Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
Add more filters











Publication year range
1.
Biochem Biophys Res Commun ; 350(1): 39-49, 2006 Nov 10.
Article in English | MEDLINE | ID: mdl-16997278

ABSTRACT

Large conductance, calcium-activated potassium channels (maxi K- or BK-channels) can be regulated by arachidonic acid produced by c-Phospholipase A2 (c-PLA2). Since in every excised patch from GH3 cells where there was BK-channel activity, treatment with either a stimulator or inhibitor of c-PLA2 resulted in a corresponding increase or decrease in BK-channel activity, we hypothesized that there must be a close association between BK-channel proteins and c-PLA2 in the cell membrane. To test this hypothesis, we first determined whether the two proteins would co-immunoprecipitate. We then used confocal imaging of fluorescently tagged proteins to determine where in the cells BK-channel proteins and c-PLA2 co-localize. The alpha-subunit of the BK-channel was strongly co-immunoprecipitated by c-PLA2 antibodies, suggesting that most of the BK channel alpha-subunits are associated with c-PLA2. This interaction was not affected by pharmacologically inhibiting c-PLA2 suggesting that the association does not require functionally active c-PLA2. Following dual immunohistochemical labeling and confocal microscopy, image analysis revealed that in the cytosol there was some co-localization, but most of the c-PLA2 was separate from BK-channel proteins. On the other hand, the c-PLA2 and BK-channel proteins at the plasma membrane were strongly co-localized. Immunoprecipitation experiments conducted with plasma membrane proteins support these findings. We conclude that c-PLA2 is likely physically associated with BK-channel proteins at the cell surface.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Phospholipases A/metabolism , Animals , Calcimycin/pharmacology , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Immunohistochemistry , Phospholipases A2 , Protein Binding , Protein Subunits/metabolism , Protein Transport , Rats
2.
Am J Physiol Renal Physiol ; 290(6): F1285-94, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16682484

ABSTRACT

Amiloride-sensitive epithelial Na+ channels (ENaC) play a crucial role in Na+ transport and fluid reabsorption in the kidney, lung, and colon. The magnitude of ENaC-mediated Na+ transport in epithelial cells depends on the average open probability of the channels and the number of channels on the apical surface of epithelial cells. The number of channels in the apical membrane, in turn, depends on a balance between the rate of ENaC insertion and the rate of removal from the apical membrane. ENaC is made up of three homologous subunits: alpha, beta, and gamma. The COOH-terminal domain of all three subunits is intracellular and contains a proline-rich motif (PPxY). Mutations or deletion of this PPxY motif in the beta- and gamma-subunits prevent the binding of one isoform of a specific ubiquitin ligase, neural precursor cell-expressed, developmentally downregulated protein (Nedd4-2), to the channel in vitro and in transfected cell systems, thereby impeding ubiquitin conjugation of the channel subunits. Ubiquitin conjugation would seem to imply that ENaC turnover is determined by the ubiquitin-proteasome system, but when Madin-Darby canine kidney cells are transfected with ENaC, ubiquitin conjugation apparently leads to lysosomal degradation. However, in untransfected renal cells (A6) expressing endogenous ENaC, ENaC is indeed degraded by the ubiquitin-proteasome system. Nonetheless, in both transfected and untransfected cells, the rate of ENaC degradation is apparently controlled by Nedd4-2 activity. In this review, we discuss the role of the ubiquitin conjugation and the alternative degradative pathways (lysosomal or proteasomal) in regulating the rate of ENaC turnover in untransfected renal cells and compare this regulation to that of transfected cell systems.


Subject(s)
Epithelial Cells/chemistry , Proteasome Endopeptidase Complex/metabolism , Sodium Channels/physiology , Ubiquitin/metabolism , Animals , Biological Transport/physiology , Cell Line , Dogs , Endosomal Sorting Complexes Required for Transport , Epithelial Cells/metabolism , Epithelial Sodium Channels , Humans , Kidney , Membrane Proteins/metabolism , Mutation , Nedd4 Ubiquitin Protein Ligases , Proline , Protein Subunits/chemistry , Protein Subunits/physiology , Sodium/metabolism , Sodium Channels/chemistry , Sodium Channels/genetics , Structure-Activity Relationship , Transfection , Ubiquitin-Protein Ligases/metabolism
3.
Am J Physiol Renal Physiol ; 289(1): F107-16, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15769939

ABSTRACT

Amiloride-sensitive epithelial sodium channels (ENaC) are responsible for transepithelial Na(+) transport in the kidney, lung, and colon. The channel consists of three subunits (alpha, beta, and gamma). In Madin-Darby canine kidney (MDCK) cells and Xenopus laevis oocytes transfected with all three ENaC subunits, neural precursor cell-expressed developmentally downregulated protein (Nedd4-2) promotes ubiquitin conjugation of ENaC. For native proteins in some cells, ubiquitin conjugation is a signal for their degradation by the ubiquitin-proteasome pathway, whereas in other cell types ubiquitin conjugation is a signal for endocytosis and lysosomal protein degradation. When ENaC are transfected into MDCK cells, ubiquitin conjugation leads to lysosomal degradation. In this paper, we characterize the involvement of the ubiquitin-proteasome proteolytic pathway in the regulation of functional ENaC in untransfected renal A6 cells expressing native ENaC subunits. In contrast to transfected cells, we show that total cellular alpha-, beta-, and gamma-ENaC subunits are polyubiquitinated and that ubiquitin conjugation of subunits increases when the cells are treated with a proteasome inhibitor. We show that Nedd4-2 is associated with alpha- and beta-subunits and is associated with the apical membrane. We also show the Nedd4-2 can regulate the number of functional ENaC subunits in the apical membrane. The results reported here suggest that the ubiquitin-proteasome proteolytic pathway is an important determinant of ENaC function in untransfected renal cells expressing endogenous ENaC.


Subject(s)
Kidney/metabolism , Polyubiquitin/physiology , Sodium Channels/metabolism , Ubiquitin-Protein Ligases/physiology , Animals , Cell Line , Endosomal Sorting Complexes Required for Transport , Epithelial Cells/metabolism , Epithelial Sodium Channels , Kidney/cytology , Nedd4 Ubiquitin Protein Ligases , Xenopus Proteins , Xenopus laevis
4.
J Neurophysiol ; 93(6): 3146-56, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15647401

ABSTRACT

BK-channels in GH3 cells are activated by arachidonic acid produced by c-PLA2. beta-adrenergic agonists also activate BK channels and were presumed to do so via production of cAMP. We, however, show for the first time in GH3 cells that a beta-adrenergic agonist activates a pertussis-toxin-sensitive G protein that activates c-PLA2. The arachidonic acid produced by c-PLA2 then activates BK channels. We examined BK channels in cell-attached patches and in excised patches from untreated GH3 cells and from GH3 cells exposed to c-PLA2 antisense oligonucleotides. For the cell-attached patch experiments, physiologic pipette and bath solutions were used. For the excised patches, 150 mM KCl was used in both the pipette and bath solutions, and the cytosolic surface contained 1 microM free Ca2+ (buffered with 5 mM K2EGTA). Treatment of GH3 cells with the G protein activator, fluoroaluminate, (AlF4-) produced an increase in the Po of BK channels of 177 +/- 41% (mean +/- SD) in cell-attached patches. Because G proteins are membrane associated, we also added an activator of G proteins, 100 microM GTP-gamma-S, to the cytosolic surface of excised patches. This treatment leads to an increase in Po of 50 +/- 9%. Similar treatment of excised patches with GDP-beta-S had no effect on Po. Isoproterenol (1 microM), an activator of beta-adrenergic receptors and, consequently, some G proteins, increased BK channel activity 229 +/- 37% in cell-attached patches from cultured GH3 cells. Western blot analysis showed that GH3 cells have beta-adrenergic receptor protein and that isoproterenol acts through these receptors because the beta-adrenergic receptor antagonist, propanolol, blocks the action of isoproterenol. To test whether G protein activation of BK channels involves c-PLA2, we studied the effects of GTP-gamma-S on excised patches and isoproterenol on cell attached patches from GH3 cells previously treated with c-PLA2 antisense oligonucleotides or pharmacological inhibitors of c-PLA2. Neither isoproterenol nor GTP-gamma-S had any effect on Po in these patches. Similarly, neither isoproterenol nor GTP-gamma-S had any effect on Po in cultured GH3 cells pretreated with pertussis toxin. Isoproterenol also significantly increased the rate of arachidonic production in GH3 cells. These results show that some receptor-linked, pertussis-toxin-sensitive G protein in GH3 cells can activate c-PLA2 to increase the amount of arachidonic acid present and ultimately increase BK-channel activity.


Subject(s)
GTP-Binding Proteins/physiology , Phospholipases A/metabolism , Pituitary Gland/metabolism , Potassium Channels, Calcium-Activated/metabolism , Signal Transduction/physiology , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Aluminum Compounds/pharmacology , Animals , Arachidonic Acids/pharmacology , Calcium/metabolism , Cell Line, Tumor , Drug Interactions , Enzyme Inhibitors/pharmacology , Fluorides/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Ion Channel Gating/drug effects , Isoproterenol/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Models, Biological , Oligodeoxyribonucleotides, Antisense/pharmacology , Patch-Clamp Techniques/methods , Phospholipases A2 , Picrotoxin/pharmacology , Pituitary Gland/drug effects , Pituitary Gland/physiology , Propranolol/pharmacology , Rats , Time Factors
5.
Am J Physiol Renal Physiol ; 283(5): F1030-45, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12372779

ABSTRACT

We studied the cellular phosphatase inhibitors okadaic acid (OKA), calyculin A, and microcystin on the epithelial sodium channel (ENaC) in A6 renal cells. OKA increased the amiloride-sensitive current after approximately 30 min with maximal stimulation at 1-2 h. Fluctuation analysis of cell-attached patches containing a large number of ENaC yielded power spectra with corner frequencies in untreated cells almost two times as large as in cells pretreated for 30 min with OKA, implying an increase in single channel open probability (P(o)) that doubled after OKA. Single channel analysis showed that, in cells pretreated with OKA, P(o) and mean open time approximately doubled. Two other phosphatase inhibitors, calyculin A and microcystin, had similar effects on P(o) and mean open time. An analog of OKA, okadaone, that does not inhibit phosphatases had no effect. Pretreatment with 10 nM OKA, which blocks protein phosphatase 2A (PP2A) but not PP1 in mammalian cells, had no effect even though both phosphatases are present in A6 cells. Several proteins were differentially phosphorylated after OKA, but ENaC subunit phosphorylation did not increase. We conclude that, in A6 cells, there is an OKA-sensitive phosphatase that suppresses ENaC activity by altering the phosphorylation of a regulatory molecule associated with the channel.


Subject(s)
Kidney/enzymology , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism , Sodium Channels/metabolism , Animals , Cell Line , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Inhibitors/pharmacology , Epithelial Sodium Channels , Ion Channel Gating/physiology , Isomerism , Kidney/cytology , Marine Toxins , Microcystins , Okadaic Acid/analogs & derivatives , Okadaic Acid/pharmacology , Oxazoles/pharmacology , Patch-Clamp Techniques/methods , Peptides, Cyclic/pharmacology , Phosphorylation , Protein Kinase C/metabolism , Protein Phosphatase 2 , Sodium Channels/chemistry
6.
Am J Physiol Cell Physiol ; 281(3): C773-85, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11502554

ABSTRACT

The A6 cell line was used to study the role of S-adenosyl-L-homocysteine hydrolase (SAHHase) in the aldosterone-induced activation of the epithelial Na(+) channel (ENaC). Because aldosterone increases methylation of several different molecules, and because this methylation is associated with increased Na(+) reabsorption, we tested the hypothesis that aldosterone increases the expression and activity of SAHHase protein. The rationale for this work is that general methylation may be promoted by activation of SAHHase, the only enzyme known to metabolize SAH, a potent end-product inhibitor of methylation. Although aldosterone increased SAHHase activity, steroid did not affect SAHHase expression. Antisense SAHHase oligonucleotide decreased SAHHase expression and activity. Moreover, this oligonucleotide, as well as a pharmacological inhibitor of SAHHase, decreased aldosterone-induced activity of ENaC via a decrease in ENaC open probability. The kinetics of ENaC in cells treated with antisense plus aldosterone were similar to those reported previously for the channel in the absence of steroid. This is the first report showing that active SAHHase, in part, increases ENaC open probability by reducing the transition rate from open states in response to aldosterone. Thus aldosterone-induced SAHHase activity plays a critical role in shifting ENaC from a gating mode with short open and closed times to one with longer open and closed times.


Subject(s)
Aldosterone/pharmacology , Hydrolases/genetics , Hydrolases/metabolism , Kidney/physiology , Sodium Channels/physiology , Urothelium/physiology , Adenosylhomocysteinase , Animals , Cell Membrane/physiology , Epithelial Sodium Channels , Isomerism , Kidney/cytology , Kinetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Methylation , Oligodeoxyribonucleotides/pharmacology , Oligodeoxyribonucleotides, Antisense/pharmacology , Patch-Clamp Techniques , RNA, Messenger/genetics , Recombinant Proteins/metabolism , Sodium/metabolism , Sodium Channels/drug effects , Transcription, Genetic , Transfection , Tubercidin/pharmacology , Urothelium/cytology , Urothelium/drug effects , Xenopus laevis
7.
Am J Physiol Renal Physiol ; 281(2): F213-21, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11457713

ABSTRACT

The renal epithelial cell line A6, derived from Xenopus laevis, expresses epithelial Na(+) channels (ENaCs) and serves as a model system to study hormonal regulation and turnover of ENaCs. Our previous studies suggest that the alpha-subunit of Xenopus ENaC (alpha-xENaC) is detectable as 150- and 180-kDa polypeptides, putative immature and mature alpha-subunit heterodimers. The 150- and 180-kDa alpha-xENaC were present in distinct fractions after sedimentation of A6 cell lysate through a sucrose density gradient. Two anti-alpha-xENaC antibodies directed against distinct domains demonstrated that only 180-kDa alpha-xENaC was expressed at the apical cell surface. The half-life of cell surface-expressed alpha-xENaC was 24-30 h, suggesting that once ENaC matures and is expressed at the plasma membrane, its turnover is similar to that reported for mature cystic fibrosis transmembrane conductance regulator. No significant changes in apical surface expression of alpha-xENaC were observed after treatment of A6 cells with aldosterone for 24 h, despite a 5.3-fold increase in short-circuit current. This lack of change in surface expression is consistent with previous observations in A6 cells and suggests that aldosterone regulates ENaC gating and increases channel open probability.


Subject(s)
Epithelial Cells/metabolism , Kidney Tubules/metabolism , Sodium Channels/metabolism , Urothelium/metabolism , Aldosterone/pharmacology , Animals , Cell Line , Cell Polarity , Centrifugation, Density Gradient , Epithelial Cells/drug effects , Epithelial Sodium Channels , Immunoblotting , Kidney Tubules/cytology , Kidney Tubules/drug effects , Precipitin Tests , Protein Subunits , Time Factors , Urothelium/cytology , Urothelium/drug effects , Xenopus laevis
8.
Am J Physiol Cell Physiol ; 281(1): C147-56, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11401837

ABSTRACT

Activity of the epithelial Na+ channel (ENaC) is the limiting step for discretionary Na+ reabsorption in the cortical collecting duct. Xenopus laevis kidney A6 cells were used to investigate the effects of cytosolic phospholipase A2 (cPLA2) activity on Na+ transport. Application of aristolochic acid, a cPLA2 inhibitor, to the apical membrane of monolayers produced a decrease in apical [3H]arachidonic acid (AA) release and led to an approximate twofold increase in transepithelial Na+ current. Increased current was abolished by the nonmetabolized AA analog 5,8,11,14-eicosatetraynoic acid (ETYA), suggesting that AA, rather than one of its metabolic products, affected current. In single channel studies, ETYA produced a decrease in ENaC open probability. This suggests that cPLA2 is tonically active in A6 cells and that the end effect of liberated AA at the apical membrane is to reduce Na+ transport via actions on ENaC. In contrast, aristolochic acid applied basolaterally inhibited current, and the effect was not reversed by ETYA. Basolateral application of the cyclooxygenase inhibitor ibuprofen also inhibited current. Both effects were reversed by prostaglandin E2 (PGE2). This suggests that cPLA2 activity and free AA, which is metabolized to PGE2, are necessary to support transport. This study supports the fine-tuning of Na+ transport and reabsorption through the regulation of free AA and AA metabolism.


Subject(s)
Aristolochic Acids , Phospholipases A/pharmacology , Sodium Channels/metabolism , Sodium/metabolism , 5,8,11,14-Eicosatetraynoic Acid/pharmacology , Animals , Cell Line , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/physiology , Dinoprostone/metabolism , Enzyme Inhibitors/pharmacology , Epithelial Sodium Channels , Fatty Acids, Unsaturated/pharmacology , Ion Transport , Kidney/cytology , Patch-Clamp Techniques , Phenanthrenes/pharmacology , Phospholipases A/antagonists & inhibitors , Phospholipases A2 , Xenopus laevis
9.
Am J Physiol Lung Cell Mol Physiol ; 280(4): L646-58, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11238004

ABSTRACT

Alveolar fluid clearance in the developing and mature lungs is believed to be mediated by some form of epithelial Na channels (ENaC). However, single-channel studies using isolated alveolar type II (ATII) cells have failed to demonstrate consistently the presence of highly selective Na+ channels that would be expected from ENaC expression. We postulated that in vitro culture conditions might be responsible for alterations in the biophysical properties of Na+ conductances observed in cultured ATII cells. When ATII cells were grown on glass plates submerged in media that lacked steroids, the predominant channel was a 21-pS nonselective cation channel (NSC) with a Na+-to-K+ selectivity of 1; however, when grown on permeable supports in the presence of steroids and air interface, the predominant channel was a low-conductance (6.6 +/- 3.4 pS, n = 94), highly Na+-selective channel (HSC) with a P(Na)/P(K) >80 that is inhibited by submicromolar concentrations of amiloride (K(0.5) = 37 nM) and is similar in biophysical properties to ENaC channels described in other epithelia. To establish the relationship of this HSC channel to the cloned ENaC, we employed antisense oligonucleotide methods to inhibit the individual subunit proteins of ENaC (alpha, beta, and gamma) and used patch-clamp techniques to determine the density of this channel in apical membrane patches of ATII cells. Overnight treatment of cells with antisense oligonucleotides to any of the three subunits of ENaC resulted in a significant decrease in the density of HSC channels in the apical membrane cell-attached patches. Taken together, these results show that when grown on permeable supports in the presence of steroids and air interface, the predominant channels expressed in ATII cells have single-channel characteristics resembling channels that are associated with the coexpression of the three cloned ENaC subunits alpha-, beta-, and gamma-ENaC.


Subject(s)
Pulmonary Alveoli/metabolism , Sodium Channels/metabolism , Animals , Cell Membrane/metabolism , Cells, Cultured , Culture Media/pharmacology , Cytological Techniques , Electrophysiology , Oxygen/pharmacology , Protein Isoforms/physiology , Pulmonary Alveoli/cytology , Rats , Rats, Sprague-Dawley , Sodium Channels/physiology
10.
J Biol Chem ; 276(16): 12903-10, 2001 Apr 20.
Article in English | MEDLINE | ID: mdl-11278712

ABSTRACT

Amiloride-sensitive epithelial Na(+) channels (ENaC) are responsible for trans-epithelial Na(+) transport in the kidney, lung, and colon. The channel consists of three subunits (alpha, beta, gamma) each containing a proline rich region (PPXY) in their carboxyl-terminal end. Mutations in this PPXY domain cause Liddle's syndrome, an autosomal dominant, salt-sensitive hypertension, by preventing the channel's interactions with the ubiquitin ligase Neural precursor cell-expressed developmentally down-regulated protein (Nedd4). It is postulated that this results in defective endocytosis and lysosomal degradation of ENaC leading to an increase in ENaC activity. To show the pathway that degrades ENaC in epithelial cells that express functioning ENaC channels, we used inhibitors of the proteosome and measured sodium channel activity. We found that the inhibitor, MG-132, increases amiloride-sensitive trans-epithelial current in Xenopus distal nephron A6 cells. There also is an increase of total cellular as well as membrane-associated ENaC subunit molecules by Western blotting. MG-132-treated cells also have increased channel density in patch clamp experiments. Inhibitors of lysosomal function did not reproduce these findings. Our results suggest that in native renal cells the proteosomal pathway is an important regulator of ENaC function.


Subject(s)
Cysteine Endopeptidases/metabolism , Multienzyme Complexes/metabolism , Sodium Channels/metabolism , Ubiquitins/metabolism , Amino Acid Sequence , Animals , Antibody Specificity , Cell Line , Cell Membrane/drug effects , Cell Membrane/physiology , Cysteine Proteinase Inhibitors/pharmacology , Epithelial Sodium Channels , Humans , Kinetics , Leupeptins/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Models, Biological , Molecular Sequence Data , Nephrons , Peptide Fragments/chemistry , Peptide Fragments/immunology , Proteasome Endopeptidase Complex , Protein Subunits , Sodium Channels/chemistry , Sodium Channels/genetics , Urothelium/cytology , Urothelium/physiology , Xenopus laevis
11.
J Biol Chem ; 276(10): 7136-42, 2001 Mar 09.
Article in English | MEDLINE | ID: mdl-11113145

ABSTRACT

To test the hypothesis that ATP activation of BK channels in GH(3) cells involves cytosolic phospholipase A(2) (cPLA(2)) as a potential protein target for phosphorylation, we first inhibited the activity of cPLA(2) by both pharmacologic and molecular biologic approaches. Both approaches resulted in a decrease rather than an increase in BK channel activity by ATP, suggesting that in the absence of cPLA(2), phosphorylation of other regulatory elements, possibly the BK channel protein itself, results in inactivation rather than activation of the channel. The absence of changes in activity in the presence of the non-substrate ATP analog 5'-adenylyl-beta,gamma-imidodiphosphate verified that ATP hydrolysis was required for channel activation by ATP. Experiments with an activator and inhibitor of protein kinase C (PKC) support the hypothesis that PKC can be involved in the activation of BK channels by ATP; and in the absence of PKC, other kinases appear to phosphorylate additional elements in the regulatory pathway that reduce channel activity. Our data point to cPLA(2)-alpha (but not cPLA(2)-gamma) as one target protein for phosphorylation that is intimately associated with the BK channel protein.


Subject(s)
Adenosine Triphosphate/metabolism , Aristolochic Acids , Cytosol/enzymology , Phospholipases A/metabolism , Phospholipases A/physiology , Potassium Channels, Calcium-Activated , Potassium Channels/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Adenylyl Imidodiphosphate/pharmacology , Animals , Blotting, Western , Cell Line , Cell Membrane/enzymology , Enzyme Inhibitors/pharmacology , Hydrolysis , Large-Conductance Calcium-Activated Potassium Channels , Oligonucleotides, Antisense/pharmacology , Patch-Clamp Techniques , Phenanthrenes/pharmacology , Phospholipases A/chemistry , Phospholipases A2 , Phosphorylation , Protein Isoforms , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Rats
12.
J Membr Biol ; 184(3): 313-9, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11891557

ABSTRACT

Aldosterone maintains total organism sodium balance in all higher vertebrates. The level of sodium reabsorption is primarily determined by the action of aldosterone on epithelial sodium channels (ENaC) in the distal nephron. Recent work shows that, in an aldosterone-sensitive renal cell line (A6), aldosterone regulates sodium reabsorption by short- and long-term processes. In the short term, aldosterone regulates sodium transport by inducing expression of the small G-protein, K-Ras2A, by stimulating the activity of methyl transferase and S-adenosyl-homocysteine hydrolase to activate Ras by methylation, and, possibly, by subsequent activation by K-Ras2A of phosphatidylinositol phosphate-5-kinase (PIP-5-K) and phosphatidylinositol-3-kinase (PI-3-K), which ultimately activates ENaC. In the long term, aldosterone regulates sodium transport by altering trafficking, assembly, and degradation of ENaC.


Subject(s)
Aldosterone/pharmacology , Epithelial Cells/metabolism , Sodium Channels/pharmacology , Sodium/pharmacokinetics , Aldosterone/metabolism , Aldosterone/physiology , Animals , Anura , Biological Transport/physiology , Cell Membrane/physiology , Electric Conductivity , Epithelial Cells/drug effects , Epithelial Sodium Channels , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Patch-Clamp Techniques/methods , Sodium Channels/pharmacokinetics , Sodium-Potassium-Exchanging ATPase/physiology
13.
DNA Seq ; 12(5-6): 425-9, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11913791

ABSTRACT

The cellular homolog of the oncogene v-src, the proto-oncogene c-src, was cloned from rat testis using a high stringency polymerase chain reaction. Rat c-src cDNA shared identity with chicken and mouse, and Rous sarcoma virus c-src and v-src, respectively. Rat c-Src protein was 98% homologous to both human and mouse c-Src. Interestingly, rat Src contained one extra amino acid compared to the mouse protein. As expected, the rat testis Src lacked the six extra residues common to the neuronal Src identified in human and mouse. Reporting of the cDNA sequence for non-neuronal, rat c-src should facilitate experimentation into cell growth and transformation using rat tissues as models of human disease.


Subject(s)
Genes, src , Testis/metabolism , Amino Acid Sequence , Animals , Base Sequence , Humans , Male , Mice , Molecular Sequence Data , Proto-Oncogene Mas , Rats , Sequence Alignment
14.
Am J Physiol Cell Physiol ; 279(4): C1211-9, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11003601

ABSTRACT

Ca(2+)-activated K(+) (BK) channels in GH(3) cells are activated by arachidonic acid (AA). Because cytosolic phospholipase A(2) can produce other unsaturated free fatty acids (FFA), we examined the effects of FFA on BK channels in excised patches. Control recordings were made at several holding potentials. The desired FFA was added to the bath solution, and the voltage paradigm was repeated. AA increased the activity of BK channels by 3.6 +/- 1.6-fold. The cis FFA, palmitoleic, oleic, linoleic, linolenic, eicosapentaenoic, and the triple bond analog of AA, eicosatetraynoic acid, all increased BK channel activity, whereas stearic (saturated) or the trans isomers elaidic, linolelaidic, and linolenelaidic had no effect. The cis unsaturated FFA shifted the open probability vs. voltage relationships to the left without a change in slope, suggesting no change in the sensitivity of the voltage sensor. Measurements of membrane fluidity showed no correlation between the change of membrane fluidity and the change in BK channel activation. In addition, AA effects on BK channels were unaffected in the presence of N-acetylcysteine. Arachidonyl-CoA, a membrane impermeable analog of AA, activates channels when applied to the cytosolic surface of excised patches, suggesting an effect of FFAs from the cytosolic surface of BK channels. Our data imply a direct interaction between cis FFA and the BK channel protein.


Subject(s)
Fatty Acids/metabolism , Neurosecretory Systems/metabolism , Pituitary Gland/metabolism , Potassium Channels, Calcium-Activated , Potassium Channels/metabolism , Acetylcysteine/pharmacology , Acyl Coenzyme A/metabolism , Acyl Coenzyme A/pharmacology , Animals , Arachidonic Acid/metabolism , Arachidonic Acid/pharmacology , Cell Line , Cell Membrane/metabolism , Fatty Acids/pharmacology , Fatty Acids, Unsaturated/metabolism , Fatty Acids, Unsaturated/pharmacology , Free Radical Scavengers/pharmacology , Large-Conductance Calcium-Activated Potassium Channels , Membrane Fluidity/drug effects , Membrane Potentials/drug effects , Neurosecretory Systems/cytology , Neurosecretory Systems/drug effects , Patch-Clamp Techniques , Pituitary Gland/cytology , Pituitary Gland/drug effects , Potassium Channels/drug effects , Rats , Reactive Oxygen Species/metabolism , Structure-Activity Relationship
15.
Am J Physiol Cell Physiol ; 279(1): C81-8, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10898719

ABSTRACT

Rapamycin and FK-506 are immunosuppressive drugs that bind a ubiquitous immunophilin, FKBP12, but immunosuppressive mechanisms and side effects appear to be different. Rapamycin binds renal FKBP12 to change renal transport. We used cell-attached patch clamp to examine rapamycin's effect on Na(+) channels in A6 cells. Channel NP(o) was 0.5 +/- 0.08 (n = 6) during the first 5 min but fell close to zero after 20 min. Application of 1 microM rapamycin reactivated Na(+) channels (NP(o) = 0.47 +/- 0.1; n=6), but 1 microM FK-506 did not. Also, GF-109203X, a protein kinase C (PKC) inhibitor, mimicked the rapamycin-induced reactivation in a nonadditive manner. However, rapamycin did not reactivate Na(+) channels if cells were exposed to 1 microM FK-506 before rapamycin. In PKC assays, rapamycin was as effective as the PKC inhibitor; however, epithelial Na(+) channel (ENaC) phosphorylation was low under baseline conditions and was not altered by PKC inhibitors or activators. These results suggest that rapamycin activates Na(+) channels by binding FKBP12 and inhibiting PKC, and, in renal cells, despite binding the same immunophilin, rapamycin and FK-506 activate different intracellular signaling pathways.


Subject(s)
Immunosuppressive Agents/pharmacology , Sirolimus/pharmacology , Sodium Channels/drug effects , Sodium Channels/metabolism , Drug Interactions , Enzyme Inhibitors/pharmacology , Epithelial Sodium Channels , Immunophilins/genetics , Indoles/pharmacology , Maleimides/pharmacology , Patch-Clamp Techniques , Phosphorylation/drug effects , Protein Kinase C/antagonists & inhibitors , Sodium Channels/physiology , Tacrolimus/pharmacology , Tacrolimus Binding Proteins , Tetradecanoylphorbol Acetate/pharmacology
16.
Am J Physiol Cell Physiol ; 279(2): C429-39, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10913010

ABSTRACT

Aldosterone increases Na(+) reabsorption by renal epithelial cells: the acute actions (<4 h) appear to be promoted by protein methylation. This paper describes the relationship between protein methylation and aldosterone's action and describes aldosterone-mediated targets for methylation in cultured renal cells (A6). Aldosterone increases protein methylation from 7.90 +/- 0.60 to 20.1 +/- 0.80 methyl ester cpm/microg protein. Aldosterone stimulates protein methylation by increasing methyltransferase activity from 14.0 +/- 0.64 in aldosterone-depleted cells to 31.8 +/- 2.60 methyl ester cpm/microg protein per hour in aldosterone-treated cells. Three known methyltransferase inhibitors reduce the aldosterone-induced increase in methyltransferase activity. One of these inhibitors, the isoprenyl-cysteine methyltransferase-specific inhibitor, S-trans, trans-farnesylthiosalicylic acid, completely blocks aldosterone-induced protein methylation and also aldosterone-induced short-circuit current. Aldosterone induces protein methylation in two molecular weight ranges: near 90 kDa and around 20 kDa. The lower molecular weight range is the weight of small G proteins, and aldosterone does increase both Ras protein 1.6-fold and Ras methylation almost 12-fold. Also, Ras antisense oligonucleotides reduce the activity of Na(+) channels by about fivefold. We conclude that 1) protein methylation is essential for aldosterone-induced increases in Na(+) transport; 2) one target for methylation is p21(ras); and 3) inhibition of Ras expression or Ras methylation inhibits Na(+) channel activity.


Subject(s)
Aldosterone/pharmacology , Epithelial Cells/drug effects , Protein Methyltransferases/drug effects , Sodium Channels/drug effects , ras Proteins/drug effects , Animals , Cells, Cultured , Epithelial Cells/metabolism , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Methylation , Protein Methyltransferases/metabolism , Sodium Channels/metabolism , ras Proteins/metabolism
17.
J Biol Chem ; 275(33): 25760-5, 2000 Aug 18.
Article in English | MEDLINE | ID: mdl-10829029

ABSTRACT

Regulation of epithelial Na(+) channel (ENaC) subunit levels by protein kinase C (PKC) was investigated in A6 cells. PKC activation altered ENaC subunit levels, differentially decreasing the levels of both beta and gamma, but not alphaENaC. Temporal regulation of beta and gammaENaC by PKC differed; gammaENaC decreased with a time constant of 3.7 +/- 1.0 h, whereas betaENaC decreased in 13.9 +/- 3. 0 h. Activation of PKC also resulted in a decrease in trans-epithelial Na(+) reabsorption for up to 48 h. PMA activation of PKC resulted in negative feedback inhibition of PKC protein levels beginning within 4 h. Both beta and gammaENaC levels, as well as transport tended toward pretreatment values after 48 h of PMA treatment. PKC inhibitors attenuated the effects of PMA on ENaC subunit levels and Na(+) transport. These results directly show for the first time that PKC differentially regulates ENaC subunit levels by decreasing the levels of beta and gamma but not alphaENaC protein. These results imply a PKC-dependent, long term decrease in Na(+) reabsorption.


Subject(s)
Protein Kinase C/metabolism , Sodium Channels/metabolism , Aldosterone/pharmacology , Animals , Biological Transport , Blotting, Western , Cell Line , Dose-Response Relationship, Drug , Electrophysiology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Epithelial Sodium Channels , Indoles/pharmacology , Kinetics , Maleimides/pharmacology , Naphthalenes/pharmacology , Protein Kinase C/chemistry , Protein Structure, Tertiary , RNA, Messenger/metabolism , Sodium/metabolism , Sodium Channels/chemistry , Tetradecanoylphorbol Acetate/pharmacology , Time Factors , Xenopus
18.
J Biol Chem ; 275(22): 16550-9, 2000 Jun 02.
Article in English | MEDLINE | ID: mdl-10747971

ABSTRACT

We used single channel methods on A6 renal cells to study the regulation by methylation reactions of epithelial sodium channels. 3-Deazaadenosine (3-DZA), a methyltransferase blocker, produced a 5-fold decrease in sodium transport and a 6-fold decrease in apical sodium channel activity by decreasing channel open probability (P(o)). 3-Deazaadenosine also blocked the increase in channel open probability associated with addition of aldosterone. Sodium channel activity in excised "inside-out" patches usually decreased within 1-2 min; in the presence of S-adenosyl-l-methionine (AdoMet), activity persisted for 5-8 min. Sodium channel mean time open (t(open)) before and after patch excision was higher in the presence of AdoMet than in untreated excised patches but less than t(open) in cell-attached patches. Sodium channel activity in excised patches exposed to both AdoMet and GTP usually remained stable for more than 10 min, and P(o) and the number of active channels per patch were close to values in cell-attached patches from untreated cells. These findings suggest that a methylation reaction contributes to the activity of epithelial sodium channels in A6 cells and is directed to some regulatory element closely connected with the channel, whose activity also depends on the presence of intracellular GTP.


Subject(s)
Sodium Channels/metabolism , Cell Line , Epithelial Cells/metabolism , Epithelial Sodium Channels , Guanosine Triphosphate/pharmacology , Methylation , Methyltransferases/pharmacology , Mineralocorticoid Receptor Antagonists/pharmacology , S-Adenosylmethionine/pharmacology , Tubercidin/pharmacology
19.
J Biol Chem ; 274(50): 35449-54, 1999 Dec 10.
Article in English | MEDLINE | ID: mdl-10585415

ABSTRACT

Xenopus laevis A6 cells were used as model epithelia to test the hypothesis that K-Ras2A is an aldosterone-induced protein necessary for steroid-regulated Na(+) transport. The possibility that increased K-Ras2A alone is sufficient to mimic aldosterone action on Na(+) transport also was tested. Aldosterone treatment increased K-Ras2A protein expression 2.8-fold within 4 h. Active Ras is membrane associated. After aldosterone treatment, 75% of K-Ras was localized to the plasma membrane compared with 25% in the absence of steroid. Aldosterone also increased the amount of active (phosphorylated) mitogen-activated protein kinase kinase likely through K-Ras2A signaling. Steroid-induced K-Ras2A protein levels and Na(+) transport were decreased with antisense K-ras2A oligonucleotides, showing that K-Ras2A is necessary for the natriferic actions of aldosterone. Aldosterone-induced Na(+) channel activity, was decreased from 0.40 to 0.09 by pretreatment with antisense ras oligonucleotide, implicating the luminal Na(+) channel as one final effector of Ras signaling. Overexpression of K-Ras2A increased Na(+) transport approximately 2.2-fold in the absence of aldosterone. These results suggest that aldosterone signals to the luminal Na(+) channel via multiple pathways and that K-Ras2A levels are limiting for a portion of the aldosterone-sensitive Na(+) transport.


Subject(s)
Aldosterone/pharmacology , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Oligodeoxyribonucleotides, Antisense/pharmacology , Sodium/metabolism , Animals , Biological Transport , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Epithelial Cells/physiology , Epithelial Sodium Channels , Gene Expression Regulation/drug effects , Kinetics , Models, Biological , Signal Transduction , Sodium Channels/metabolism , Xenopus laevis
20.
J Biol Chem ; 274(38): 26912-6, 1999 Sep 17.
Article in English | MEDLINE | ID: mdl-10480901

ABSTRACT

The Xenopus laevis distal tubule epithelial cell line A6 was used as a model epithelia to study the role of isoprenylcysteine-O-carboxyl methyltransferase (pcMTase) in aldosterone-mediated stimulation of Na(+) transport. Polyclonal antibodies raised against X. laevis pcMTase were immunoreactive with a 33-kDa protein in whole cell lysate. These antibodies were also reactive with a 33-kDa product from in vitro translation of the pcMTase cDNA. Aldosterone application increased pcMTase activity resulting in elevation of total protein methyl esterification in vivo, but pcMTase protein levels were not affected by steroid, suggesting that aldosterone increased activity independent of enzyme number. Inhibition of pcMTase resulted in a reduction of aldosterone-induced Na(+) transport demonstrating the necessity of pcMTase-mediated transmethylation for steroid induced Na(+) reabsorption. Transfection with an eukaryotic expression construct containing pcMTase cDNA increased pcMTase protein level and activity. This resulted in potentiation of the natriferic actions of aldosterone. However, overexpression did not change Na(+) reabsorption in the absence of steroid, suggesting that pcMTase activity is not limiting Na(+) transport in the absence of steroid, but that subsequent to aldosterone addition, pcMTase activity becomes limiting. These results suggest that a critical transmethylation is necessary for aldosterone-induction of Na(+) transport. It is likely that the protein catalyzing this methylation is isoprenylcysteine-O-carboxyl methyltransferase and that aldosterone activates pcMTase without affecting transferase expression.


Subject(s)
Aldosterone/metabolism , Protein Methyltransferases/metabolism , Sodium/metabolism , Sodium/pharmacokinetics , Absorption , Animals , Biological Transport, Active , Cells, Cultured , Kidney/metabolism , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL