Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Am J Physiol Cell Physiol ; 283(1): C126-34, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12055080

ABSTRACT

Some members of the epithelial Na+ channel/degenerin (ENaC/DEG) family of ion channels have been detected in mammalian brain. Therefore, we examined the RNA and protein expression of these channels in another part of the central nervous system, the rabbit retina. We next sought to demonstrate physiological evidence for an amiloride-sensitive current in Müller glia, which, on the basis of a previous study, are thought to express alpha-ENaC (Golestaneh N, de Kozak Y, Klein C, and Mirshahi M. Glia 33: 160-168, 2001). RT-PCR of retinal RNA revealed the presence of alpha-, beta-, gamma-, and delta-ENaC as well as acid-sensing ion channel (ASIC)1, ASIC2, ASIC3, and ASIC4. Immunohistochemical localization with antibodies against alpha-ENaC and beta-ENaC showed labeling in Müller cells and neurons, respectively. The presence of alpha-ENaC, beta-ENaC, and ASIC1 was detected by Western blotting. Cultured Müller cells were whole cell patch clamped. These cells exhibited an inward Na+ current that was blocked by amiloride. These data demonstrate for the first time both the expression of a variety of ENaC and ASIC subunits in the rabbit retina as well as distinct cellular expression patterns of specific subunits in neurons and glia.


Subject(s)
Ion Channels/metabolism , Membrane Proteins , Nerve Tissue Proteins/metabolism , Neuroglia/metabolism , Neurons/metabolism , Retina/metabolism , Sodium Channels/metabolism , Acid Sensing Ion Channels , Animals , Cell Line , Cells, Cultured , Degenerin Sodium Channels , Dogs , Electric Conductivity , Epithelial Sodium Channels , Ion Channels/genetics , Nerve Tissue Proteins/genetics , Neuroglia/physiology , Neurons/physiology , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Rabbits , Retina/cytology , Retina/physiology , Sodium Channels/genetics
2.
J Biol Chem ; 276(9): 6621-30, 2001 Mar 02.
Article in English | MEDLINE | ID: mdl-11110786

ABSTRACT

These studies provide evidence that cystic fibrosis transmembrane conductance regulator (CFTR) potentiates and accelerates regulatory volume decrease (RVD) following hypotonic challenge by an autocrine mechanism involving ATP release and signaling. In wild-type CFTR-expressing cells, CFTR augments constitutive ATP release and enhances ATP release stimulated by hypotonic challenge. CFTR itself does not appear to conduct ATP. Instead, ATP is released by a separate channel, whose activity is potentiated by CFTR. Blockade of ATP release by ion channel blocking drugs, gadolinium chloride (Gd(3+)) and 4,4'-diisothiocyanatostilbene-2,2'disulfonic acid (DIDS), attenuated the effects of CFTR on acceleration and potentiation of RVD. These results support a key role for extracellular ATP and autocrine and paracrine purinergic signaling in the regulation of membrane ion permeability and suggest that CFTR potentiates ATP release by stimulating a separate ATP channel to strengthen autocrine control of cell volume regulation.


Subject(s)
Adenosine Triphosphate/metabolism , Cell Size , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Animals , COS Cells , Chloride Channels/physiology , Gadolinium/pharmacology
3.
J Biol Chem ; 275(36): 27947-56, 2000 Sep 08.
Article in English | MEDLINE | ID: mdl-10821834

ABSTRACT

Epithelial sodium channel (ENaC) and cystic fibrosis transmembrane conductance regulator (CFTR) are co-localized in the apical membrane of many epithelia. These channels are essential for electrolyte and water secretion and/or reabsorption. In cystic fibrosis airway epithelia, a hyperactivated epithelial Na(+) conductance operates in parallel with defective Cl(-) secretion. Several groups have shown that CFTR down-regulates ENaC activity, but the mechanisms and the regulation of CFTR by ENaC are unknown. To test the hypothesis that ENaC and CFTR regulate each other, and to identify the region(s) of ENaC involved in the interaction between CFTR and ENaC, rENaC and its mutants were co-expressed with CFTR in Xenopus oocytes. Whole cell macroscopic sodium currents revealed that wild type (wt) alphabetagamma-rENaC-induced Na(+) current was inhibited by co-expression of CFTR, and further inhibited when CFTR was activated with a cAMP-raising mixture (CKT). Conversely, alphabetagamma-rENaC stimulated CFTR-mediated Cl(-) currents up to approximately 6-fold. Deletion mutations in the intracellular tails of the three rENaC subunits suggested that the carboxyl terminus of the beta subunit was required both for the down-regulation of ENaC by activated CFTR and the up-regulation of CFTR by ENaC. However, both the carboxyl terminus of the beta subunit and the amino terminus of the gamma subunit were essential for the down-regulation of rENaC by unstimulated CFTR. Interestingly, down-regulation of rENaC by activated CFTR was Cl(-)-dependent, while stimulation of CFTR by rENaC was not dependent on either cytoplasmic Na(+) or a depolarized membrane potential. In summary, there appear to be at least two different sites in ENaC involved in the intermolecular interaction between CFTR and ENaC.


Subject(s)
Cell Membrane/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Sodium Channels/chemistry , Sodium Channels/physiology , Amiloride/pharmacology , Animals , Binding Sites , Cell Membrane/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Epithelial Sodium Channels , Gene Expression Regulation , Green Fluorescent Proteins , Luminescent Proteins/analysis , Macromolecular Substances , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mutagenesis, Site-Directed , Oocytes/physiology , Protein Biosynthesis , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Sodium/metabolism , Sodium Channels/genetics , Xenopus laevis
4.
J Biol Chem ; 274(53): 37845-54, 1999 Dec 31.
Article in English | MEDLINE | ID: mdl-10608849

ABSTRACT

The hypothesis that actin interactions account for the signature biophysical properties of cloned epithelial Na(+) channels (ENaC) (conductance, ion selectivity, and long mean open and closed times) was tested using planar lipid bilayer reconstitution and patch clamp techniques. We found the following. 1) In bilayers, actin produced a more than 2-fold decrease in single channel conductance, a 5-fold increase in Na(+) versus K(+) permselectivity, and a substantial increase in mean open and closed times of wild-type alphabetagamma-rENaC but had no effect on a mutant form of rENaC in which the majority of the C terminus of the alpha subunit was deleted (alpha(R613X)betagamma-rENaC). 2) When alpha(R613X)betagamma-rENaC was heterologously expressed in oocytes and single channels examined by patch clamp, 12.5-pS channels of relatively low cation permeability were recorded. These characteristics were identical to those recorded in bilayers for either alpha(R613X)betagamma-rENaC or wild-type alphabetagamma-rENaC in the absence of actin. Moreover, we show that rENaC subunits tightly associate, forming either homo- or heteromeric complexes when prepared by in vitro translation or when expressed in oocytes. Finally, we show that alpha-rENaC is properly assembled but retained in the endoplasmic reticulum compartment. We conclude that actin subserves an important regulatory function for ENaC and that planar bilayers are an appropriate system in which to study the biophysical and regulatory properties of these cloned channels.


Subject(s)
Actins/metabolism , Lipid Bilayers , Sodium Channels/metabolism , Amino Acid Sequence , Animals , Base Sequence , DNA Primers , Endoplasmic Reticulum/metabolism , Epithelial Sodium Channels , Microscopy, Confocal , Microscopy, Fluorescence , Oocytes/metabolism , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Sodium Channels/genetics , Xenopus
5.
Biophys J ; 75(5): 2292-301, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9788924

ABSTRACT

The molecular composition of a core conduction element formed by the alpha-subunit of cloned epithelial Na+ channels (ENaC) was studied in planar lipid bilayers. Two pairs of in vitro translated proteins were employed in combinatorial experiments: 1) wild-type (WT) and an N-terminally truncated alphaDeltaN-rENaC that displays accelerated kinetics (tauo = 32 +/- 13 ms, tauc = 42 +/- 11 ms), as compared with the WT channel (tauc1 = 18 +/- 8 ms, tauc2 = 252 +/- 31 ms, and tauo = 157 +/- 43 ms); and 2) WT and an amiloride binding mutant, alphaDelta278-283-rENaC. The channels that formed in a alphaWT:alphaDeltaN mixture fell into two groups: one with tauo and tauc that corresponded to those exhibited by the alphaDeltaN-rENaC alone, and another with a double-exponentially distributed closed time and a single-exponentially distributed open time that corresponded to the alphaWT-rENaC alone. Five channel subtypes with distinct sensitivities to amiloride were found in a 1alphaWT:1alphaDelta278-283 protein mixture. Statistical analyses of the distributions of channel phenotypes observed for either set of the WT:mutant combinations suggest a tetrameric organization of alpha-subunits as a minimal model for the core conduction element in ENaCs.


Subject(s)
Amiloride/pharmacology , Sodium Channels/chemistry , Animals , Cloning, Molecular , Electrophysiology , Kinetics , Lipid Bilayers/metabolism , Protein Binding/physiology , Proteolipids/chemistry , Recombinant Proteins/metabolism , Sequence Deletion/genetics , Sodium Channels/genetics , Xenopus
6.
Am J Physiol ; 275(2): C449-58, 1998 08.
Article in English | MEDLINE | ID: mdl-9688599

ABSTRACT

We reported the identification of three outwardly rectified Cl- channel (ORCC) candidate proteins (115, 85, and 52 kDa) from bovine tracheal epithelia. We have raised polyclonal antibodies against these isolated proteins. Incorporation into planar lipid bilayers of material partly purified from bovine tracheal apical membranes with one of these antibodies as a ligand (anti-p115) resulted in the incorporation of an ORCC identical in biophysical characteristics to one we previously described. We developed a new purification procedure to increase the yield and purity of this polypeptide. The purification scheme that gave the best results in terms of overall protein yield and purity was a combination of anion- and cation-exchange chromatography followed by immunopurification. By use of this purification scheme, 7 microg of the 115-kDa protein were purified from 20 mg of tracheal apical membrane proteins. Incorporation of this highly purified material into planar lipid bilayers revealed a DIDS-inhibitable channel with the following properties: linear conductance of 87 +/- 9 pS in symmetrical Cl- solutions, halide selectivity sequence of I- > Cl- > Br-, and lack of sensitivity to protein kinase A, Ca2+, or dithiothreitol. Using anti-Galphai antibodies to precipitate Galphai protein(s) from the partly purified preparations, we demonstrated that the loss of rectification of the ORCC was due to uncoupling of Galphai protein(s) from the ORCC protein and that the 115-kDa polypeptide is an ORCC.


Subject(s)
Calcium Channels/physiology , Epithelial Cells/physiology , Trachea/physiology , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Animals , Antibodies , Blotting, Western , Calcium Channels/biosynthesis , Calcium Channels/isolation & purification , Cattle , Chlorides/metabolism , Chromatography, Ion Exchange , Electric Conductivity , Electrophoresis, Polyacrylamide Gel , Epithelial Cells/cytology , GTP-Binding Proteins/metabolism , Lipid Bilayers , Macromolecular Substances , Membrane Potentials , Molecular Weight , Trachea/cytology
7.
Nature ; 390(6657): 302-5, 1997 Nov 20.
Article in English | MEDLINE | ID: mdl-9384384

ABSTRACT

The cystic fibrosis gene encodes a cyclic AMP-gated chloride channel (CFTR) that mediates electrolyte transport across the luminal surfaces of a variety of epithelial cells. The molecular mechanisms that modulate CFTR activity in epithelial tissues are poorly understood. Here we show that CFTR is regulated by an epithelially expressed syntaxin (syntaxin 1A), a membrane protein that also modulates neurosecretion and calcium-channel gating in brain. Syntaxin 1A physically interacts with CFTR chloride channels and regulates CFTR-mediated currents both in Xenopus oocytes and in epithelial cells that normally express these proteins. The physical and functional interactions between syntaxin 1A and CFTR are blocked by a syntaxin-binding protein of the Munc18 protein family (also called n-Secl). Our results indicate that CFTR function in epithelial cells is regulated by an interplay between syntaxin and Munc18 isoforms.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Cystic Fibrosis , Membrane Proteins/physiology , Nerve Tissue Proteins/physiology , Vesicular Transport Proteins , Animals , Cell Line , Chlorides/metabolism , Colon/cytology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Humans , Isomerism , Membrane Proteins/genetics , Molecular Sequence Data , Munc18 Proteins , Oocytes , Patch-Clamp Techniques , Qa-SNARE Proteins , Recombinant Proteins , Syntaxin 1 , Xenopus , Xenopus Proteins
8.
Am J Physiol ; 272(4 Pt 1): C1077-86, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9142832

ABSTRACT

Cystic fibrosis (CF) airway epithelia exhibit enhanced Na+ reabsorption in parallel with diminished Cl- secretion. We tested the hypothesis that actin plays a role in the regulation of a cloned epithelial Na+ channel (ENaC) by the cystic fibrosis transmembrane conductance regulator (CFTR). We found that immunopurified bovine tracheal CFTR coreconstituted into a planar lipid bilayer with alpha,beta,gamma-rat ENaC (rENaC) decreased single-channel open probability (Po) of rENaC in the presence of actin by over 60%, a significantly greater effect than was observed in the absence of actin (approximately 20%). In the presence of actin, protein kinase A plus ATP activated both CFTR and rENaC, but CFTR was activated in a sustained manner, whereas the activation of rENaC was transitory. ATP alone could also activate ENaC transiently in the presence ofactin but had no effect on CFTR. Stabilizing short actin filaments at a fixed length with gelsolin (at a ratio to actin of 2:1) produced a sustained activation of alpha,beta,gamma-rENaC in both the presence or absence of CFTR. Gelsolin alone (i.e., in the absence of actin) had no effect on the conductance or Po of either CFTR or rENaC. We have also found that short actin filaments produced their modulatory action on alpha-rENaC independent of the presence of the beta- or gamma-rENaC subunits. In contrast, CFTR did not affect any properties of the channel formed by alpha-rENaC alone, i.e., in the absence of beta- or gamma-rENaC. These results indicate that CFTR can directly downregulate single Na+ channel activity, which may account for the observed differences between Na+ transport in normal and CF-affected airway epithelia. Moreover, the presence of actin confers an enhanced modulatory ability of CFTR on Na+ channels.


Subject(s)
Actins/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Respiratory System/metabolism , Sodium Channels/metabolism , Actins/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Cattle , Cyclic AMP-Dependent Protein Kinases/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/pharmacology , Epithelium/metabolism , Gelsolin/pharmacology , Isomerism , Lipid Bilayers/metabolism , Rabbits , Rats , Sodium Channels/drug effects
9.
J Biol Chem ; 271(30): 17704-10, 1996 Jul 26.
Article in English | MEDLINE | ID: mdl-8663510

ABSTRACT

Cytoskeletal elements play an important role in the regulation of ion transport in epithelia. We have studied the effects of actin filaments of different length on the alpha, beta, gamma-rENaC (rat epithelial Na+ channel) in planar lipid bilayers. We found the following. 1) Short actin filaments caused a 2-fold decrease in unitary conductance and a 2-fold increase in open probability (Po) of alpha,beta,gamma-rENaC. 2) alpha,beta,gamma-rENaC could be transiently activated by protein kinase A (PKA) plus ATP in the presence, but not in the absence, of actin. 3) ATP in the presence of actin was also able to induce a transitory activation of alpha, beta,gamma-rENaC, although with a shortened time course and with a lower magnitude of change in Po. 4) DNase I, an agent known to prohibit elongation of actin filaments, prevented activation of alpha,beta,gamma-rENaC by ATP or PKA plus ATP. 5) Cytochalasin D, added after rundown of alpha,beta,gamma-rENaC activity following ATP or PKA plus ATP treatment, produced a second transient activation of alpha,beta,gamma-rENaC. 6) Gelsolin, a protein that stabilizes polymerization of actin filaments at certain lengths, evoked a sustained activation of alpha,beta,gamma-rENaC at actin/gelsolin ratios of <32:1, with a maximal effect at an actin/gelsolin ratio of 2:1. These results suggest that short actin filaments activate alpha, beta,gamma-rENaC. PKA-mediated phosphorylation augments activation of this channel by decreasing the rate of elongation of actin filaments. These results are consistent with the hypothesis that cloned alpha,beta,gamma-rENaCs form a core conduction unit of epithelial Na+ channels and that interaction of these channels with other associated proteins, such as short actin filaments, confers regulation to channel activity.


Subject(s)
Actins/metabolism , Cytoskeleton/metabolism , Ion Channel Gating , Sodium Channels/metabolism , Actins/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Epithelium/metabolism , Lipid Bilayers/metabolism , Rats , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Sodium Channels/drug effects , Sodium Channels/genetics
10.
J Biol Chem ; 271(9): 4725-32, 1996 Mar 01.
Article in English | MEDLINE | ID: mdl-8617738

ABSTRACT

Cystic fibrosis airway epithelia exhibit enhanced Na+ reabsorption in parallel with diminished Cl- secretion. We tested the hypothesis that the cystic fibrosis transmembrane conductance regulator (CFTR) directly affects epithelial Na+ channel activity by co-incorporating into planar lipid bilayers immunopurified bovine tracheal CFTR and either heterologously expressed rat epithelial Na+ channel ( alpha,b eta,gamma-rENaC) or an immunopurified bovine renal Na+ channel protein complex. The single channel open probability (Po) of rENaC was decreased by 24% in the presence of CFTR. Protein kinase A (PKA) plus ATP activated CFTR, but did not have any effect on rENaC. CFTR also decreased the extent of elevation of the renal Na+ channel Po following PKA-mediated phosphorylation. Moreover, the presence of CFTR prohibited the inward rectification of the gating of this renal Na+ channel normally induced by PKA-mediated phosphorylation, thus down-regulating inward Na+ current. This interaction between CFTR and Na+ channels occurs independently of whether or not wild-type CFTR is conducting anions. However, the nonconductive CFTR mutant, G551D CFTR, cannot substitute for the wild-type molecule. Our results indicate that CFTR can directly down-regulate single Na+ channel activity, thus accounting, at least in part, for the observed differences in Na+ transport between normal and cystic fibrosis-affected airway epithelia.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Kidney Medulla/physiology , Sodium Channels/physiology , Adenosine Triphosphate/metabolism , Amiloride/pharmacology , Animals , Cattle , Cell Membrane/drug effects , Cell Membrane/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/pharmacology , Electric Conductivity , Epithelium/physiology , Female , Gene Expression , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Lipid Bilayers , Membrane Potentials/drug effects , Oocytes/physiology , Phosphorylation , Point Mutation , Protein Kinases/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Sodium Channels/biosynthesis , Sodium Channels/isolation & purification
11.
J Biol Chem ; 271(9): 4776-80, 1996 Mar 01.
Article in English | MEDLINE | ID: mdl-8617745

ABSTRACT

Experiments were designed to test if immunopurified outwardly rectified chloride channels (ORCCs) and the cystic fibrosis transmembrane conductance regulator (CFTR) incorporated into planar lipid bilayers are regulated by G-proteins. pertussis toxin (PTX) (100 ng/ml) + NAD (1 mM) + ATP (1 mM) treatment of ORCC and CFTR in bilayers resulted in a 2-fold increase in single channel open probability (Po) of ORCC but not of CFTR. Neither PTX, NAD, nor ATP alone affected the biophysical properties of either channel. Further, PTX conferred a linearity to the ORCC current-voltage curve, with a slope conductance of 80 +/- 3 picosiemens (pS) in the +/- 100 mV range of holding potentials. PKA-mediated phosphorylation of these PTX + NAD-treated channels further increased the Po of the linear 80-pS channels from 0.66 +/- 0.05 to >0.9, and revealed the presence of a small (16 +/- 2 pS) linear channel in the membrane. PTX treatment of a CFTR-immunodepleted protein preparation incorporated into bilayer membranes resulted in a similar increase in the Po of the larger conductance channel and restored PKA-sensitivity that was lost after CFTR immunodepletion. The addition of guanosine 5'-3-O-(thio)triphosphate (100 mum) to the cytoplasmic bathing solutions decreased the activity of the ORCC and increased its rectification at both negative and positive voltages. ADP-ribosylation of immunopurified material revealed the presence of a 41-kDa protein. These results demonstrate copurification of a channel-associated G-protein that is involved in the regulation of ORCC function.


Subject(s)
Chloride Channels/physiology , GTP-Binding Proteins/metabolism , Lipid Bilayers , Adenosine Diphosphate Ribose/metabolism , Adenosine Triphosphate/pharmacology , Animals , Cattle , Cell Membrane/physiology , Chloride Channels/drug effects , Chloride Channels/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Epithelium/physiology , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Kinetics , Membrane Potentials/drug effects , NAD/pharmacology , Pertussis Toxin , Phosphorylation , Trachea/physiology , Virulence Factors, Bordetella/pharmacology
12.
J Biol Chem ; 270(49): 29194-200, 1995 Dec 08.
Article in English | MEDLINE | ID: mdl-7493947

ABSTRACT

We have previously described a protocol for the simultaneous isolation and reconstitution of a protein kinase A (PKA)-sensitive outwardly rectified chloride channel (ORCC) and the cystic fibrosis transmembrane conductance regulator (CFTR) from bovine tracheal epithelium. Immunoprecipitation of CFTR from this preparation prevented PKA activation of the ORCC, suggesting that CFTR regulated the ORCC and that this regulatory relationship was preserved throughout the purification procedure. We now report the purification of CFTR from bovine tracheal epithelia and the purification of a CFTR conduction mutant (G551D CFTR) from retrovirally transduced mouse L cells using a combination of alkali stripping, Triton-X extraction, and immunoaffinity chromatography. Immunopurified CFTR proteins were reconstituted in the absence and presence of ORCC. To test the hypothesis that only functional CFTR can support activation of ORCC by PKA and ATP, we used an inhibitory anti-CFTR505-511 peptide antibody or G551D CFTR. When anti-CFTR505-511 peptide antibodies were present prior to the addition of PKA and ATP, activation of both the ORCC and CFTR was prevented. If the antibody was added after activation of the ORCC and CFTR Cl- channels by PKA and ATP, only the CFTR Cl- channel was inhibited. When ORCC and G551D CFTR were co-incorporated into planar bilayers, only the ORCC was recorded and this channel could not be further activated by the addition of PKA and ATP. Thus, functional CFTR is required for activation of the ORCC by PKA and ATP. We also tested the hypothesis that PKA activation of ORCC was dependent on the extracellular presence of ATP. We added ATP on the presumed extracellular side of the lipid bilayer under conditions where it was not possible to activate the ORCC, i.e. in the presence of inhibitory anti-CFTR505-511 antibody or G551D CFTR. In both cases the ORCC regained PKA sensitivity. Moreover, the addition of hexokinase + glucose to the extracellular side prevented activation of the ORCCs by PKA and ATP in the presence of CFTR. These experiments confirm that both the presence of CFTR as well as the presence of ATP on the extracellular side is required for activation of the ORCC by PKA and ATP.


Subject(s)
Chloride Channels/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Adenosine Triphosphate/pharmacology , Animals , Cattle , Cyclic AMP-Dependent Protein Kinases/pharmacology , Mice , Rabbits , Recombinant Proteins/pharmacology
13.
J Biol Chem ; 270(4): 1521-8, 1995 Jan 27.
Article in English | MEDLINE | ID: mdl-7530244

ABSTRACT

Our laboratory has developed a protocol for the isolation of a 140-kDa protein that forms an anion-selective channel when reconstituted into planar lipid bilayers. Polyclonal antibodies have been raised against the 38-kDa component of this purified protein. This channel has a linear current-voltage relationship and is not activated by protein kinase A (PKA) plus ATP. Using the same antibody and a modified purification protocol (eliminating the ion exchange chromatography steps), we isolated and reconstituted two other anion channels from tracheal membrane vesicles. In vitro phosphorylation of these isolated proteins by PKA and ATP revealed four bands migrating at 52, 85, 120, and 174 kDa. Immunoprecipitation experiments with anti-CFTR antibodies indicate that the 174-kDa phosphoprotein was CFTR. Upon incorporation of these isolated proteins into planar bilayers, an anion channel that exhibited a marked outward rectification in symmetrical Cl- solutions with a slope conductance of 82 pS at depolarizing voltages was observed. PKA and ATP increased channel activity but only from one side of the bilayer. However, channel activity was unaffected by addition of ATP alone from either side of the membrane. DIDS (100 microM) applied to the opposite side of the bilayer to which PKA and ATP act, blocked channel activity. A linear anion-selective channel with a conductance of 16 pS could be also resolved after inhibition of the outwardly rectified anion channel by DIDS in the presence of PKA and ATP. This small conductance channel was inhibited by 300 microM diphenylamine-2-carboxylic acid. Immunodepletion of the 174-kDa phosphoprotein from the preparation prevented activation of the 82-pS outwardly rectified anion channel by PKA and ATP. However, the PKA-dependent in vitro phosphorylation of the 52-, 85-, and 120-kDa phosphoproteins was unaffected by the absence of CFTR. Our results suggest a direct regulatory relationship between an outwardly rectified anion channel and CFTR.


Subject(s)
Chloride Channels/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Membrane Proteins/physiology , Trachea/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Animals , Antibodies , Autoradiography , Blotting, Western , Cattle , Cell Membrane/metabolism , Chloride Channels/drug effects , Chloride Channels/isolation & purification , Chromatography, Ion Exchange , Cystic Fibrosis Transmembrane Conductance Regulator , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Epithelium/metabolism , Kinetics , Lipid Bilayers , Membrane Potentials/drug effects , Membrane Proteins/drug effects , Membrane Proteins/isolation & purification , Phosphorus Radioisotopes , Phosphorylation
14.
Am J Physiol ; 266(5 Pt 2): F775-84, 1994 May.
Article in English | MEDLINE | ID: mdl-8203561

ABSTRACT

The role of intracellular Ca2+ in the development and maintenance of epithelial tight junctional integrity is poorly understood. We assessed tight junctional resistance (Rj) in confluent monolayers of A6 cells that were treated with mucosal amiloride such that the transepithelial resistance (Rt) reflects Rj. Solution Ca2+ concentration [Ca2+] was reduced by ethylene glycol-bis(beta-aminoethyl ether)-N,N,N',N'-tetraacetic acid (EGTA) addition to the bathing solutions. Reduction of mucosal [Ca2+] to 1 microM or reduction of serosal Ca2+ to 100 microM did not significantly alter Rt. However, a further decrease of serosal Ca2+ to 40 microM caused the resistance to fall to < 12% of the control value. Following restoration of serosal [Ca2+], Rt increased to a new steady-state value within approximately 15 min. The magnitude of recovery of Rt was inversely correlated with the length of time the epithelium was exposed to low serosal [Ca2+]. To further test the effects of asymmetric Ca2+ removal, the serosal [Ca2+] was chelated using EGTA to reduce Rt. When the Ca2+ ionophore A-23187 was subsequently added to the mucosal solution, Rt increased from 20% to 60% of the control level. In addition, cells were loaded with the fluorescent Ca2+ indicator, Calcium Green, and the temporal relationship between changes in Rt and intracellular Ca2+ was determined. Following removal of serosal Ca2+, cell Ca2+ decreased, followed by a decrease in Rt. In contrast, returning Ca2+ to the serosal bathing solution resulted in a parallel increase of both Rt and cell [Ca2+]. These data strongly suggest that changes in intracellular [Ca2+] play an important role in the regulation of Rj.


Subject(s)
Amiloride/pharmacology , Calcium/metabolism , Intercellular Junctions/physiology , Animals , Calcimycin/pharmacology , Cell Line , Egtazic Acid/pharmacology , Epithelium/drug effects , Epithelium/physiology , Intercellular Junctions/drug effects , Kidney , Kinetics , Mathematics , Mucous Membrane/drug effects , Mucous Membrane/physiology , Time Factors
15.
Am J Physiol ; 261(6 Pt 1): C1196-203, 1991 Dec.
Article in English | MEDLINE | ID: mdl-1767820

ABSTRACT

We describe a convenient nonelectrophysiological technique for assessing cell proliferation and subsequent tight junction formation for epithelial monolayers grown on permeable supports. The method involves the use of phenol red (PR), a standard pH indicator in most cell culture media. In addition, we report a systematic error in a commercially available system for measuring transepithelial electrical properties. Briefly, the flux of PR across the epithelium was measured from the serosal solution into the mucosal solution. The mucosal solution was first replaced with a PR-free solution and then collected at timed intervals. The PR concentration was measured using a spectrophotometer set at the isosbestic point for PR (479 nm). PR flux was then calculated and used as an index of the permeability of the epithelium to PR. This method was tested using the renal epithelial cell line A6. After cell seeding, PR flux decreased in two phases: an initial large decrease, associated with cell growth and monolayer confluence, and a second decrease associated with tight junction formation [assessed by measuring transepithelial conductance (Gt)]. In addition to monitoring tight junction formation, PR flux measurements were also used to estimate the net movement of solution by the epithelial cells between the mucosal and serosal compartments. For convenience, Gt was initially measured in culture dishes using a commercially available "chopstick" electrode system. However, the chopstick system yielded Gt values that were on average 51% lower than values for the same preparations when measured in standard Ussing-type chambers. The discrepancy was due to a nonuniform current field produced by the chopstick electrodes.


Subject(s)
Cell Division , Epithelial Cells , Intercellular Junctions/physiology , Phenolsulfonphthalein , Spectrum Analysis/methods , Cell Line , Cell Membrane Permeability , Electric Conductivity , Epithelium/physiology , Evaluation Studies as Topic , Hydrogen-Ion Concentration , Kinetics , Osmolar Concentration , Phenolsulfonphthalein/metabolism , Solutions
16.
Am J Physiol ; 259(6 Pt 1): C869-82, 1990 Dec.
Article in English | MEDLINE | ID: mdl-1701967

ABSTRACT

Urinary kallikreins are proteolytic enzymes known to be secreted by distal nephron tubules. In this study, we demonstrate (using the chromogenic tripeptide substrate S 2266) that the renal cell line A6 from Xenopus laevis secretes a kallikrein-like enzyme. Secretion is present only when the cells are grown on filters, and enzyme is secreted only into the apical membrane bathing solution. Enzyme secretion consists of two components, one soybean trypsin inhibitor (SBTI) sensitive (SSBTI) and the other insensitive to SBTI (ISBTI). Both enzymes were inhibited by aprotinin, a kallikrein-like enzyme inhibitor. Using a bioassay, only the ISBTI enzyme produced a hypotensive effect on blood pressure and is thus a kallikrein-like enzyme. The apical membrane of cells grown on filters contains both enzyme species, whereas the basolateral membrane contains only the ISBTI (kallikrein-like) enzyme. Both enzymes were present in the apical membrane of cells grown on plastic. Initiation of enzyme secretion occurred after the cells formed electrically tight monolayers and the increase in membrane activity always preceded enzyme secretion. Using an irreversible inhibitor of the apical membrane-bound enzymes, the turnover rate for the SSBTI and ISBTI enzymes (cells on filters) was 3 and 7 h, respectively. Because the recovery of enzyme secretion was proportional to the recovery of membrane-bound enzyme activities, this suggests that enzyme secretion is due to the release of membrane-bound enzyme.


Subject(s)
Isoenzymes/metabolism , Kallikreins/metabolism , Kidney/enzymology , Animals , Aprotinin/pharmacology , Blood Pressure/drug effects , Cell Line , Cell Membrane/enzymology , Culture Media , Epithelium/enzymology , Isoenzymes/pharmacology , Kallikreins/pharmacology , Kinetics , Mathematics , Models, Theoretical , Rats , Rats, Inbred Strains , Thermodynamics , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...