Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Physiol Renal Physiol ; 302(1): F1-8, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21993886

ABSTRACT

The epithelial sodium channel (ENaC) is activated by a unique mechanism, whereby inhibitory tracts are released by proteolytic cleavage within the extracellular loops of two of its three homologous subunits. While cleavage by furin within the biosynthetic pathway releases one inhibitory tract from the α-subunit and moderately activates the channel, full activation through release of a second inhibitory tract from the γ-subunit requires cleavage once by furin and then at a distal site by a second protease, such as prostasin, plasmin, or elastase. We now report that coexpression of mouse transmembrane protease serine 4 (TMPRSS4) with mouse ENaC in Xenopus oocytes was associated with a two- to threefold increase in channel activity and production of a unique ∼70-kDa carboxyl-terminal fragment of the γ-subunit, similar to the ∼70-kDa γ-subunit fragment that we previously observed with prostasin-dependent channel activation. TMPRSS4-dependent channel activation and production of the ∼70-kDa fragment were partially blocked by mutation of the prostasin-dependent cleavage site (γRKRK186QQQQ). Complete inhibition of TMPRSS4-dependent activation of ENaC and γ-subunit cleavage was observed when three basic residues between the furin and prostasin cleavage sites were mutated (γK173Q, γK175Q, and γR177Q), in addition to γRKRK186QQQQ. Mutation of the four basic residues associated with the furin cleavage site (γRKRR143QQQQ) also prevented TMPRSS4-dependent channel activation. We conclude that TMPRSS4 primarily activates ENaC by cleaving basic residues within the tract γK173-K186 distal to the furin cleavage site, thereby releasing a previously defined key inhibitory tract encompassing γR158-F168 from the γ-subunit.


Subject(s)
Epithelial Sodium Channels/physiology , Membrane Proteins/physiology , Serine Endopeptidases/physiology , Amino Acid Sequence , Animals , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/metabolism , Furin/metabolism , Mice , Oocytes/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , Serine Endopeptidases/metabolism , Xenopus laevis
2.
J Biol Chem ; 286(31): 27436-46, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21659509

ABSTRACT

Epithelial Na(+) channels (ENaCs) play an essential role in the regulation of body fluid homeostasis. Certain transition metals activate or inhibit the activity of ENaCs. In this study, we examined the effect of extracellular Cu(2+) on human ENaC expressed in Xenopus oocytes and investigated the structural basis for its effects. External Cu(2+) inhibited human αßγ ENaC with an estimated IC(50) of 0.3 µM. The slow time course and a lack of change in the current-voltage relationship were consistent with an allosteric (non pore-plugging) inhibition of human ENaC by Cu(2+). Experiments with mixed human and mouse ENaC subunits suggested that both the α and ß subunits were primarily responsible for the inhibitory effect of Cu(2+) on human ENaC. Lowering bath solution pH diminished the inhibition by Cu(2+). Mutations of two α, two ß, and two γ His residues within extracellular domains significantly reduced the inhibition of human ENaC by Cu(2+). We identified a pair of residues as potential Cu(2+)-binding sites at the subunit interface between thumb subdomain of αhENaC and palm subdomain of ßhENaC, suggesting a counterclockwise arrangement of α, ß, and γ ENaC subunits in a trimeric channel complex when viewed from above. We conclude that extracellular Cu(2+) is a potent inhibitor of human ENaC and binds to multiple sites within the extracellular domains including a subunit interface.


Subject(s)
Copper/pharmacology , Epithelial Sodium Channel Blockers , Animals , Cells, Cultured , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/metabolism , Humans , Mutagenesis, Site-Directed , Protein Binding , Xenopus
4.
Am J Physiol Renal Physiol ; 299(4): F854-61, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20630937

ABSTRACT

Proteases activate the epithelial sodium channel (ENaC) by cleaving the large extracellular domains of the α- and γ-subunits and releasing peptides with inhibitory properties. Furin and prostasin activate mouse ENaC by cleaving the γ-subunit at sites flanking a 43 residue inhibitory tract (γE144-K186). To determine whether there is a minimal inhibitory region within this 43 residue tract, we generated serial deletions in the inhibitory tract of the γ-subunit in channels resistant to cleavage by furin and prostasin. We found that partial or complete deletion of a short segment in the γ-subunit, R158-N171, enhanced channel activity. Synthetic peptides overlapping this segment in the γ-subunit further identified a key 11-mer tract, R158-F168 (RFLNLIPLLVF), which inhibited wild-type ENaC expressed in Xenopus laevis oocytes, and endogenous channels in mpkCCD cells and human airway epithelia. Further studies with amino acid-substituted peptides defined residues that are required for inhibition in this key 11-mer tract. The presence of the native γ inhibitory tract in ENaC weakened the intrinsic binding constant of the 11-mer peptide inhibitor, suggesting that the γ inhibitory tract and the 11-mer peptide interact at overlapping sites within the channel.


Subject(s)
Epithelial Sodium Channels/analysis , Protein Structure, Tertiary , Animals , Cell Line , Cells, Cultured , Epithelial Sodium Channels/drug effects , Epithelial Sodium Channels/metabolism , Female , Furin/pharmacology , Humans , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/metabolism , Mice , Oocytes/cytology , Oocytes/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , Serine Endopeptidases/pharmacology , Xenopus laevis
5.
Curr Opin Nephrol Hypertens ; 19(1): 13-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19864949

ABSTRACT

PURPOSE OF REVIEW: Hypertension and edema are clinical manifestations of the extracellular volume expansion generated by abnormal renal sodium handling. Perturbations in epithelial sodium channel (ENaC) activity disrupt volume homeostasis. ENaC activity can be enhanced by proteases that cleave its long extracellular domains. Recent evidence suggests that this mechanism may be involved in individuals with volume overload and proteinuria. RECENT FINDINGS: Several observations indicate a link between proteinuria and hypertension, with proteinuria preceding and predicting the onset of incident hypertension in some individuals. Recently, enhanced cleavage of ENaC's extracellular loops was identified in kidney tissue of proteinuric mice. Plasmin, a serine protease known for its role in fibrinolysis, has been implicated as an activator of ENaC in proteinuric states as nephrotic urine activates ENaC expressed in a mouse collecting duct cell line, aprotinin-affinity precipitation of nephrotic urine abolishes its ability to activate ENaC, plasmin is a major component within aprotinin-affinity purified nephrotic urine and is absent in nonproteinuric urine, and plasmin activates ENaC by cleaving the extracellular loop of its gamma subunit. SUMMARY: Enhancement of ENaC activity by proteases represents a likely mechanism for extracellular volume overload relevant to some individuals with proteinuria. Proteases not normally found in the urine can enter the urinary space across damaged glomeruli and activate ENaC. Further understanding of this mechanism may guide targeted therapeutics in individuals with proteinuria, edema, and hypertension.


Subject(s)
Fibrinolysin/metabolism , Sodium/metabolism , Animals , Edema/metabolism , Epithelial Sodium Channels/chemistry , Epithelial Sodium Channels/metabolism , Homeostasis , Humans , Hypertension/metabolism , Kidney Tubules/metabolism , Mice , Models, Biological , Models, Molecular , Proteinuria/metabolism
6.
J Biol Chem ; 284(52): 36473-36481, 2009 Dec 25.
Article in English | MEDLINE | ID: mdl-19858190

ABSTRACT

Acid-sensing ion channels are proton-gated Na(+) channels expressed predominantly in neurons. How channel structure translates an environmental stimulus into changes in pore permeability remains largely undefined. The pore of ASIC1 is defined by residues in the second transmembrane domain (TM2), although a segment of the outer vestibule is formed by residues of TM1. We used the voltage clamp fluorometry technique to define the role of the region preceding TM2 (pre-TM2) in activation and desensitization of mouse ASIC1a. Oocytes expressing E425C channels labeled with Alexa Fluor 488 C5-maleimide showed a change in the emission of the fluorescent probe in response to extracellular acidification. The time course of the change in fluorescence correlated with activation but not desensitization of E425C channels. The fluorescence emission did not change following extracellular acidification in oocytes carrying an inactivating mutation (W287G/E425C), although these channels were labeled and expressed at the plasma membrane. Our data indicate that pore opening occurs in conjunction with a conformational rearrangement of the pre-TM2. We observed a change in the emission of the fluorescent probe when labeled E425C channels transition from the desensitized to the resting state. The substituted-cysteine-accessibility method was used to determine whether the pre-TM2 has different conformations in the resting and desensitized states. State-dependent changes in accessibility to 2-[(trimethylammonium)ethyl]methanethiosulfonate bromide modification were observed in oocytes expressing K421C, K422C, Y424C, and E425C channels. Our results suggest that the pre-TM2 of ASIC1a undergoes dynamic conformational rearrangements during proton-dependent gating.


Subject(s)
Ion Channel Gating/physiology , Nerve Tissue Proteins/metabolism , Protons , Sodium Channels/metabolism , Acid Sensing Ion Channels , Amino Acid Substitution , Animals , Fluorometry/methods , Gene Expression , Maleimides/chemistry , Mice , Mutation, Missense , Nerve Tissue Proteins/genetics , Oocytes/cytology , Oocytes/metabolism , Protein Structure, Tertiary/physiology , Sodium Channels/genetics , Xenopus laevis
7.
J Biol Chem ; 283(52): 36586-91, 2008 Dec 26.
Article in English | MEDLINE | ID: mdl-18981180

ABSTRACT

Proteolytic processing of epithelial sodium channel (ENaC) subunits occurs as channels mature within the biosynthetic pathway. The proteolytic processing events of the alpha and gamma subunits are associated with channel activation. Furin cleaves the alpha subunit ectodomain at two sites, releasing an inhibitory tract and activating the channel. However, furin cleaves the gamma subunit ectodomain only once. A second distal cleavage in the gamma subunit induced by other proteases, such as prostasin and elastase, is required to release a second inhibitory tract and further activate the channel. We found that the serine protease plasmin activates ENaC in association with inducing cleavage of the gamma subunit at gammaLys194, a site distal to the furin site. A gammaK194A mutant prevented both plasmin-dependent activation of ENaC and plasmin-dependent production of a unique 70-kDa carboxyl-terminal gamma subunit cleavage fragment. Plasmin-dependent cleavage and activation of ENaC may have a role in extracellular volume expansion in human disorders associated with proteinuria, as filtered plasminogen may be processed by urokinase, released from renal tubular epithelium, to generate active plasmin.


Subject(s)
Epithelial Sodium Channels/metabolism , Fibrinolysin/metabolism , Furin/metabolism , Animals , Binding Sites , Dogs , Humans , Male , Mice , Mutagenesis, Site-Directed , Oocytes/metabolism , Protein Structure, Tertiary , Proteinuria/metabolism , Rats , Xenopus
8.
Am J Physiol Renal Physiol ; 294(1): F47-52, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18032549

ABSTRACT

Epithelial sodium channels (ENaC) are processed by proteases as they transit the biosynthetic pathway. We recently observed that furin-dependent processing of the alpha-subunit of ENaC at two sites within its extracellular domain is required for channel activation due to release of a 26-residue inhibitory domain. While channels with alpha-subunits lacking the furin sites are not cleaved and have very low activity, channels lacking the furin consensus sites as well as the tract between these sites (alphaD206-R231) are active. We analyzed channels with a series of deletions in the tract alphaD206-R231 and lacking the alpha-subunit furin consensus sites in Xenopus laevis oocytes. We found an eight-residue tract that, when deleted, restored channel activity to the level found in oocytes expressing wild-type ENaC. A synthetic peptide, LPHPLQRL, representing the tract alphaL211-L218, inhibited wild-type ENaC expressed in oocytes with an IC(50) of 0.9 microM, and inhibited channels expressed in collecting duct cells and human primary airway epithelial cells with an IC(50)s of between approximately 50 and 100 microM. Analyses of peptides with deletions within this inhibitory tract indicate that eight residues is the minimal backbone length that is required for ENaC inhibition. Analyses of 8-mer peptides with conserved and nonconserved substitutions suggest that L(1), P(2), H(3), P(4), and L(8) are required for inhibitory activity. Our findings suggest that this eight-residue tract is a key conserved inhibitory domain that provides epithelial cells with a reserve of inactive channels that can be activated as required by proteases.


Subject(s)
Epithelial Sodium Channels/physiology , Kidney Tubules, Collecting/physiology , Protein Structure, Tertiary/physiology , Amino Acid Sequence , Animals , Cell Membrane/physiology , Cells, Cultured , Epithelial Sodium Channels/analysis , Epithelial Sodium Channels/genetics , Female , Furin/metabolism , Humans , Kidney Tubules, Collecting/cytology , Mice , Molecular Sequence Data , Oocytes/cytology , Oocytes/metabolism , Patch-Clamp Techniques , Protein Structure, Tertiary/genetics , Respiratory Mucosa/cytology , Respiratory Mucosa/physiology , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...