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










Publication year range
2.
Pflugers Arch ; 474(7): 681-697, 2022 07.
Article in English | MEDLINE | ID: mdl-35525869

ABSTRACT

How phosphorylation of the epithelial sodium channel (ENaC) contributes to its regulation is incompletely understood. Previously, we demonstrated that in outside-out patches ENaC activation by serum- and glucocorticoid-inducible kinase isoform 1 (SGK1) was abolished by mutating a serine residue in a putative SGK1 consensus motif RXRXX(S/T) in the channel's α-subunit (S621 in rat). Interestingly, this serine residue is followed by a highly conserved proline residue rather than by a hydrophobic amino acid thought to be required for a functional SGK1 consensus motif according to in vitro data. This suggests that this serine residue is a potential phosphorylation site for the dual-specificity tyrosine phosphorylated and regulated kinase 2 (DYRK2), a prototypical proline-directed kinase. Its phosphorylation may prime a highly conserved preceding serine residue (S617 in rat) to be phosphorylated by glycogen synthase kinase 3 ß (GSK3ß). Therefore, we investigated the effect of DYRK2 on ENaC activity in outside-out patches of Xenopus laevis oocytes heterologously expressing rat ENaC. DYRK2 included in the pipette solution significantly increased ENaC activity. In contrast, GSK3ß had an inhibitory effect. Replacing S621 in αENaC with alanine (S621A) abolished the effects of both kinases. A S617A mutation reduced the inhibitory effect of GKS3ß but did not prevent ENaC activation by DYRK2. Our findings suggest that phosphorylation of S621 activates ENaC and primes S617 for subsequent phosphorylation by GSK3ß resulting in channel inhibition. In proof-of-concept experiments, we demonstrated that DYRK2 can also stimulate ENaC currents in microdissected mouse distal nephron, whereas GSK3ß inhibits the currents.


Subject(s)
Epithelial Sodium Channels , Protein Serine-Threonine Kinases , Animals , Epithelial Sodium Channels/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Mice , Oocytes/metabolism , Phosphorylation , Proline/metabolism , Rats , Serine/metabolism , Xenopus laevis/metabolism
3.
J Biol Chem ; 298(6): 102004, 2022 06.
Article in English | MEDLINE | ID: mdl-35504352

ABSTRACT

The epithelial sodium channel (ENaC) is a heterotrimer consisting of α-, ß-, and γ-subunits. Channel activation requires proteolytic release of inhibitory tracts from the extracellular domains of α-ENaC and γ-ENaC; however, the proteases involved in the removal of the γ-inhibitory tract remain unclear. In several epithelial tissues, ENaC is coexpressed with the transmembrane serine protease 2 (TMPRSS2). Here, we explored the effect of human TMPRSS2 on human αßγ-ENaC heterologously expressed in Xenopus laevis oocytes. We found that coexpression of TMPRSS2 stimulated ENaC-mediated whole-cell currents by approximately threefold, likely because of an increase in average channel open probability. Furthermore, TMPRSS2-dependent ENaC stimulation was not observed using a catalytically inactive TMPRSS2 mutant and was associated with fully cleaved γ-ENaC in the intracellular and cell surface protein fractions. This stimulatory effect of TMPRSS2 on ENaC was partially preserved when inhibiting its proteolytic activity at the cell surface using aprotinin but was abolished when the γ-inhibitory tract remained attached to its binding site following introduction of two cysteine residues (S155C-Q426C) to form a disulfide bridge. In addition, computer simulations and site-directed mutagenesis experiments indicated that TMPRSS2 can cleave γ-ENaC at sites both proximal and distal to the γ-inhibitory tract. This suggests a dual role of TMPRSS2 in the proteolytic release of the γ-inhibitory tract. Finally, we demonstrated that TMPRSS2 knockdown in cultured human airway epithelial cells (H441) reduced baseline proteolytic activation of endogenously expressed ENaC. Thus, we conclude that TMPRSS2 is likely to contribute to proteolytic ENaC activation in epithelial tissues in vivo.


Subject(s)
Epithelial Sodium Channels , Oocytes , Serine Endopeptidases , Animals , Epithelial Sodium Channels/metabolism , Humans , Ion Transport/physiology , Oocytes/metabolism , Peptide Hydrolases/metabolism , Proteolysis , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Xenopus laevis/genetics
4.
J Biol Chem ; 296: 100404, 2021.
Article in English | MEDLINE | ID: mdl-33577799

ABSTRACT

Mice lacking connexin 30 (Cx30) display increased epithelial sodium channel (ENaC) activity in the distal nephron and develop salt-sensitive hypertension. This indicates a functional link between Cx30 and ENaC, which remains incompletely understood. Here, we explore the effect of Cx30 on ENaC function using the Xenopus laevis oocyte expression system. Coexpression of human Cx30 with human αßγENaC significantly reduced ENaC-mediated whole-cell currents. The size of the inhibitory effect on ENaC depended on the expression level of Cx30 and required Cx30 ion channel activity. ENaC inhibition by Cx30 was mainly due to reduced cell surface ENaC expression resulting from enhanced ENaC retrieval without discernible effects on proteolytic channel activation and single-channel properties. ENaC retrieval from the cell surface involves the interaction of the ubiquitin ligase Nedd4-2 with PPPxY-motifs in the C-termini of ENaC. Truncating the C- termini of ß- or γENaC significantly reduced the inhibitory effect of Cx30 on ENaC. In contrast, mutating the prolines belonging to the PPPxY-motif in γENaC or coexpressing a dominant-negative Xenopus Nedd4 (xNedd4-CS) did not significantly alter ENaC inhibition by Cx30. Importantly, the inhibitory effect of Cx30 on ENaC was significantly reduced by Pitstop-2, an inhibitor of clathrin-mediated endocytosis, or by mutating putative clathrin adaptor protein 2 (AP-2) recognition motifs (YxxФ) in the C termini of ß- or γ-ENaC. In conclusion, our findings suggest that Cx30 inhibits ENaC by promoting channel retrieval from the plasma membrane via clathrin-dependent endocytosis. Lack of this inhibition may contribute to increased ENaC activity and salt-sensitive hypertension in mice with Cx30 deficiency.


Subject(s)
Clathrin/metabolism , Connexin 30/pharmacology , Epithelial Sodium Channels/chemistry , Nedd4 Ubiquitin Protein Ligases/metabolism , Oocytes/physiology , Animals , Endocytosis , Epithelial Sodium Channels/metabolism , Humans , Oocytes/cytology , Patch-Clamp Techniques/methods , Signal Transduction , Xenopus laevis
5.
Pflugers Arch ; 470(7): 1087-1102, 2018 07.
Article in English | MEDLINE | ID: mdl-29589117

ABSTRACT

The bile acid-sensitive ion channel (BASIC) is a member of the ENaC/degenerin family of ion channels. It is activated by bile acids and inhibited by extracellular Ca2+. The aim of this study was to explore the molecular mechanisms mediating these effects. The modulation of BASIC function by extracellular Ca2+ and tauro-deoxycholic acid (t-DCA) was studied in Xenopus laevis oocytes heterologously expressing human BASIC using the two-electrode voltage-clamp and outside-out patch-clamp techniques. Substitution of aspartate D444 to alanine or cysteine in the degenerin region of BASIC, a region known to be critically involved in channel gating, resulted in a substantial reduction of BASIC Ca2+ sensitivity. Moreover, mutating D444 or the neighboring alanine (A443) to cysteine significantly reduced the t-DCA-mediated BASIC stimulation. A combined molecular docking/simulation approach demonstrated that t-DCA may temporarily form hydrogen bonds with several amino acid residues including D444 in the outer vestibule of the BASIC pore or in the inter-subunit space. By these interactions, t-DCA may stabilize the open state of the channel. Indeed, single-channel recordings provided evidence that t-DCA activates BASIC by stabilizing the open state of the channel, whereas extracellular Ca2+ inhibits BASIC by stabilizing its closed state. In conclusion, our results highlight the potential role of the degenerin region as a critical regulatory site involved in the functional interaction of Ca2+ and t-DCA with BASIC.


Subject(s)
Bile Acids and Salts/metabolism , Calcium/metabolism , Degenerin Sodium Channels/metabolism , Amino Acid Sequence , Animals , Bile/metabolism , Humans , Ion Channel Gating/physiology , Molecular Docking Simulation/methods , Oocytes/metabolism , Xenopus laevis/metabolism
7.
Physiol Rep ; 5(3)2017 Feb.
Article in English | MEDLINE | ID: mdl-28193786

ABSTRACT

Acid-sensing ion channels (ASICs) are nonvoltage-gated sodium channels transiently activated by extracellular protons and belong to the epithelial sodium channel (ENaC)/Degenerin (DEG) family of ion channels. Bile acids have been shown to activate two members of this family, the bile acid-sensitive ion channel (BASIC) and ENaC. To investigate whether bile acids also modulate ASIC function, human ASIC1a was heterologously expressed in Xenopus laevis oocytes. Exposing oocytes to tauro-conjugated cholic (t-CA), deoxycholic (t-DCA), and chenodeoxycholic (t-CDCA) acid at pH 7.4 did not activate ASIC1a-mediated whole-cell currents. However, in ASIC1a expressing oocytes the whole-cell currents elicited by pH 5.5 were significantly increased in the presence of these bile acids. Single-channel recordings in outside-out patches confirmed that t-DCA enhanced the stimulatory effect of pH 5.5 on ASIC1a channel activity. Interestingly, t-DCA reduced single-channel current amplitude by ~15% which suggests an interaction of t-DCA with a region close to the channel pore. Molecular docking predicted binding of bile acids to the pore region near the degenerin site (G433) in the open conformation of the channel. Site-directed mutagenesis demonstrated that the amino acid residue G433 is critically involved in the potentiating effect of bile acids on ASIC1a activation by protons.


Subject(s)
Acid Sensing Ion Channels/physiology , Bile Acids and Salts/physiology , Acid Sensing Ion Channels/metabolism , Animals , Bile Acids and Salts/metabolism , Bile Acids and Salts/pharmacology , Binding Sites , Humans , Hydrogen-Ion Concentration , Molecular Docking Simulation , Oocytes , Protons , Xenopus laevis
8.
J Biol Chem ; 291(38): 19835-47, 2016 09 16.
Article in English | MEDLINE | ID: mdl-27489102

ABSTRACT

The epithelial sodium channel (ENaC) is a member of the ENaC/degenerin ion channel family, which also includes the bile acid-sensitive ion channel (BASIC). So far little is known about the effects of bile acids on ENaC function. ENaC is probably a heterotrimer consisting of three well characterized subunits (αßγ). In humans, but not in mice and rats, an additional δ-subunit exists. The aim of this study was to investigate the effects of chenodeoxycholic, cholic, and deoxycholic acid in unconjugated (CDCA, CA, and DCA) and tauro-conjugated (t-CDCA, t-CA, t-DCA) form on human ENaC in its αßγ- and δßγ-configuration. We demonstrated that tauro-conjugated bile acids significantly stimulate ENaC in the αßγ- and in the δßγ-configuration. In contrast, non-conjugated bile acids have a robust stimulatory effect only on δßγENaC. Bile acids stimulate ENaC-mediated currents by increasing the open probability of active channels without recruiting additional near-silent channels known to be activated by proteases. Stimulation of ENaC activity by bile acids is accompanied by a significant reduction of the single-channel current amplitude, indicating an interaction of bile acids with a region close to the channel pore. Analysis of the known ASIC1 (acid-sensing ion channel) crystal structure suggested that bile acids may bind to the pore region at the degenerin site of ENaC. Substitution of a single amino acid residue within the degenerin region of ßENaC (N521C or N521A) significantly reduced the stimulatory effect of bile acids on ENaC, suggesting that this site is critical for the functional interaction of bile acids with the channel.


Subject(s)
Bile Acids and Salts/chemistry , Bile Acids and Salts/metabolism , Epithelial Sodium Channels/chemistry , Epithelial Sodium Channels/metabolism , Acid Sensing Ion Channels/chemistry , Acid Sensing Ion Channels/genetics , Acid Sensing Ion Channels/metabolism , Animals , Crystallography, X-Ray , Epithelial Sodium Channels/genetics , Humans , Mice , Protein Domains , Rats , Xenopus laevis
9.
Pflugers Arch ; 467(4): 687-701, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24906497

ABSTRACT

Proteolytic activation of protease-activated receptor 2 (PAR2) may represent a major mechanism of regulating the transient receptor potential vanilloid 4 (TRPV4) non-selective cation channel in pathophysiological conditions associated with protease activation (e.g. during inflammation). To provide electrophysiological evidence for PAR2-mediated TRPV4 regulation, we characterised the properties of human TRPV4 heterologously expressed in Xenopus laevis oocytes in the presence and absence of co-expressed human PAR2. In outside-out patches from TRPV4 expressing oocytes, we detected single-channel activity typical for TRPV4 with a single-channel conductance of about 100 pS for outward and 55 pS for inward currents. The synthetic TRPV4 activator GSK1016790A stimulated TRPV4 mainly by converting previously silent channels into active channels with an open probability of nearly one. In oocytes co-expressing TRPV4 and PAR2, PAR2 activation by trypsin or by specific PAR2 agonist SLIGRL-NH2 potentiated the GSK1016790A-stimulated TRPV4 whole-cell currents several fold, indicative of channel sensitisation. Pre-incubation of oocytes with the calcium chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA)-AM did not reduce the stimulatory effect of PAR2 activation on TRPV4, which indicates that the effect is independent of intracellular calcium signalling. Neutrophil elastase, a biased agonist of PAR2 that does not induce intracellular calcium signalling, also caused a PAR2-dependent sensitisation of TRPV4. The Rho-kinase inhibitor Y27362 abolished elastase-stimulated sensitisation of TRPV4, which indicates that Rho-kinase signalling plays a critical role in PAR2-mediated TRPV4 sensitisation by the biased agonist neutrophil elastase. During acute inflammation, neutrophil elastase may sensitise TRPV4 by a mechanism involving biased agonism of PAR2 and activation of Rho-kinase.


Subject(s)
Calcium Signaling , Leukocyte Elastase/metabolism , Receptor, PAR-2/metabolism , TRPV Cation Channels/metabolism , Animals , Humans , Leucine/analogs & derivatives , Leucine/pharmacology , Oligopeptides/pharmacology , Proteolysis , Receptor, PAR-2/agonists , Sulfonamides/pharmacology , TRPV Cation Channels/agonists , Trypsin/pharmacology , Xenopus , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
10.
Pflugers Arch ; 466(2): 253-63, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23842738

ABSTRACT

The human bile acid-sensitive ion channel (hBASIC) is a cation channel of the degenerin/epithelial Na(+) channel gene family that is expressed in the intestinal tract and can be activated by bile acids. Here, we show that in addition to its sensitivity for bile acids, hBASIC shares further key features with its rat ortholog: it is blocked by extracellular divalent cations, is inhibited by micromolar concentrations of the diarylamidine diminazene, and activated by millimolar concentrations of flufenamic acid. Furthermore, we demonstrate that two major bile acids present in human bile, chenodeoxycholic acid and deoxycholic acid, activate hBASIC in a synergistic manner. In addition, we determined the single-channel properties of hBASIC in outside-out patch clamp recordings, revealing a single-channel conductance of about 11 pS and a high Na(+) selectivity. Deoxycholic acid activates hBASIC in patch clamp recordings mainly by reducing the single-channel closed time. In summary, we provide a thorough functional characterization of hBASIC.


Subject(s)
Acid Sensing Ion Channels/physiology , Bile Acids and Salts/pharmacology , Degenerin Sodium Channels/physiology , Acid Sensing Ion Channels/drug effects , Cations, Divalent/pharmacology , Degenerin Sodium Channels/drug effects , Diminazene/pharmacology , Epithelial Sodium Channels/drug effects , Epithelial Sodium Channels/physiology , Flufenamic Acid/pharmacology , Humans , Ion Channel Gating/physiology , Patch-Clamp Techniques
11.
Biochim Biophys Acta ; 1828(11): 2399-409, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23791703

ABSTRACT

Several Cl(-) channels have been described in the native renal tubule, but their correspondence with ClC-K1 and ClC-K2 channels (orthologs of human ClC-Ka and ClC-Kb), which play a major role in transcellular Cl(-) absorption in the kidney, has yet to be established. This is partly because investigation of heterologous expression has involved rat or human ClC-K models, whereas characterization of the native renal tubule has been done in mice. Here, we investigate the electrophysiological properties of mouse ClC-K1 channels heterologously expressed in Xenopus laevis oocytes and in HEK293 cells with or without their accessory Barttin subunit. Current amplitudes and plasma membrane insertion of mouse ClC-K1 were enhanced by Barttin. External basic pH or elevated calcium stimulated currents followed the anion permeability sequence Cl(-)>Br(-)>NO3(-)>I(-). Single-channel recordings revealed a unit conductance of ~40pS. Channel activity in cell-attached patches increased with membrane depolarization (voltage for half-maximal activation: ~-65mV). Insertion of the V166E mutation, which introduces a glutamate in mouse ClC-K1, which is crucial for channel gating, reduced the unit conductance to ~20pS. This mutation shifted the depolarizing voltage for half-maximal channel activation to ~+25mV. The unit conductance and voltage dependence of wild-type and V166E ClC-K1 were not affected by Barttin. Owing to their strikingly similar properties, we propose that the ClC-K1/Barttin complex is the molecular substrate of a chloride channel previously detected in the mouse thick ascending limb (Paulais et al., J Membr. Biol, 1990, 113:253-260).


Subject(s)
Chloride Channels/metabolism , Animals , HEK293 Cells , Humans , Kidney Tubules/metabolism , Patch-Clamp Techniques , Recombinant Proteins/metabolism , Xenopus laevis
12.
Am J Physiol Lung Cell Mol Physiol ; 304(1): L43-55, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23087020

ABSTRACT

In some patients with atypical cystic fibrosis (CF), only one allele of the CF transmembrane conductance regulator (CFTR) gene is affected. Mutations of the epithelial sodium channel (ENaC) may contribute to the pathophysiology of the disease in these patients. To functionally characterize a mutation in the ß-subunit of ENaC (ßV348M) recently identified in a patient with severe CF-like symptoms (Mutesa et al. 2009), we expressed wild-type (wt) αßγENaC or mutant αßV348MγENaC in Xenopus laevis oocytes. The ßV348M mutation stimulated amiloride-sensitive whole-cell current (ΔI(ami)) by ∼40% but had no effect on surface expression or single-channel conductance of ENaC. Instead the mutation increased channel open probability (P(o)). Proteolytic activation of mutant ENaC by chymotrypsin was reduced compared with that of wt ENaC (∼3.0-fold vs. ∼4.2-fold), which is consistent with the increased baseline P(o) of mutant ENaC. Similarly, the ENaC activator S3969 stimulated mutant ENaC currents to a lesser degree (by ∼2.6-fold) than wt ENaC currents (by ∼3.5-fold). The gain-of-function effect of the ßV348M mutation was confirmed by whole-cell current measurements in HEK293 cells transiently transfected with wt or mutant ENaC. Computational channel modeling in combination with functional expression of different ßV348 mutants in oocytes suggests that the ßV348M mutation increases channel P(o) by destabilizing the closed channel state. Our findings indicate that the gain-of-function effect of the ßV348M mutation may contribute to CF pathophysiology by inappropriately increasing sodium and fluid absorption in the respiratory tract.


Subject(s)
Epithelial Sodium Channels/genetics , Amino Acid Substitution , Animals , Cystic Fibrosis/genetics , Epithelial Sodium Channels/metabolism , Female , HEK293 Cells , Humans , Indoles/pharmacology , Mutation , Patch-Clamp Techniques , Xenopus laevis
13.
J Gen Physiol ; 140(4): 375-89, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22966015

ABSTRACT

Proteolytic activation of the epithelial sodium channel (ENaC) involves cleavage of its γ subunit in a critical region targeted by several proteases. Our aim was to identify cleavage sites in this region that are functionally important for activation of human ENaC by plasmin and chymotrypsin. Sequence alignment revealed a putative plasmin cleavage site in human γENaC (K189) that corresponds to a plasmin cleavage site (K194) in mouse γENaC. We mutated this site to alanine (K189A) and expressed human wild-type (wt) αßγENaC and αßγ(K189A)ENaC in Xenopus laevis oocytes. The γ(K189A) mutation reduced but did not abolish activation of ENaC whole cell currents by plasmin. Mutating a putative prostasin site (γ(RKRK178AAAA)) had no effect on the stimulatory response to plasmin. In contrast, a double mutation (γ(RKRK178AAAA;K189A)) prevented the stimulatory effect of plasmin. We conclude that in addition to the preferential plasmin cleavage site K189, the putative prostasin cleavage site RKRK178 may serve as an alternative site for proteolytic channel activation by plasmin. Interestingly, the double mutation delayed but did not abolish ENaC activation by chymotrypsin. The time-dependent appearance of cleavage products at the cell surface nicely correlated with the stimulatory effect of chymotrypsin on ENaC currents in oocytes expressing wt or double mutant ENaC. Delayed proteolytic activation of the double mutant channel with a stepwise recruitment of so-called near-silent channels was confirmed in single-channel recordings from outside-out patches. Mutating two phenylalanines (FF174) in the vicinity of the prostasin cleavage site prevented proteolytic activation by chymotrypsin. This indicates that chymotrypsin preferentially cleaves at FF174. The close proximity of FF174 to the prostasin site may explain why mutating the prostasin site impedes channel activation by chymotrypsin. In conclusion, this study supports the concept that different proteases have distinct preferences for certain cleavage sites in γENaC, which may be relevant for tissue-specific proteolytic ENaC activation.


Subject(s)
Chymotrypsin/metabolism , Epithelial Sodium Channels/chemistry , Epithelial Sodium Channels/metabolism , Fibrinolysin/metabolism , Ion Channel Gating , Amino Acid Motifs , Animals , Epithelial Sodium Channels/genetics , Humans , Mutation, Missense , Protein Subunits/metabolism , Proteolysis , Serine Endopeptidases/pharmacology , Xenopus
14.
J Biol Chem ; 286(37): 31944-52, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21775436

ABSTRACT

The epithelial sodium channel (ENaC) is a member of the ENaC/degenerin superfamily. ENaC is a heteromultimer containing three homologous subunits (α, ß, and γ); however, the subunit stoichiometry is still controversial. Here, we addressed this issue using atomic force microscopy imaging of complexes between isolated ENaC and antibodies/Fab fragments directed against specific epitope tags on the α-, ß- and γ-subunits. We show that for α-, ß- and γ-ENaC alone, pairs of antibodies decorate the channel at an angle of 120°, indicating that the individual subunits assemble as homotrimers. A similar approach demonstrates that αßγ-ENaC assembles as a heterotrimer containing one copy of each subunit. Intriguingly, all four subunit combinations also produce higher-order structures containing two or three individual trimers. The trimer-of-trimers organization would account for earlier reports that ENaC contains eight to nine subunits.


Subject(s)
Cell Membrane/chemistry , Cell Membrane/ultrastructure , Epithelial Sodium Channels/chemistry , Microscopy, Atomic Force , Protein Subunits/chemistry , Animals , Cell Membrane/metabolism , Epithelial Sodium Channels/metabolism , HEK293 Cells , Humans , Immunoglobulin Fab Fragments/chemistry , Protein Structure, Quaternary , Protein Subunits/metabolism , Xenopus laevis
15.
J Physiol ; 588(Pt 8): 1211-25, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20194130

ABSTRACT

Increased activity of the epithelial sodium channel (ENaC) in the respiratory airways contributes to the pathophysiology of cystic fibrosis (CF), a genetic disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. In some patients suffering from atypical CF a mutation can be identified in only one CFTR allele. We recently identified in this group of CF patients a heterozygous mutation (W493R) in the alpha-subunit of ENaC. Here, we investigate the functional effects of this mutation by expressing wild-type alpha beta gamma ENaC or mutant alpha(W493R)beta gamma ENaC in Xenopus oocytes. The alpha W493R mutation stimulated amiloride-sensitive whole-cell currents (Delta I(ami)) by approximately 4-fold without altering the single-channel conductance or surface expression of ENaC. As these data suggest that the open probability (P(o)) of the mutant channel is increased, we investigated the proteolytic activation of ENaC by chymotrypsin. Single-channel recordings revealed that chymotrypsin activated near-silent channels in outside-out membrane patches from oocytes expressing wild-type ENaC, but not in membrane patches from oocytes expressing the mutant channel. In addition, the alpha W493R mutation abolished Na(+) self inhibition of ENaC, which might also contribute to its gain-of-function effects. We conclude that the alpha W493R mutation promotes constitutive activation of ENaC by reducing the inhibitory effect of extracellular Na(+) and decreasing the pool of near-silent channels. The resulting gain-of-function phenotype of the mutant channel might contribute to the pathophysiology of CF in patients carrying this mutation.


Subject(s)
Cystic Fibrosis/physiopathology , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/physiology , Mutation/genetics , Sodium/metabolism , Animals , Cells, Cultured , Chymotrypsin/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Feedback, Physiological/physiology , Female , Humans , Oocytes/cytology , Oocytes/metabolism , Patch-Clamp Techniques , Phenotype , Plasmids , Xenopus laevis
16.
Cell Physiol Biochem ; 25(1): 145-58, 2010.
Article in English | MEDLINE | ID: mdl-20054153

ABSTRACT

Loss-of-function mutations of the epithelial sodium channel (ENaC) may contribute to pulmonary symptoms resembling those of patients with atypical cystic fibrosis (CF). Recently, we identified a loss-of-function mutation in the alpha-subunit of ENaC (alphaF61L) in an atypical CF patient without mutations in CFTR. To investigate the functional effect of this mutation, we expressed human wild-type alpha beta gamma-ENaC or mutant alpha(F61L) beta gamma-ENaC in Xenopus laevis oocytes. The alphaF61L mutation reduced the ENaC mediated whole-cell currents by approximately 90%. In contrast, the mutation decreased channel surface expression only by approximately 40% and did not alter the single-channel conductance. These findings indicate that the major effect of the mutation is a reduction of the average channel open probability (P(o)). This was confirmed by experiments using the betaS520C mutant ENaC which can be converted to a channel with a P(o) of nearly one, and by experiments using chymotrypsin to proteolytically activate the channel. These experiments revealed that the mutation reduced the average P(o) of ENaC by approximately 75%. Na(+) self inhibition of the mutant channel was significantly enhanced, but the observed effect was too small to account for the large reduction in average channel P(o). The ENaC-activator S3969 partially rescued the loss-of-function phenotype of the alphaF61L mutation. We conclude that the alphaF61L mutation may contribute to respiratory symptoms in atypical CF patients.


Subject(s)
Cystic Fibrosis/genetics , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/metabolism , Mutation , Animals , Chymotrypsin/metabolism , Cystic Fibrosis/metabolism , Epithelial Sodium Channels/analysis , Female , Gene Expression , Humans , Oocytes/metabolism , Sodium/metabolism , Xenopus laevis
17.
Cell Physiol Biochem ; 26(6): 913-24, 2010.
Article in English | MEDLINE | ID: mdl-21220922

ABSTRACT

Kinases contribute to the regulation of the epithelial sodium channel (ENaC) in a complex manner. For example, SGK1 (serum- and glucocorticoid-inducible kinase type 1) enhances ENaC surface expression by phosphorylating Nedd4-2, thereby preventing ENaC retrieval and degradation. An additional mechanism of ENaC activation by SGK1 involves an SGK consensus motif ((616)RSRYWS(621)) in the C-terminus of the channel's α-subunit. This consensus motif may also be a target for ENaC regulation by protein kinase B α (PKBα) known to be activated by insulin and growth factors. Therefore, we investigated a possible role of PKBα in the regulation of rat ENaC heterologously expressed in Xenopus laevis oocytes. We found that recombinant PKBα included in the pipette solution increased ENaC currents in outside-out patches by about 4-fold within 15-20 min. Replacing the serine residue S621 of the SGK consensus motif by an alanine (S621A) abolished this stimulatory effect. In co-expression experiments active PKBα but not catalytically inactive PKBα significantly increased ENaC whole-cell currents and surface expression by more than 50 % within 24 hours of co-expression. Interestingly, this stimulatory effect was preserved in oocytes expressing ENaC with the S621A mutation. We conclude that the acute stimulatory effect of PKBα involves a specific kinase consensus motif in the C-terminus of the channel's α-subunit. In contrast, the increase in channel surface expression caused by co-expression of PKBα does not depend on this site in the channel and is probably mediated by an effect on channel trafficking.


Subject(s)
Epithelial Sodium Channels/metabolism , Oocytes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Amino Acid Substitution , Animals , Epithelial Sodium Channels/physiology , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Mutagenesis, Site-Directed , Oocytes/physiology , Patch-Clamp Techniques , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/pharmacology , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Xenopus laevis/embryology , Xenopus laevis/metabolism
18.
Cell Physiol Biochem ; 24(5-6): 605-18, 2009.
Article in English | MEDLINE | ID: mdl-19910701

ABSTRACT

The lipid environment of the epithelial sodium channel (ENaC) and its possible association with so-called lipid rafts may be relevant to its function. The aim of our study was to confirm the association of ENaC with lipid rafts and to analyze the effect of cholesterol depletion of the plasma membrane by methyl-beta-cyclodextrin (MbetaCD) on channel function and regulation. Using sucrose density gradient centrifugation we demonstrated that a significant portion of ENaC protein distributes to low density fractions thought to be typical lipid raft fractions. Importantly, cholesterol depletion of cell lysate by MbetaCD shifted ENaC to non-raft fractions of higher density. Live cell imaging demonstrated that treatment with MbetaCD largely reduced filipin staining over time, confirming cholesterol depletion of the plasma membrane. For electrophysiological studies intact oocytes were exposed to 20 mM MbetaCD for three hours. MbetaCD treatment had no consistent effect on baseline whole-cell ENaC currents. In addition to the typical single channel conductance of about 5 pS, subconductance states of ENaC were occasionally observed in patches from MbetaCD treated but not from control oocytes. Importantly, in outside-out patch clamp recordings the stimulatory effect of recombinant SGK1 in the pipette solution was essentially abolished in oocytes pretreated with MbetaCD. These results indicate that ENaC activation by cytosolic SGK1 is compromised by removing cholesterol from the plasma membrane. Thus, ENaC activation by SGK1 may require the presence of an intact lipid environment and/or of lipid rafts as signalling platform.


Subject(s)
Cell Membrane/metabolism , Cholesterol/metabolism , Epithelial Sodium Channels/metabolism , Immediate-Early Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Centrifugation, Density Gradient , Electrophysiological Phenomena , Membrane Microdomains , Oocytes/metabolism , Xenopus laevis , beta-Cyclodextrins/pharmacology
19.
J Physiol ; 586(19): 4587-608, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18669538

ABSTRACT

The mechanisms by which proteases activate the epithelial sodium channel (ENaC) are not yet fully understood. We investigated the effect of extracellular proteases on rat ENaC heterologously expressed in Xenopus laevis oocytes. Application of trypsin increased ENaC whole-oocyte currents by about 8-fold without a concomitant increase in channel surface expression. The stimulatory effect of trypsin was preserved in oocytes expressing alphagamma-ENaC, but was abolished in oocytes expressing alphabeta-ENaC. Thus, the gamma-subunit appears to be essential for channel activation by extracellular proteases. Site-directed mutagenesis of a putative prostasin cleavage site in the extracellular loop of the gamma-subunit revealed that mutating the 181Lys residue to alanine (gammaK181A) increases ENaC baseline whole-oocyte currents, decreases channel surface expression, and largely reduces the stimulatory effect of extracellular proteases (trypsin, chymotrypsin and human neutrophil elastase). In single-channel recordings from outside-out patches we demonstrated that the gammaK181A mutation essentially abolishes the activation of near-silent channels by trypsin, while a stimulatory effect of trypsin on channel gating is preserved. This apparent dual effect of trypsin on channel gating and on the recruitment of near-silent channels was confirmed by experiments using the beta518C mutant ENaC which can be converted to a channel with an open probability of nearly one by exposure to a sulfhydryl reagent. Interestingly, the gammaK181A mutation results in the spontaneous appearance of a 67 kDa fragment of the gamma-subunit in the plasma membrane which can be prevented by a furin inhibitor and also occurs after channel activation by extracellular trypsin. This suggests that the mutation promotes channel cleavage and activation by endogenous proteases. This would lower the pool of near-silent channels and explain the constitutive activation and reduced responsiveness of the mutant channel to extracellular proteases. We conclude that the mutated site (K181A) affects a region in the gamma-subunit of ENaC that is functionally important for the activation of near-silent channels by extracellular proteases.


Subject(s)
Epithelial Sodium Channels/metabolism , Ion Channel Gating , Protein Subunits/metabolism , Trypsin/metabolism , Animals , Cell Membrane/enzymology , Chymotrypsin/metabolism , Epithelial Sodium Channels/genetics , Furin/metabolism , Leukocyte Elastase/metabolism , Mutagenesis, Site-Directed , Mutation , Oocytes , Patch-Clamp Techniques , Phenotype , Rats , Serine Endopeptidases/metabolism , Xenopus laevis
20.
J Biol Chem ; 279(37): 38134-42, 2004 Sep 10.
Article in English | MEDLINE | ID: mdl-15234985

ABSTRACT

Aldosterone-induced serum- and glucocorticoid-inducible kinase isoform 1 (SGK1) contributes to the regulation of the epithelial sodium channel (ENaC), the activity of which is critical for long term blood pressure control. Aldosterone-induced SGK1 is thought to enhance ENaC surface expression by phosphorylating Nedd4-2 and thereby preventing ENaC retrieval and degradation. In outside-out membrane patches of Xenopus laevis oocytes heterologously expressing ENaC, amiloride-sensitive ENaC currents were enhanced by phosphatase inhibitors and were dependent on cytosolic Mg(2+). This indicates that a kinase is involved in channel regulation. Indeed, recombinant constitutively active SGK1, included in the pipette solution, caused a sustained 2- to 3-fold increase of ENaC currents. Deletion of the C terminus of alphaENaC largely reduced the stimulatory effect of SGK1, whereas stimulation by SGK1 did not require the presence of the C termini of the beta- or gamma-subunits. Replacing the serine residue Ser(621) of the SGK1 consensus motif in the C terminus of the alpha-subunit by an alanine specifically abolished the stimulatory effect of SGK. Our findings indicate that SGK1 can stimulate ENaC activity independently of an inhibition of Nedd4-2-mediated channel retrieval. This defines a novel regulatory pathway likely to be relevant for aldosterone-induced stimulation of ENaC in vivo.


Subject(s)
Nuclear Proteins , Protein Serine-Threonine Kinases/metabolism , Sodium Channels/metabolism , Alanine/chemistry , Amiloride/pharmacology , Amino Acid Motifs , Animals , Cytosol/metabolism , DNA, Complementary/metabolism , Enzyme Activation , Epithelial Sodium Channels , Gene Deletion , Immediate-Early Proteins , Magnesium/chemistry , Models, Biological , Mutagenesis, Site-Directed , Oocytes/metabolism , Patch-Clamp Techniques , Phosphorylation , Protein Isoforms , Protein Structure, Tertiary , RNA, Complementary/metabolism , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Serine/chemistry , Time Factors , Xenopus , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...