestinal crypt cells to adenosine 3',5'-cyclic monophosphate (cAMP). Now we report that the cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel is the cellular target for this rice inhibitor. METHODS: We used RF, the same previously described extract prepared from boiled rice, to assess chloride channel activation in vitro, measuring (1) cell volume regulation of guinea pig intestinal crypt epithelial cell suspensions using standard Coulter counter technology, (2) transepithelial chloride current in monolayers of T84 cells mounted in Ussing chambers, and (3) whole-cell and single-channel currents using the patch-clamp technique in cells transfected to express CFTR. RESULTS: RF inhibited activation by cAMP of CFTR chloride channels in all experimental preparations; RF did not block volume-stimulated Cl- secretion, suggesting that its effect might be specific for CFTR chloride channels. RF inhibited transepithelial cAMP-stimulated Cl- current in T84 cells and inhibited forskolin (i.e., cAMP)-induced current in cells transfected with CFTR. Excised patch and single-channel patch-clamp recordings supported the view that the response was a direct effect on CFTR rather than on cAMP signal transduction. CONCLUSIONS: RF exerts a specific inhibitory effect on CFTR chloride channels, blocking activation from the luminal surface of the cell and reversing established activation. Many major diarrheal states are based on cAMP-induced CFTR activation, leading to excessive gut secretion; our findings could have clinical relevance.


Subject(s)
Chlorides/metabolism , Cooking , Intestinal Mucosa/metabolism , Oryza , Animals , CHO Cells , Cell Line/drug effects , Cell Line/metabolism , Chlorides/physiology , Cricetinae , Cyclic AMP/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Electric Conductivity , Guinea Pigs , Humans , Intestines/cytology , Oryza/chemistry , Patch-Clamp Techniques , Plant Extracts/pharmacology , Transfection
14.
Br J Pharmacol ; 126(6): 1471-7, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10217542

ABSTRACT

1. The effects of physiological substrates of multidrug resistance-associated proteins (MRPs) on cystic fibrosis transmembrane conductance regulator (CFTR) Cl- channel currents were examined using patch clamp recording from CFTR-transfected mammalian cell lines. 2. Two MRP substrates, taurolithocholate-3-sulphate (TLCS) and beta-estradiol 17-(beta-D-glucuronide) (E217betaG) caused a voltage-dependent block of macroscopic CFTR Cl- currents when applied to the intracellular face of excised membrane patches, with mean apparent dissociation constants (KDs) of 96+/-10 and 563+/-103 microM (at 0 mV) respectively. The unconjugated bile salts taurocholate and cholate were also effective CFTR channel blockers under these conditions, with KDs of 453+/-44 and 3760+/-710 microM (at 0 mV) respectively. 3. Reducing the extracellular Cl- concentration from 154 to 20 mM decreased the KD for block intracellular TLCS to 54+/-1 microM, and also significantly reduced the voltage dependence of block, by suggesting that TLCS blocks Cl- permeation through CFTR by binding within the channel pore. 4. Intracellular TLCS reduced the apparent amplitude of CFTR single channel currents, suggesting that the duration of block is very fast compared to the gating of the channel. 5. The apparent affinity of block by TLCs is comparable to that of other well-known CFTR channel blockers, suggesting that MRP substrates may comprise a novel class of probes of the CFTR channel pore. 6. These results also suggest that the related proteins CFTR and MRP may share a structurally similar anion binding site at the cytoplasmic face of the membrane.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Estradiol/pharmacology , Taurolithocholic Acid/analogs & derivatives , Animals , CHO Cells , Calcium Channel Blockers/pharmacology , Chlorides/pharmacology , Cholates/pharmacology , Cricetinae , Dose-Response Relationship, Drug , Electric Stimulation , Estradiol/metabolism , Glyburide/pharmacology , Membrane Potentials/drug effects , Multidrug Resistance-Associated Proteins , Stilbenes/pharmacology , Taurocholic Acid/pharmacology , Taurolithocholic Acid/metabolism , Taurolithocholic Acid/pharmacology , ortho-Aminobenzoates/pharmacology
15.
J Physiol ; 512 ( Pt 1): 1-16, 1998 Oct 01.
Article in English | MEDLINE | ID: mdl-9729613

ABSTRACT

1. The effects of individually mutating two adjacent threonine residues in the sixth membrane-spanning region (TM6) of the cystic fibrosis transmembrane conductance regulator (CFTR) Cl- channel on permeation properties were examined using patch clamp recording from mammalian cell lines stably expressing human CFTR. 2. A number of mutations of T338 significantly affected the permeation properties of the channel. Increases and decreases in single channel conductance were observed for different mutants. Anion selectivity was strongly affected, with no two channel variants sharing the same selectivity sequence. Several mutations led to strong inward rectification of the macroscopic current-voltage relationship. The effects of these mutations on permeation properties were correlated with the size of the amino acid side chain substituted, rather than its chemical nature. 3. Most mutations of T339 resulted in a lack of functional channel expression and apparent misprocessing of the protein. One mutant, T339V, was characterized in detail; its permeation properties were significantly altered, although these effects were not as strong as for T338 mutations. 4. These results suggest an important role for T338 in controlling the permeation properties of the CFTR Cl- channel. It is suggested that mutation of this residue alters the interaction between permeating anions and the channel pore via an indirect effect on the orientation of the TM6 helix.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Protein Structure, Secondary , Amino Acid Substitution , Animals , Anions , CHO Cells , Cell Line , Cell Membrane Permeability , Cricetinae , Cystic Fibrosis Transmembrane Conductance Regulator/biosynthesis , Humans , Kidney , Mammals , Membrane Potentials , Methotrexate/pharmacology , Models, Molecular , Mutagenesis, Site-Directed , Patch-Clamp Techniques , Point Mutation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transfection
16.
Am J Physiol ; 275(1): C323-6, 1998 07.
Article in English | MEDLINE | ID: mdl-9688865

ABSTRACT

The cystic fibrosis transmembrane conductance regulator (CFTR) forms an ion channel that is permeable both to Cl- and to larger organic anions. Here we show, using macroscopic current recording from excised membrane patches, that the anionic antioxidant tripeptide glutathione is permeant in the CFTR channel. This permeability may account for the high concentrations of glutathione that have been measured in the surface fluid that coats airway epithelial cells. Furthermore, loss of this pathway for glutathione transport may contribute to the reduced levels of glutathione observed in airway surface fluid of cystic fibrosis patients, which has been suggested to contribute to the oxidative stress observed in the lung in cystic fibrosis. We suggest that release of glutathione into airway surface fluid may be a novel function of CFTR.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Glutathione/metabolism , Animals , Biological Transport , Cell Line , Cell Membrane/physiology , Cell Membrane Permeability , Cricetinae , Glutathione Disulfide/metabolism , Kinetics , Membrane Potentials , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transfection
18.
Am J Physiol ; 274(5): C1397-410, 1998 05.
Article in English | MEDLINE | ID: mdl-9612228

ABSTRACT

Cystic fibrosis transmembrane conductance regulator (CFTR) Cl- channel activity declines rapidly when excised from transfected Chinese hamster ovary (CHO) or human airway cells because of membrane-associated phosphatase activity. In the present study, we found that CFTR channels usually remained active in patches excised from baby hamster kidney (BHK) cells overexpressing CFTR. Those patches with stable channel activity were used to investigate the regulation of CFTR by exogenous protein phosphatases (PP). Adding PP2A, PP2C, or alkaline phosphatase to excised patches reduced CFTR channel activity by > 90% but did not abolish it completely. PP2B caused weak deactivation, whereas PP1 had no detectable effect on open probability (Po). Interestingly, the time course of deactivation by PP2C was identical to that of the spontaneous rundown observed in some patches after excision. PP2C and PP2A had distinct effects on channel gating Po declined during exposure to exogenous PP2C (and during spontaneous rundown, when it was observed) without any change in mean burst duration. By contrast, deactivation by exogenous PP2A was associated with a dramatic shortening of burst duration similar to that reported previously in patches from cardiac cells during deactivation of CFTR by endogenous phosphatases. Rundown of CFTR-mediated current across intact T84 epithelial cell monolayers was insensitive to toxic levels of the PP2A inhibitor calyculin A. These results demonstrate that exogenous PP2C is a potent regulator of CFTR activity, that its effects on single-channel gating are distinct from those of PP2A but similar to those of endogenous phosphatases in CHO, BHK, and T84 epithelial cells, and that multiple protein phosphatases may be required for complete deactivation of CFTR channels.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Ion Channels/metabolism , Kidney/metabolism , Phosphoprotein Phosphatases/pharmacology , Phosphoric Monoester Hydrolases/pharmacology , Saccharomyces cerevisiae Proteins , Animals , Animals, Newborn/physiology , CHO Cells/metabolism , Cell Line , Cricetinae , Electrophysiology , Enzyme Inhibitors/pharmacology , Humans , Ion Channels/drug effects , Ion Channels/physiology , Kidney/cytology , Magnesium/pharmacology , Marine Toxins , Oxazoles/pharmacology , Patch-Clamp Techniques , Protein Phosphatase 2 , Protein Phosphatase 2C
19.
J Physiol ; 508 ( Pt 2): 365-77, 1998 Apr 15.
Article in English | MEDLINE | ID: mdl-9508802

ABSTRACT

1. The relationship between phosphorylation of the cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel and its gating by nucleotides was examined using the patch clamp technique by comparing strongly phosphorylated wild-type (WT) channels with weakly phosphorylated mutant channels lacking four (4SA) or all ten (10SA) dibasic consensus sequences for phosphorylation by protein kinase A (PKA). 2. The open probability (Po) of strongly phosphorylated WT channels in excised patches was about twice that of 4SA and 10SA channels, after correcting for the number of functional channels per patch by addition of adenylylimidodiphosphate (AMP-PNP). The mean burst durations of WT and mutant channels were similar, and therefore the elevated Po of WT was due to its higher bursting rate. 3. The ATP dependence of the 10SA mutant was shifted to higher nucleotide concentrations compared with WT channels. The relationship between Po and [ATP] was noticeably sigmoid for 10SA channels (Hill coefficient, 1.8), consistent with positive co-operativity between two sites. Increasing ATP concentration to 10 mM caused the Po of both WT and 10SA channels to decline. 4. Wild-type and mutant CFTR channels became locked in open bursts when exposed to mixtures of ATP and the non-hydrolysable analogue AMP-PNP. The rate at which the low phosphorylation mutants became locked open was about half that of WT channels, consistent with Po being the principal determinant of locking rate in WT and mutant channels. 5. We conclude that phosphorylation at 'weak' PKA sites is sufficient to sustain the interactions between the ATP binding domains that mediate locking by AMP-PNP. Phosphorylation of the strong dibasic PKA sites controls the bursting rate and Po of WT channels by increasing the apparent affinity of CFTR for ATP.


Subject(s)
Chloride Channels/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Nucleotides/pharmacology , Adenosine Triphosphate/pharmacology , Animals , CHO Cells , Chloride Channels/drug effects , Chloride Channels/genetics , Cricetinae , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Ion Channel Gating/drug effects , Ion Channel Gating/genetics , Kinetics , Mutation , Phosphorylation
20.
J Gen Physiol ; 111(4): 601-14, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9524141

ABSTRACT

The cystic fibrosis transmembrane conductance regulator (CFTR) forms a tightly regulated channel that mediates the passive diffusion of Cl- ions. Here we show, using macroscopic current recording from excised membrane patches, that CFTR also shows significant, but highly asymmetrical, permeability to a broad range of large organic anions. Thus, all large organic anions tested were permeant when present in the intracellular solution under biionic conditions (PX/PCl = 0.048-0.25), whereas most were not measurably permeant when present in the extracellular solution. This asymmetry was not observed for smaller anions. ATPase inhibitors that "lock" CFTR channels in the open state (pyrophosphate, 5'-adenylylimidodiphosphate) disrupted the asymmetry of large anion permeation by allowing their influx from the extracellular solution, which suggests that ATP hydrolysis is required to maintain asymmetric permeability. The ability of CFTR to allow efflux of large organic anions represents a novel function of CFTR. Loss of this function may contribute to the pleiotropic symptoms seen in cystic fibrosis.


Subject(s)
Adenosine Triphosphate/physiology , Anions/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Adenosine Triphosphate/metabolism , Animals , CHO Cells , Cell Line , Chlorides/antagonists & inhibitors , Chlorides/physiology , Cricetinae , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Electric Conductivity , Gluconates/antagonists & inhibitors , Gluconates/metabolism , Hydrolysis , Mutation/physiology , Permeability
SELECTION OF CITATIONS
SEARCH DETAIL
...