Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters











Publication year range
1.
Viruses ; 12(10)2020 10 09.
Article in English | MEDLINE | ID: mdl-33050303

ABSTRACT

The integrins function as the primary receptor molecules for the pathogenic infection of foot-and-mouth disease virus (FMDV) in vivo, while the acquisition of a high affinity for heparan sulfate (HS) of some FMDV variants could be privileged to facilitate viral infection and expanded cell tropism in vitro. Here, we noted that a BHK-adapted Cathay topotype derivative (O/HN/CHA/93tc) but not its genetically engineered virus (rHN), was able to infect HS-positive CHO-K1 cells and mutant pgsD-677 cells. There were one or three residue changes in the capsid proteins of O/HN/CHA/93tc and rHN, as compared with that of their tissue-originated isolate (O/HN/CHA/93wt). The phenotypic properties of a set of site-directed mutants of rHN revealed that E83K of VP1 surrounding the fivefold symmetry axis was necessary for the integrin-independent infection of O/HN/CHA/93tc. L80 in VP2 was essential for the occurrence of E83K in VP1 during the adaptation of O/HN/CHA/93wt to BHK-21 cells. L80M in VP2 and D138G in VP1 of rHN was deleterious, which could be compensated by K83R of VP1 for restoring an efficient infection of integrin-negative CHO cell lines. These might have important implications for understanding the molecular and evolutionary mechanisms of the recognition and binding of FMDV with alternative cellular receptors.


Subject(s)
Binding Sites/physiology , Capsid Proteins/genetics , Capsid Proteins/metabolism , Foot-and-Mouth Disease Virus/metabolism , Receptors, Virus/metabolism , Virus Attachment , Amino Acid Sequence , Amino Acid Substitution , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , Foot-and-Mouth Disease/virology , Foot-and-Mouth Disease Virus/genetics , Genome, Viral/genetics , Heparitin Sulfate/metabolism , Mice , Receptors, Virus/genetics , Virus Internalization
2.
J Virol ; 93(7)2019 04 01.
Article in English | MEDLINE | ID: mdl-30700601

ABSTRACT

The presence of sequence divergence through adaptive mutations in the major capsid protein VP1, and also in VP0 (VP4 and VP2) and VP3, of foot-and-mouth disease virus (FMDV) is relevant to a broad range of viral characteristics. To explore the potential role of isolate-specific residues in the VP0 and VP3 coding regions of PanAsia-1 strains in genetic and phenotypic properties of FMDV, a series of recombinant full-length genomic clones were constructed using Cathay topotype infectious cDNA as the original backbone. The deleterious and compensatory effects of individual amino acid substitutions at positions 4008 and 3060 and in several different domains of VP2 illustrated that the chain-based spatial interaction patterns of VP1, VP2, and VP3 (VP1-3), as well as between the internal VP4 and the three external capsid proteins of FMDV, might contribute to the assembly of eventually viable viruses. The Y2079H site-directed mutants dramatically induced a decrease in plaque size on BHK-21 cells and viral pathogenicity in suckling mice. Remarkably, the 2079H-encoding viruses displayed a moderate increase in acid sensitivity correlated with NH4Cl resistance compared to the Y2079-encoding viruses. Interestingly, none of all the 16 rescued viruses were able to infect heparan sulfate-expressing CHO-K1 cells. However, viral infection in BHK-21 cells was facilitated by utilizing non-integrin-dependent, heparin-sensitive receptor(s) and replacements of four uncharged amino acids at position 3174 in VP3 of FMDV had no apparent influence on heparin affinity. These results provide particular insights into the correlation of evolutionary biology with genetic diversity in adapting populations of FMDV.IMPORTANCE The sequence variation within the capsid proteins occurs frequently in the infection of susceptible tissue cultures, reflecting the high levels of genetic diversity of FMDV. A systematic study for the functional significance of isolate-specific residues in VP0 and VP3 of FMDV PanAsia-1 strains suggested that the interaction of amino acid side chains between the N terminus of VP4 and several potential domains of VP1-3 had cascading effects on the viability and developmental characteristics of progeny viruses. Y2079H in VP0 of the indicated FMDVs could affect plaque size and pathogenicity, as well as acid sensitivity correlated with NH4Cl resistance, whereas there was no inevitable correlation in viral plaque and acid-sensitive phenotypes. The high affinity of non-integrin-dependent FMDVs for heparin might be explained by the differences in structures of heparan sulfate proteoglycans on the surfaces of different cell lines. These results may contribute to our understanding of the distinct phenotypic properties of FMDV in vitro and in vivo.


Subject(s)
Amino Acid Substitution/genetics , Capsid Proteins/genetics , Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease/virology , Animals , CHO Cells , Cricetulus , Heparitin Sulfate/genetics , Mice , Open Reading Frames/genetics , Serogroup , Virion/genetics
3.
Am J Physiol Renal Physiol ; 316(3): F539-F549, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30539654

ABSTRACT

Although the role of urea in urine concentration is known, the effect of urea handling by the urea transporters (UTs), UT-A1 and UT-A3, on sodium balance remains elusive. Serum and urinary sodium concentration is similar between wild-type mice (WT) and UT-A3 null (UT-A3 KO) mice; however, mice lacking both UT-A1 and UT-A3 (UT-A1/A3 KO) have significantly lower serum sodium and higher urinary sodium. Protein expression of renal sodium transporters is unchanged among all three genotypes. WT, UT-A3 KO, and UT-A1/A3 KO acutely respond to hydrochlorothiazide and furosemide; however, UT-A1/A3 KO fail to show a diuretic or natriuretic response following amiloride administration, indicating that baseline epithelial Na+ channel (ENaC) activity is impaired. UT-A1/A3 KO have more ENaC at the apical membrane than WT mice, and single-channel analysis of ENaC in split-open inner medullary collecting duct (IMCD) isolated in saline shows that ENaC channel density and open probability is higher in UT-A1/A3 KO than WT. UT-A1/A3 KO excrete more urinary nitric oxide (NO), a paracrine inhibitor of ENaC, and inner medullary nitric oxide synthase 1 mRNA expression is ~40-fold higher than WT. Because endogenous NO is unstable, ENaC activity was reassessed in split-open IMCD with the NO donor PAPA NONOate [1-propanamine-3-(2-hydroxy-2-nitroso-1-propylhydrazine)], and ENaC activity was almost abolished in UT-A1/A3 KO. In summary, loss of both UT-A1 and UT-A3 (but not UT-A3 alone) causes elevated medullary NO production and salt wasting. NO inhibition of ENaC, despite elevated apical accumulation of ENaC in UT-A1/A3 KO IMCD, appears to be the main contributor to natriuresis in UT-A1/A3 KO mice.


Subject(s)
Epithelial Sodium Channels/metabolism , Kidney Medulla/metabolism , Membrane Transport Proteins/metabolism , Nitric Oxide/metabolism , Sodium/metabolism , Animals , Ion Transport/physiology , Kidney Concentrating Ability/physiology , Membrane Transport Proteins/genetics , Mice , Mice, Knockout , Urea Transporters
4.
J Biol Chem ; 293(5): 1666-1675, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29180450

ABSTRACT

It has been suggested that voltage-dependent anion channels (VDACs) control the release of superoxide from mitochondria. We have previously shown that reactive oxygen species (ROS) such as superoxide (O2̇̄) and hydrogen peroxide (H2O2) stimulate epithelial sodium channels (ENaCs) in sodium-transporting epithelial tissue, including cortical collecting duct (CCD) principal cells. Therefore, we hypothesized that VDACs could regulate ENaC by modulating cytosolic ROS levels. Herein, we find that VDAC3-knockout(KO) mice can maintain normal salt and water balance on low-salt and high-salt diets. However, on a high-salt diet for 2 weeks, VDAC3-KO mice had significantly higher systolic blood pressure than wildtype mice. Consistent with this observation, after a high-salt diet for 2 weeks, ENaC activity in VDAC3-KO mice was significantly higher than wildtype mice. EM analysis disclosed a significant morphological change of mitochondria in the CCD cells of VDAC3-KO mice compared with wildtype mice, which may have been caused by mitochondrial superoxide overload. Of note, compared with wildtype animals, ROS levels in VDAC3-KO animals fed a normal or high-salt diet were consistently and significantly increased in renal tubules. Both the ROS scavenger 1-oxyl-2,2,6,6-tetramethyl-4-hydroxypiperidine (TEMPOL) and the mitochondrial ROS scavenger (2-(2,2,6,6-tetramethylpiperidin-1-oxyl-4-ylamino)-2-oxoethyl)triphenylphosphonium chloride (mito-TEMPO) could reverse the effect of high-salt on ENaC activity and systolic blood pressure in the VDAC3-KO mice. Mito-TEMPO partially correct the morphological changes in VDAC3-KO mice. Our results suggest that knocking out mitochondrial VDAC3 increases ROS, alters renal sodium transport, and leads to hypertension.


Subject(s)
Epithelial Sodium Channels/metabolism , Hydrogen Peroxide/metabolism , Kidney/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/deficiency , Sodium/metabolism , Superoxides/metabolism , Voltage-Dependent Anion Channels/deficiency , Animals , Blood Pressure/drug effects , Blood Pressure/genetics , Cyclic N-Oxides/pharmacology , Epithelial Sodium Channels/genetics , Hypertension/genetics , Hypertension/metabolism , Hypertension/pathology , Ion Transport/drug effects , Ion Transport/genetics , Kidney/pathology , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Membrane Transport Proteins/metabolism , Organophosphorus Compounds/pharmacology , Piperidines/pharmacology , Spin Labels , Voltage-Dependent Anion Channels/metabolism
5.
Biochem J ; 473(19): 3237-52, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27422782

ABSTRACT

The thiazide-sensitive sodium chloride cotransporter (NCC) and the epithelial sodium channel (ENaC) are two of the most important determinants of salt balance and thus systemic blood pressure. Abnormalities in either result in profound changes in blood pressure. There is one segment of the nephron where these two sodium transporters are coexpressed, the second part of the distal convoluted tubule. This is a key part of the aldosterone-sensitive distal nephron, the final regulator of salt handling in the kidney. Aldosterone is the key hormonal regulator for both of these proteins. Despite these shared regulators and coexpression in a key nephron segment, associations between these proteins have not been investigated. After confirming apical localization of these proteins, we demonstrated the presence of functional transport proteins and native association by blue native PAGE. Extensive coimmunoprecipitation experiments demonstrated a consistent interaction of NCC with α- and γ-ENaC. Mammalian two-hybrid studies demonstrated direct binding of NCC to ENaC subunits. Fluorescence resonance energy transfer and immunogold EM studies confirmed that these transport proteins are within appropriate proximity for direct binding. Additionally, we demonstrate that there are functional consequences of this interaction, with inhibition of NCC affecting the function of ENaC. This novel finding of an association between ENaC and NCC could alter our understanding of salt transport in the distal tubule.


Subject(s)
Epithelial Sodium Channels/metabolism , Sodium Chloride Symporters/metabolism , Animals , Cell Line , Fluorescence Resonance Energy Transfer , Kidney Cortex/metabolism , Mice , Microscopy, Confocal , Protein Binding , Two-Hybrid System Techniques
6.
J Biol Chem ; 290(48): 28805-11, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26451045

ABSTRACT

The renal epithelial sodium channel (ENaC) provides regulated sodium transport in the distal nephron. The effects of intracellular calcium ([Ca(2+)]i) on this channel are only beginning to be elucidated. It appears from previous studies that the [Ca(2+)]i increases downstream of ATP administration may have a polarized effect on ENaC, where apical application of ATP and the subsequent [Ca(2+)]i increase have an inhibitory effect on the channel, whereas basolateral ATP and [Ca(2+)]i have a stimulatory effect. We asked whether this polarized effect of ATP is, in fact, reflective of a polarized effect of increased [Ca(2+)]i on ENaC and what underlying mechanism is responsible. We began by performing patch clamp experiments in which ENaC activity was measured during apical or basolateral application of ionomycin to increase [Ca(2+)]i near the apical or basolateral membrane, respectively. We found that ENaC does indeed respond to increased [Ca(2+)]i in a polarized fashion, with apical increases being inhibitory and basolateral increases stimulating channel activity. In other epithelial cell types, mitochondria sequester [Ca(2+)]i, creating [Ca(2+)]i signaling microdomains within the cell that are dependent on mitochondrial localization. We found that mitochondria localize in bands just beneath the apical and basolateral membranes in two different cortical collecting duct principal cell lines and in cortical collecting duct principal cells in mouse kidney tissue. We found that inhibiting mitochondrial [Ca(2+)]i uptake destroyed the polarized response of ENaC to [Ca(2+)]i. Overall, our data suggest that ENaC is regulated by [Ca(2+)]i in a polarized fashion and that this polarization is maintained by mitochondrial [Ca(2+)]i sequestration.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Epithelial Sodium Channels/metabolism , Kidney Tubules, Collecting/metabolism , Mitochondria/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Line , Mice , Xenopus laevis
7.
Biochim Biophys Acta ; 1848(11 Pt A): 2859-67, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26277265

ABSTRACT

This investigation was conducted to study the relationship between intracellular Ca(2+) and activation of large conductance Ca(2+)-activated K(+) (BK) currents by unoprostone, the first synthetic docosanoid. We used HEK293 cells stably transfected with two BK channel splice variants, one sensitive to unoprostone and the other insensitive. We examined the effects of unoprostone on channel activity in excised inside-out patches and cell-attached patches. The half-maximal stimulation of the sensitive BK channels by Ca(2+) was shifted from 3.4±0.017 nM to 0.81±.0058 nM in the presence of 10 nM unoprostone. There was no effect on insensitive channels even at unoprostone concentrations as high as 1000 nM. There was no effect of unoprostone on the voltage dependence of the BK channels. Changes in open probability and effects of Ca(2+) and unoprostone were best described by a synergistic binding model. These data would suggest that Ca(2+) and unoprostone were binding to sites close to one another on the channel protein and that unoprostone binding causes the affinity of the calcium binding site to increase. This idea is consistent with three dimensional models of the Ca(2+) binding site and a putative unoprostone binding domain. Our results have important implications for the clinical use of unoprostone to activate BK channels. Channel activation will be limited if intracellular Ca(2+) is not elevated.


Subject(s)
Calcium/metabolism , Dinoprost/analogs & derivatives , Ion Channel Gating/drug effects , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/physiology , Alternative Splicing , Amino Acid Sequence , Animals , Dinoprost/pharmacology , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Membrane Potentials/drug effects , Molecular Sequence Data , Patch-Clamp Techniques , Protein Isoforms/genetics , Protein Isoforms/physiology , Rats , Sequence Homology, Amino Acid , Transfection
8.
Am J Physiol Renal Physiol ; 309(5): F456-63, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26136560

ABSTRACT

Phosphatidylinositol bisphosphate (PIP2) regulates epithelial sodium channel (ENaC) open probability. In turn, myristoylated alanine-rich C kinase substrate (MARCKS) protein or MARCKS-like protein 1 (MLP-1) at the plasma membrane regulates the delivery of PIP2 to ENaC. MARCKS and MLP-1 are regulated by changes in cytosolic calcium; increasing calcium promotes dissociation of MARCKS from the membrane, but the calcium-regulatory mechanisms are unclear. However, it is known that increased intracellular calcium can activate calmodulin and we show that inhibition of calmodulin with calmidazolium increases ENaC activity presumably by regulating MARCKS and MLP-1. Activated calmodulin can regulate MARCKS and MLP-1 in two ways. Calmodulin can bind to the effector domain of MARCKS or MLP-1, inactivating both proteins by causing their dissociation from the membrane. Mutations in MARCKS that prevent calmodulin association prevent dissociation of MARCKS from the membrane. Calmodulin also activates CaM kinase II (CaMKII). An inhibitor of CaMKII (KN93) increases ENaC activity, MARCKS association with ENaC, and promotes MARCKS movement to a membrane fraction. CaMKII phosphorylates filamin. Filamin is an essential component of the cytoskeleton and promotes association of ENaC, MARCKS, and MLP-1. Disruption of the cytoskeleton with cytochalasin E reduces ENaC activity. CaMKII phosphorylation of filamin disrupts the cytoskeleton and the association of MARCKS, MLP-1, and ENaC, thereby reducing ENaC open probability. Taken together, these findings suggest calmodulin and CaMKII modulate ENaC activity by destabilizing the association between the actin cytoskeleton, ENaC, and MARCKS, or MLP-1 at the apical membrane.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calmodulin/metabolism , Cytoskeleton/metabolism , Epithelial Sodium Channels/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Nephrons/metabolism , Animals , Calcium/metabolism , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cytoskeleton/drug effects , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Filamins/metabolism , Imidazoles/pharmacology , Myristoylated Alanine-Rich C Kinase Substrate , Nephrons/cytology , Nephrons/drug effects , Xenopus
9.
Am J Physiol Renal Physiol ; 309(3): F251-8, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25925258

ABSTRACT

Inhibition of the epithelial Na(+) channel (ENaC) reduces Cl(-) absorption in cortical collecting ducts (CCDs) from aldosterone-treated rats and mice. Since ENaC does not transport Cl(-), the purpose of the present study was to explore how ENaC modulates Cl(-) absorption in mouse CCDs perfused in vitro. Therefore, we measured transepithelial Cl(-) flux and transepithelial voltage in CCDs perfused in vitro taken from mice that consumed a NaCl-replete diet alone or the diet with aldosterone administered by minipump. We observed that application of an ENaC inhibitor [benzamil (3 µM)] to the luminal fluid unmasks conductive Cl(-) secretion. During ENaC blockade, this Cl(-) secretion fell with the application of a nonselective Cl(-) channel blocker [DIDS (100 µM)] to the perfusate. While single channel recordings of intercalated cell apical membranes in split-open CCDs demonstrated a Cl(-) channel with properties that resemble the ClC family of Cl(-) channels, ClC-5 is not the primary pathway for benzamil-sensitive Cl(-) flux. In conclusion, first, in CCDs from aldosterone-treated mice, most Cl(-) absorption is benzamil sensitive, and, second, benzamil application stimulates stilbene-sensitive conductive Cl(-) secretion, which occurs through a ClC-5-independent pathway.


Subject(s)
4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Chlorides/metabolism , Hydrochloric Acid/metabolism , Kidney Tubules, Collecting/metabolism , Sodium Channel Blockers/pharmacology , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/antagonists & inhibitors , Algorithms , Amiloride/pharmacology , Animals , Cell Membrane/drug effects , Diuretics/pharmacology , Epithelial Sodium Channels/genetics , Female , Male , Mice , Mice, Knockout
10.
Am J Physiol Renal Physiol ; 309(2): F154-63, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25972513

ABSTRACT

The present study explored whether the intercalated cell Cl(-)/HCO3(-) exchanger pendrin modulates epithelial Na(+) channel (ENaC) function by changing channel open probability and/or channel density. To do so, we measured ENaC subunit subcellular distribution by immunohistochemistry, single channel recordings in split open cortical collecting ducts (CCDs), as well as transepithelial voltage and Na(+) absorption in CCDs from aldosterone-treated wild-type and pendrin-null mice. Because pendrin gene ablation reduced 70-kDa more than 85-kDa γ-ENaC band density, we asked if pendrin gene ablation interferes with ENaC cleavage. We observed that ENaC-cleaving protease application (trypsin) increased the lumen-negative transepithelial voltage in pendrin-null mice but not in wild-type mice, which raised the possibility that pendrin gene ablation blunts ENaC cleavage, thereby reducing open probability. In mice harboring wild-type ENaC, pendrin gene ablation reduced ENaC-mediated Na(+) absorption by reducing channel open probability as well as by reducing channel density through changes in subunit total protein abundance and subcellular distribution. Further experiments used mice with blunted ENaC endocytosis and degradation (Liddle's syndrome) to explore the significance of pendrin-dependent changes in ENaC open probability. In mouse models of Liddle's syndrome, pendrin gene ablation did not change ENaC subunit total protein abundance, subcellular distribution, or channel density, but markedly reduced channel open probability. We conclude that in mice harboring wild-type ENaC, pendrin modulates ENaC function through changes in subunit abundance, subcellular distribution, and channel open probability. In a mouse model of Liddle's syndrome, however, pendrin gene ablation reduces channel activity mainly through changes in open probability.


Subject(s)
Anion Transport Proteins/physiology , Epithelial Sodium Channels/metabolism , Kidney Tubules, Collecting/physiology , Sodium/metabolism , Animals , Disease Models, Animal , Female , Liddle Syndrome/genetics , Liddle Syndrome/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Sulfate Transporters , Trypsin
11.
Am J Physiol Renal Physiol ; 308(7): F697-705, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25587116

ABSTRACT

Many hormonal pathways contribute to the regulation of renal epithelial sodium channel (ENaC) function, a key process for maintaining blood volume and controlling blood pressure. In the present study, we examined whether the peptide hormone prolactin (PRL) regulates ENaC function in renal epithelial cells (A6). Basolateral application of several different concentrations of PRL dramatically stimulated the transepithelial current in A6 cells, increasing both amiloride-sensitive (ENaC) and amiloride-insensitive currents. Using cell-attached patch clamp, we determined that PRL increased both the number (N) and open probability (Po) of ENaC present in the apical membrane. Inhibition of PKA with H-89 abolished the effect of PRL on amiloride-sensitive and insensitive transepithelial currents and eliminated the increase in ENaC NPo with PRL exposure. PRL also increased cAMP in A6 cells, consistent with signaling through the cAMP-dependent PKA pathway. We also identified that PRL induced activity of a 2-pS anion channel with outward rectification, electrophysiological properties consistent with ClC4 or ClC5. RT-PCR only detected ClC4, but not ClC5 transcripts. Here, we show for the first time that PRL activates sodium and chloride transport in renal epithelial cells via ENaC and ClC4.


Subject(s)
Chloride Channels/metabolism , Epithelial Cells/drug effects , Epithelial Sodium Channels/metabolism , Prolactin/pharmacology , Sodium/metabolism , Amiloride/pharmacology , Animals , Cell Line , Cyclic AMP/metabolism , Epithelial Cells/metabolism , Mice , Patch-Clamp Techniques/methods
12.
J Am Soc Nephrol ; 26(4): 844-54, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25145935

ABSTRACT

With no lysine (WNK) kinases are members of the serine/threonine kinase family. We previously showed that WNK4 inhibits renal large-conductance Ca(2+)-activated K(+) (BK) channel activity by enhancing its degradation through a lysosomal pathway. In this study, we investigated the effect of WNK1 on BK channel activity. In HEK293 cells stably expressing the α subunit of BK (HEK-BKα cells), siRNA-mediated knockdown of WNK1 expression significantly inhibited both BKα channel activity and open probability. Knockdown of WNK1 expression also significantly inhibited BKα protein expression and increased ERK1/2 phosphorylation, whereas overexpression of WNK1 significantly enhanced BKα expression and decreased ERK1/2 phosphorylation in a dose-dependent manner in HEK293 cells. Knockdown of ERK1/2 prevented WNK1 siRNA-mediated inhibition of BKα expression. Similarly, pretreatment of HEK-BKα cells with the lysosomal inhibitor bafilomycin A1 reversed the inhibitory effects of WNK1 siRNA on BKα expression in a dose-dependent manner. Knockdown of WNK1 expression also increased the ubiquitination of BKα channels. Notably, mice fed a high-K(+) diet for 10 days had significantly higher renal protein expression levels of BKα and WNK1 and lower levels of ERK1/2 phosphorylation compared with mice fed a normal-K(+) diet. These data suggest that WNK1 enhances BK channel function by reducing ERK1/2 signaling-mediated lysosomal degradation of the channel.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , MAP Kinase Signaling System , Protein Serine-Threonine Kinases/metabolism , Animals , HEK293 Cells , Humans , Lysosomes/metabolism , Mice, Inbred C57BL , Minor Histocompatibility Antigens , WNK Lysine-Deficient Protein Kinase 1 , Water-Electrolyte Balance
13.
Am J Physiol Renal Physiol ; 307(7): F806-13, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25100278

ABSTRACT

The polarized nature of epithelial cells allows for different responses to luminal or serosal stimuli. In kidney tubules, ATP is produced luminally in response to changes in luminal flow. Luminal increases in ATP have been previously shown to inhibit the renal epithelial Na⁺ channel (ENaC). On the other hand, ATP is increased basolaterally in renal epithelia in response to aldosterone. We tested the hypothesis that basolateral ATP can stimulate ENaC function through activation of the P2X4receptor/channel. Using single channel cell-attached patch-clamp techniques, we demonstrated the existence of a basolaterally expressed channel stimulated by the P2X4agonist 2-methylthio-ATP (meSATP) in Xenopus A6 cells, a renal collecting duct principal cell line. This channel had a similar reversal potential and conductance to that of P2X4channels. Cell surface biotinylation of the basolateral side of these cells confirmed the basolateral presence of the P2X4 receptor. Basolateral addition of meSATP enhanced the activity of ENaC in single channel patch-clamp experiments, an effect that was absent in cells transfected with a dominant negative P2X4receptor construct, indicating that activation of P2X4channels stimulates ENaC activity in these cells. The effect of meSATP on ENaC activity was reduced after chelation of basolateral Ca²âº with EGTA or inhibition of phosphatidylinositol 3-kinase with LY-294002. Overall, our results show that ENaC is stimulated by P2X4receptor activation and that the stimulation is dependent on increases in intracellular Ca²âº and phosphatidylinositol 3-kinase activation.


Subject(s)
Calcium/metabolism , Epithelial Sodium Channels/metabolism , Kidney Tubules, Collecting/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Purinergic P2X4/metabolism , Animals , Cell Line , Xenopus
14.
Am J Respir Crit Care Med ; 190(5): 522-32, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25029038

ABSTRACT

RATIONALE: Alveolar liquid clearance is regulated by Na(+) uptake through the apically expressed epithelial sodium channel (ENaC) and basolaterally localized Na(+)-K(+)-ATPase in type II alveolar epithelial cells. Dysfunction of these Na(+) transporters during pulmonary inflammation can contribute to pulmonary edema. OBJECTIVES: In this study, we sought to determine the precise mechanism by which the TIP peptide, mimicking the lectin-like domain of tumor necrosis factor (TNF), stimulates Na(+) uptake in a homologous cell system in the presence or absence of the bacterial toxin pneumolysin (PLY). METHODS: We used a combined biochemical, electrophysiological, and molecular biological in vitro approach and assessed the physiological relevance of the lectin-like domain of TNF in alveolar liquid clearance in vivo by generating triple-mutant TNF knock-in mice that express a mutant TNF with deficient Na(+) uptake stimulatory activity. MEASUREMENTS AND MAIN RESULTS: TIP peptide directly activates ENaC, but not the Na(+)-K(+)-ATPase, upon binding to the carboxy-terminal domain of the α subunit of the channel. In the presence of PLY, a mediator of pneumococcal-induced pulmonary edema, this binding stabilizes the ENaC-PIP2-MARCKS complex, which is necessary for the open probability conformation of the channel and preserves ENaC-α protein expression, by means of blunting the protein kinase C-α pathway. Triple-mutant TNF knock-in mice are more prone than wild-type mice to develop edema with low-dose intratracheal PLY, correlating with reduced pulmonary ENaC-α subunit expression. CONCLUSIONS: These results demonstrate a novel TNF-mediated mechanism of direct ENaC activation and indicate a physiological role for the lectin-like domain of TNF in the resolution of alveolar edema during inflammation.


Subject(s)
Epithelial Sodium Channel Agonists/metabolism , Epithelial Sodium Channels/metabolism , Peptides, Cyclic/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Edema/metabolism , Streptolysins , Tumor Necrosis Factor-alpha/metabolism , Animals , Bacterial Proteins , Epithelial Sodium Channel Agonists/chemistry , Epithelial Sodium Channels/chemistry , Female , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Peptides, Cyclic/chemistry , Pulmonary Alveoli/microbiology , Pulmonary Edema/microbiology , Tumor Necrosis Factor-alpha/chemistry
15.
Am J Physiol Lung Cell Mol Physiol ; 307(5): L374-85, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25015976

ABSTRACT

We used a PKC-α knockout model to investigate the regulation of alveolar epithelial Na(+) channels (ENaC) by PKC. Primary alveolar type II (ATII) cells were subjected to cell-attached patch clamp. In the absence of PKC-α, the open probability (Po) of ENaC was decreased by half compared with wild-type mice. The channel density (N) was also reduced in the knockout mice. Using in vivo biotinylation, membrane localization of all three ENaC subunits (α, ß, and γ) was decreased in the PKC-α knockout lung, compared with the wild-type. Confocal microscopy of lung slices showed elevated levels of reactive oxygen species (ROS) in the lungs of the PKC-α knockout mice vs. the wild-type. High levels of ROS in the knockout lung can be explained by a decrease in both cytosolic and mitochondrial superoxide dismutase activity. Elevated levels of ROS in the knockout lung activates PKC-δ and leads to reduced dephosphorylation of ERK1/2 by MAP kinase phosphatase, which in turn causes increased internalization of ENaC via ubiquitination by the ubiquitin-ligase Nedd4-2. In addition, in the knockout lung, PKC-δ activates ERK, causing a decrease in ENaC density at the apical alveolar membrane. PKC-δ also phosphorylates MARCKS, leading to a decrease in ENaC Po. The effects of ROS and PKC-δ were confirmed with patch-clamp experiments on isolated ATII cells in which the ROS scavenger, Tempol, or a PKC-δ-specific inhibitor added to patches reversed the observed decrease in ENaC apical channel density and Po. These results explain the decrease in ENaC activity in PKC-α knockout lung.


Subject(s)
Epithelial Cells/metabolism , Epithelial Sodium Channels/metabolism , Lung/metabolism , Protein Kinase C-alpha/physiology , Pulmonary Alveoli/metabolism , Animals , Epithelial Cells/cytology , Female , Immunoblotting , Male , Mice , Mice, Knockout , Pulmonary Alveoli/cytology , Reactive Oxygen Species/metabolism
16.
Am J Physiol Renal Physiol ; 307(1): F86-95, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24829507

ABSTRACT

Numerous reports have linked cytoskeleton-associated proteins with the regulation of epithelial Na(+) channel (ENaC) activity. The purpose of the present study was to determine the effect of actin cytoskeleton disruption by cytochalasin E on ENaC activity in Xenopus 2F3 cells. Here, we show that cytochalasin E treatment for 60 min can disrupt the integrity of the actin cytoskeleton in cultured Xenopus 2F3 cells. We show using single channel patch-clamp experiments and measurements of short-circuit current that ENaC activity, but not its density, is altered by cytochalasin E-induced disruption of the cytoskeleton. In nontreated cells, 8 of 33 patches (24%) had no measurable ENaC activity, whereas in cytochalasin E-treated cells, 17 of 32 patches (53%) had no activity. Analysis of those patches that did contain ENaC activity showed channel open probability significantly decreased from 0.081 ± 0.01 in nontreated cells to 0.043 ± 0.01 in cells treated with cytochalasin E. Transepithelial current from mpkCCD cells treated with cytochalasin E, cytochalasin D, or latrunculin B for 60 min was decreased compared with vehicle-treated cells. The subcellular expression of fodrin changed significantly, and several protein elements of the cytoskeleton decreased at least twofold after 60 min of cytochalasin E treatment. Cytochalasin E treatment disrupted the association between ENaC and myristoylated alanine-rich C-kinase substrate. The results presented here suggest disruption of the actin cytoskeleton by different compounds can attenuate ENaC activity through a mechanism involving changes in the subcellular expression of fodrin, several elements of the cytoskeleton, and destabilization of the ENaC-myristoylated alanine-rich C-kinase substrate complex.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Cytochalasins/pharmacology , Cytoskeleton/metabolism , Epithelial Sodium Channels/metabolism , Actins/metabolism , Animals , Carrier Proteins/metabolism , Cell Line , Cell Membrane/metabolism , Cytochalasin D/metabolism , Cytoskeleton/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Myristoylated Alanine-Rich C Kinase Substrate , Xenopus , Xenopus laevis
17.
BMC Vet Res ; 10: 2, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24386990

ABSTRACT

BACKGROUND: Toll-like receptor (TLR) agonists reportedly have potent antiviral and antitumor activities and may be a new kind of adjuvant for enhancing immune efficacy. Resiquimod (R848) is an imidazoquinoline compound with potent antiviral activity and functions through the TLR7/TLR8 MyD88-dependent signaling pathway. Polyinosinic-polycytidylic acid [poly(I:C)] is a synthetic analog of double-stranded RNA that induces the production of pro-inflammatory cytokines by the activation of NF-κB through TLR3. This study investigated the potential of R848 and poly(I:C) as an adjuvant 146S foot-and-mouth disease virus (FMDV) vaccine formulated with aluminum hydroxide (Al(OH)3). RESULTS: Antibody titers to FMDV and CD8+ T cells were markedly enhanced in mice immunized to 146S FMDV + Al(OH)3 + R848 + poly(I:C) compared with mice immunized to FMDV + ISA206. IFN-γ secretion substantially increased compared with IL-4 secretion by splenic T cells stimulated with FMDV antigens in vitro, suggesting that R848, poly(I:C), and with Al(OH)3 together biased the immune response toward a Th1-type direction. CONCLUSIONS: These results indicated that the R848 and poly(I:C) together with Al(OH)3 enhanced humoral and cellular immune responses to immunization with 146S FMDV antigens. Thus, this new vaccine formulation can be used for FMDV prevention.


Subject(s)
Adjuvants, Immunologic/pharmacology , Aluminum Hydroxide/pharmacology , Foot-and-Mouth Disease/prevention & control , Imidazoles/pharmacology , Poly I-C/pharmacology , Viral Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Aluminum Hydroxide/administration & dosage , Aluminum Hydroxide/chemistry , Animals , Antibodies, Viral/blood , Antibody Specificity , Female , Foot-and-Mouth Disease/immunology , Imidazoles/administration & dosage , Mice , Mice, Inbred BALB C , Poly I-C/administration & dosage , Spleen/cytology , Spleen/drug effects , T-Lymphocyte Subsets , Viral Vaccines/administration & dosage
18.
Am J Physiol Renal Physiol ; 306(3): F309-20, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24338818

ABSTRACT

The epithelial Na channel (ENaC) is negatively regulated by protein kinase C (PKC) as shown using PKC activators in a cell culture model. To determine whether PKCα influences ENaC activity in vivo, we examined the regulation of ENaC in renal tubules from PKCα⁻/⁻ mice. Cortical collecting ducts were dissected and split open, and the exposed principal cells were subjected to cell-attached patch clamp. In the absence of PKCα, the open probability (P0) of ENaC was increased three-fold vs. wild-type SV129 mice (0.52 ± 0.04 vs. 0.17 ± 0.02). The number of channels per patch was also increased. Using confocal microscopy, we observed an increase in membrane localization of α-, ß-, and γ-subunits of ENaC in principal cells in the cortical collecting ducts of PKCα⁻/⁻ mice compared with wild-type mice. To confirm this increase, one kidney from each animal was perfused with biotin, and membrane protein was pulled down with streptavidin. The nonbiotinylated kidney was used to assess total protein. While total ENaC protein did not change in PKCα⁻/⁻ mice, membrane localization of all the ENaC subunits was increased. The increase in membrane ENaC could be explained by the observation that ERK1/2 phosphorylation was decreased in the knockout mice. These results imply a reduction in ENaC membrane accumulation and P0 by PKCα in vivo. The PKC-mediated increase in ENaC activity was associated with an increase in blood pressure in knockout mice fed a high-salt diet.


Subject(s)
Epithelial Sodium Channels/metabolism , Kidney Tubules, Collecting/cytology , Protein Kinase C-alpha/deficiency , Aldosterone/blood , Animals , Aquaporin 2/metabolism , Blood Pressure/drug effects , Kidney Tubules, Collecting/physiology , Mice , Mice, Knockout , Microscopy, Confocal , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Patch-Clamp Techniques , Phosphorylation , Protein Kinase C-alpha/metabolism , Sodium Chloride, Dietary/administration & dosage
19.
Am J Physiol Renal Physiol ; 305(9): F1365-73, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23863469

ABSTRACT

Epithelial Na(+) channel (ENaC) activity, which determines the rate of renal Na(+) reabsorption, can be regulated by G protein-coupled receptors. Regulation of ENaC by Gα-mediated downstream effectors has been studied extensively, but the effect of Gßγ dimers on ENaC is unclear. A6 cells endogenously contain high levels of Gß1 but low levels of Gß3, Gß4, and Gß5 were detected by Q-PCR. We tested Gγ2 combined individually with Gß1 through Gß5 expressed in A6 cells, after which we recorded single-channel ENaC activity. Among the five ß and γ2 combinations, ß1γ2 strongly inhibits ENaC activity by reducing both ENaC channel number (N) and open probability (Po) compared with control cells. In contrast, the other four ß-isoforms combined with γ2 have no significant effect on ENaC activity. By using various inhibitors to probe Gß1γ2 effects on ENaC regulation, we found that Gß1γ2-mediated ENaC inhibition involved activation of phospholipase C-ß and its enzymatic products that induce protein kinase C and ERK1/2 signaling pathways.


Subject(s)
Epithelial Sodium Channels/metabolism , GTP-Binding Protein beta Subunits/physiology , GTP-Binding Protein gamma Subunits/physiology , Kidney/metabolism , Sodium/metabolism , Animals , Cell Line , Extracellular Signal-Regulated MAP Kinases/physiology , Ion Channel Gating/physiology , Patch-Clamp Techniques , Phosphoinositide Phospholipase C/metabolism , Protein Isoforms/physiology , Signal Transduction/physiology , Xenopus laevis
20.
Am J Physiol Renal Physiol ; 305(1): F31-41, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23594824

ABSTRACT

A serine-threonine protein kinase, WNK4, reduces Na⁺ reabsorption and K⁺ secretion in the distal convoluted tubule by reducing trafficking of the thiazide-sensitive Na-Cl cotransporter to and enhancing renal outer medullary potassium channel retrieval from the apical membrane. Epithelial sodium channels (ENaC) in the distal nephron also play a role in regulating Na⁺ reabsorption and are also regulated by WNK4, but the mechanism is unclear. In A6 distal nephron cells, transepithelial current measurement and single channel recording show that WNK4 inhibits ENaC activity. Analysis of the number of channel per patch shows that WNK4 reduces channel number but has no effect on channel open probability. Western blots of apical and total ENaC provide additional evidence that WNK4 reduces apical as well as total ENaC expression. WNK4 enhances ENaC internalization independent of Nedd4-2-mediated ENaC ubiquitination. WNK4 also reduced the amount of ENaC available for recycling but has no effect on the rate of transepithelial current increase to forskolin. In contrast, Nedd4-2 not only reduced ENaC in the recycling pool but also decreased the rate of increase of current after forskolin. WNK4 associates with wild-type as well as Liddle's mutated ENaC, and WNK4 reduces both wild-type and mutated ENaC expressed in HEK293 cells.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Epithelial Sodium Channels/metabolism , Nephrons/enzymology , Protein Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Blotting, Western , Colforsin/pharmacology , Endosomal Sorting Complexes Required for Transport/genetics , Epithelial Sodium Channels/drug effects , Epithelial Sodium Channels/genetics , HEK293 Cells , Humans , Membrane Potentials , Mutation , Nedd4 Ubiquitin Protein Ligases , Nephrons/drug effects , Patch-Clamp Techniques , Protein Serine-Threonine Kinases/genetics , Protein Transport , Time Factors , Transfection , Ubiquitin-Protein Ligases/genetics , Ubiquitination , Xenopus Proteins/metabolism , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL