Thyroid hormone stimulates Na-K-ATPase activity and its plasma membrane insertion in rat alveolar epithelial cells

Jianxun Lei, Sogol Nowbar, Cary N. Mariash, and David H. Ingbar

Department of Medicine, University of Minnesota, Minneapolis, Minnesota 55455

Submitted 4 November 2002 ; accepted in final form 4 April 2003


    ABSTRACT
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 DISCLOSURES
 REFERENCES
 
Na-K-ATPase protein is critical for maintaining cellular ion gradients and volume and for transepithelial ion transport in kidney and lung. Thyroid hormone, 3,3',5-triiodo-L-thyronine (T3), given for 2 days to adult rats, increases alveolar fluid resorption by 65%, but the mechanism is undefined. We tested the hypothesis that T3 stimulates Na-K-ATPase in adult rat alveolar epithelial cells (AEC), including primary rat alveolar type II (ATII) cells, and determined mechanisms of the T3 effect on the Na-KATPase enzyme using two adult rat AEC cell lines (MP48 and RLE-6TN). T3 at 10-8 and 10-5 M increased significantly hydrolytic activity of Na-K-ATPase in primary ATII cells and both AEC cell lines. The increased activity was dose dependent in the cell lines (10-9-10-4 M) and was detected within 30 min and peaked at 6 h. Maximal increases in Na-K-ATPase activity were twofold in MP48 and RLE-6TN cells at pharmacological T3 of 10-5 and 10-4 M, respectively, but increases were statistically significant at physiological T3 as low as 10-9 M. This effect was T3 specific, because reverse T3 (3,3',5'-triiodo-L-thyronine) at 10-9-10-4 M had no effect. The T3-induced increase in Na-K-ATPase hydrolytic activity was not blocked by actinomycin D. No significant change in mRNA and total cell protein levels of Na-K-ATPase were detected with 10-9-10-5 M T3 at 6 h. However, T3 increased cell surface expression of Na-K-ATPase {alpha}1- or {beta}1-subunit proteins by 1.7- and 2-fold, respectively, and increases in Na-K-ATPase activity and cell surface expression were abolished by brefeldin A. These data indicate that T3 specifically stimulates Na-K-ATPase activity in adult rat AEC. The upregulation involves translocation of Na-K-ATPase to plasma membrane, not increased gene transcription. These results suggest a novel nontranscriptional mechanism for regulation of Na-K-ATPase by thyroid hormone.

alveolar cell lines; protein translocation; sodium pump


THE ELECTROCHEMICAL GRADIENT across the membranes of most animal cells is maintained by Na-K-ATPase (52). It is an integral membrane protein consisting of at least two subunits, {alpha} and {beta}, each of which exists as one of several isoforms ({alpha}1, {alpha}2, {alpha}3, {alpha}4, {beta}1, {beta}2, and {beta}3) (21). The {alpha}-subunit contains the catalytic and regulatory phosphorylation sites and the binding sites for cation substrates and some protein kinases such as protein kinase C and phosphoinositide 3-kinase (21, 56). The role of the {beta}-subunit is less clear, but it is required for normal processing and plasma membrane expression of the enzyme and may have a role in regulating the interaction of cations with the {alpha}-subunit (3, 12). The {gamma}-subunit peptide, which can modify Na-K-ATPase function, was at first thought to represent a third component of Na-K-ATPase, but recent evidence suggests that it is not an integral part of the enzyme complex (41, 52, 53). The pump isoforms are expressed in a tissue- and development-specific fashion and are believed to be distinct in function and modes of regulation (5, 30). The {alpha}1-isoform is predominant in most epithelia, including rat lung (30), particularly in alveolar type II (ATII) cells.

Although much information about Na-K-ATPase is available, regulation of Na-K-ATPase and the underlying mechanisms are not completely understood (52). The many mechanisms for regulation of Na-K-ATPase activity in a tissue-specific manner have been reviewed (52). Long-term regulation of Na-K-ATPase activity usually involves changes in mRNA/protein synthesis or degradation of Na-K-ATPase subunit isoforms (52). Short-term rapid alternations in Na-K-ATPase activity may be mediated by changes in subcellular distribution of pump units (52), by reversible noncatalytic-site phosphorylation of the {alpha}1-subunit (21), or by changes in intracellular Na+ concentration. Many hormones regulate Na-K-ATPase. Insulin, {beta}-adrenergic agonists, and dopamine can rapidly modulate Na-K-ATPase activity (51, 52). In some studies, dopamine (46) or {beta}-adrenergic receptor stimulation by isoproterenol (4) or cAMP analogs (24) rapidly increased cell surface expression of Na-K-ATPase {alpha}-subunits in rat lung epithelia, whereas in rat proximal tubule cells, dopamine stimulated endocytosis of this enzyme from the plasma membrane (11, 56).

Thyroid hormones play a fundamental role in regulation of normal cell function and differentiation by interacting with intracellular thyroid hormone receptors and transcriptional coregulatory factors (coactivators and corepressors) (7, 54, 55). Thyroid hormones also may generate biological responses by nongenomic mechanisms that are independent of nuclear receptors for 3,3',5-triiodo-L-thyronine (T3) (13, 18, 55). Thyroid hormone stimulates Na-K-ATPase activity in responsive tissues and differentially regulates Na-K-ATPase isoforms (6, 28). T3 response elements exist in the 5'-flanking region of Na-K-ATPase {alpha}- and {beta}-subunits (20, 29). In nonlung tissues, the T3-induced increases in Na-K-ATPase activity generally are due to thyroid hormone-induced synthesis of Na-K-ATPase mRNA or protein (36-39). T3 regulates the activity and gene expression of Na-K-ATPase in tissue- and cell type-specific manners (17, 52). For example, T3 upregulates Na-K-ATPase activity in rat liver, skeletal muscle, and kidney (39) but inhibits synaptosomal Na-K-ATPase activity in rat cerebral cortex (48). Na-K-ATPase activity is stimulated by T3 in K562 human erythroleukemia cells (1), but not in human submandibular gland cells (34). T3 increases significantly Na-K-ATPase activity in liver, skeletal muscle, kidney, small intestine, cardiac muscle, and heart; however, the effect of T3 on Na-K-ATPase in the lung has not been determined.

The clearance of alveolar fluid is essential for recovery from adult lung injury and respiratory distress syndrome. T3 pretreatment stimulates alveolar epithelial fluid clearance by 65% in normal adult rat lung (22) and exerts a synergistic effect with dexamethasone in adult rat lung (22) and with hydrocortisone in the developing sheep lung on stimulation of alveolar fluid clearance (2). Although the cellular mechanism of T3 stimulation in fluid clearance is unknown, reabsorption of fluid out of the distal air space in the lung usually is driven by active Na+ transport (16, 40), and increased edema clearance usually requires higher Na-K-ATPase activity in alveolar epithelial cells (AEC) (40, 43). T3-induced Na-K-ATPase activity may contribute to the alveolar fluid clearance stimulated by T3 in adult rat lung. Therefore, the first objective of this study was to test the hypothesis that T3 stimulates Na-K-ATPase activity in adult rat lung epithelia, and if so, the second purpose was to explore possible mechanisms involved in this effect.


    MATERIALS AND METHODS
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 DISCLOSURES
 REFERENCES
 
Reagents. T3, 3,3',5'-triiodo-L-thyronine (reverse T3), actinomycin D, brefeldin A, and antibody against actin (A-2066) were purchased from Sigma Chemical. A mouse cDNA probe for 18S rRNA was purchased from Ambion. Cell culture reagents, Waymouth's MB752/1 medium, DMEM-Ham's F-12 medium, HEPES, fetal bovine serum (FBS), and antibiotics were purchased from GIBCO BRL, Life Technologies. Bicinchoninic acid (BCA) protein assay kit was obtained from Pierce. Stripped FBS (without endogenous T3) was prepared by mixing serum with a resin slurry [5 g AG 1-X8 resin (Bio-Rad)/100 ml serum] as described elsewhere (47). Removal of endogenous T3 by this method was confirmed by using tracer amounts of radioisotope-labeled T3.

Isolation and culture of rat AEC. ATII cells were isolated from pathogen-free adult male Sprague-Dawley rats (190-220 g) as we previously described (32). Use of the animals complied with guidelines for their ethical treatment, and the studies were approved by the Institutional Animal Care Committee. ATII cells were grown in DMEM-Ham's F-12 medium with 10% FBS and antibiotics (50 U/ml penicillin and 50 U/ml streptomycin) overnight. The cells were cultured in DMEM-Ham's F-12 medium with 5% stripped FBS for 24 h before T3 treatment.

Cell culture. The rat ATII cell line RLE-6TN (15) was purchased from American Type Culture Collection, and the MP48 cell line (44) was a gift from G. Hunninghake (University of Iowa). Both cell lines exhibit characteristics of ATII cells, such as lipid-containing inclusion bodies. RLE-6TN cells are derived from male Fischer 344 rats after simian virus 40-T antigen transformation. They stain positive for cytokeratins and lipids (phosphine 3R) and have alkaline phosphatase activity but do not express T antigen. MP48 cells are adenoviral 12S E1A immortalized diploid adult rat type II cells that resemble type II cells in situ, contain lamellar bodies, bind the lectin Maclura pomifera I, and stably express cytokeratins and the E1A gene product. MP48 cells were maintained in Waymouth's MB752/1 medium. RLE-6TN cells were grown in DMEM-Ham's F-12 medium. Both media were supplemented with 10% FBS, 40 mM HEPES, and antibiotics (100 U/ml penicillin, 100 µg/ml streptomycin, and 0.25 µg/ml amphotericin B). Antibiotics, including amphotericin, were included in the standard culture media for the cell lines and during T3 exposure but were not present during the Na-K-ATPase activity assay. Removal of amphotericin did not alter Na-K-ATPase activity in MP48 cells.

Experimental design. To measure the effect of T3 on the hydrolytic activity of Na-K-ATPase, the cells were grown to ~80% confluence in the appropriate medium containing 10% FBS. In most, but not all, experiments, the cells were T3 starved for 24 h; i.e., cells were cultured in appropriate medium supplemented with stripped 10% FBS; then the monolayers were exposed to T3 or inhibitors for various time intervals in appropriate medium plus 1% stripped FBS. At the indicated times, the cell monolayers were washed twice with ice-cold phosphate-buffered saline (PBS) to terminate exposure to all agents. In some experiments, cells were not preexposed to stripped serum, but after culture with 10% FBS, T3 was added to the medium with 1 or 10% FBS, and Na+ pump activity was measured. For inhibition studies, cells were preincubated with inhibitors for 30 min at 37°C and then treated with T3 in the continued presence of inhibitors for various time intervals.

Preparation of crude AEC membranes. Crude cell membranes were prepared as previously described (51) with slight modification. Briefly, the cells were scraped in ice-cold PBS and centrifuged at 5,000 rpm at 4°C for 5 min. The cell pellets were resuspended in 500 µl of ATPase assay buffer containing 37.5 mM imidazole, 137.5 mM NaCl, 18.75 mM KCl, 6.25 mM sodium azide, 0.625 mM EGTA (pH 7.0), 5 mM MgCl2, and 0.005% SDS for MP48 cells and primary ATII cells and 0.01% SDS for RLE-6TN cells. The cells were sonicated on ice for 10 s with Vibra Cell (Sonics and Materials, Danbury, CT) at 45% duty cycle and 4.5 output control on ice and centrifuged at 10,000 rpm at 4°C for 10 min. The sediment was resuspended by sonication for 8 s in 350-500 µl of the ATPase assay buffer described above. Aliquots of crude cell membrane preparations were used for assay of protein contents by using the BCA protein assay kit (Pierce, Rockford, IL) and Na-K-ATPase activity. Na-K-ATPase activity assays were performed immediately after isolation of crude cell membranes.

Na-K-ATPase activity assay. The hydrolytic activity of Na-K-ATPase was measured as ouabain-sensitive ATP hydrolysis under maximal velocity conditions by measuring the release of inorganic phosphate (Pi) from ATP, as previously described (32, 37) with slight modification. Briefly, equal amounts of crude cell membrane (20, 30, and 50 µg for RLE-6TN, MP48, and primary ATII cells, respectively) for all treatments in an experiment were added to 1 ml of ATPase assay buffer with or without 10 mM ouabain. The tubes were incubated for 18 min at 37°C after the reaction was started by the addition of 10 µl of 0.439 M ATP (Sigma Chemical). The reaction was terminated by addition of 50 µl of 100% ice-cold trichloroacetic acid, and Pi was determined by the method of Fiske and Subbarow as described elsewhere (37). Na-K-ATPase specific activity was taken as the difference in Pi concentration per milligram of protein per minute in the absence and presence of 10 mM ouabain. Results were compared with controls and expressed as percent increase or inhibition of control Na-K-ATPase activity.

RNA isolation and Northern analysis. Total cellular RNA was isolated using TRI Reagent (Molecular Research Center, Cincinnati, OH) following the manufacturer's instructions. RNA was isolated, and Northern analysis was performed as previously described (30). The purity and concentration of RNA were assessed from the ratio of absorbance at 260 nm to that at 280 nm. After ultraviolet irradiation using UV Stratalinker 1800 (Stratagene), prehybridization, and hybridization were performed using the High-Efficiency Hybridization System (Molecular Research Center) following the instructions of the manufacturer. Transcripts were visualized with standard autoradiography or phosphorimaging (Bio-Rad) and quantitated. The integrated optical density of the RNA bands was determined with Densitometry Image software (Molecular Analyst). All RNA densitometry values were normalized to 18S rRNA. The experiments were performed at least in triplicate.

Western blot. Protein samples were incubated in boiling water for 10 min in loading buffer containing 2% SDS, 50 mM Tris · HCl (pH 7.5), 10% glycerol, 2% {beta}-mercaptoethanol, and 0.001% bromphenol blue. For the {beta}1-subunit, the concentration of SDS and {beta}-mercaptoethanol was 5.6 and 6%, respectively. Proteins were then separated on a 7.5 or 10% denaturing SDS-polyacrylamide gel for Na-K-ATPase {alpha}1- and {beta}1-subunits, respectively. Separated proteins were transferred to a polyvinylidene difluoride membrane (catalog no. IPVH00010, Millipore, Bedford, MA), which was then hybridized with primary mouse antibodies against the Na-KATPase {alpha}1-subunit (catalog no. 05-369, Upstate Biotechnology) or {beta}1-subunit (catalog no. 05-382, Upstate Biotechnology) diluted 1:2,000. After incubation with a secondary peroxidase-conjugated goat anti-mouse antibody diluted 1:2,500 (Sigma Chemical), the membranes were washed and the proteins were detected with the ECL system (Amersham, Piscataway, NJ). The chemiluminescence of each lane was quantified by densitometry in arbitrary units using a Bio-Rad Image Analysis system (Molecular Analyst).

Measurement of total cell and plasma membrane Na-KATPase protein. For measurement of total cellular Na-KATPase subunit proteins, the cells were washed twice with ice-cold 1x PBS, scraped off plates, and collected in ice-cold PBS. The cells were pelleted with centrifugation at 5,000 rpm at 4°C for 5 min. To lyse the pellets, we drew them 10 times through a syringe and 25-gauge needle in 400 µl of lysis buffer containing 50 mM Tris · HCl (pH 7.5), 150 mM NaCl, 2 mM EDTA, and 1% (vol/vol) Nonidet P-40, with protease inhibitors (1 mM phenylmethylsulfonyl fluoride, 2 µg/ml pepstatin, and 10 µg/ml each aprotinin and leupeptin). Protein concentrations were determined using the BCA protein assay kit. Equal amounts of protein were subjected to Western blot analysis as described above. The densitometry values of Na-K-ATPase subunit proteins were normalized to actin protein.

For determination of plasma membrane Na-K-ATPase, the cell surface proteins were biotinylated as described previously (24, 25) with minor modification. Cells were grown on 24-mm Transwell porous cell culture inserts (0.4-µm pore; Nucleopore polycarbonate membrane, catalog no. 3412, Costar, Cambridge, MA). T3 and/or inhibitors were added to the apical and basolateral sides of the filter for indicated time periods. The cells were incubated in PBS containing 2 mM EDTA at 4°C for 5 min after being washed twice with ice-cold 1x PBS. Then cell surface proteins were biotinylated by addition of biotinylation buffer (10 mM triethanolamine, pH 7.5, 1 mM EDTA, and 150 mM NaCl) containing 1 mg/ml water-soluble, cleavable sulfobiotin-X-NHS (Calbiochem, San Diego, CA) to the apical and basolateral sides of the filter for 30 min at 4°C. The free unreacted biotin was quenched by three washes with 1x PBS containing 100 mM glycine. Cells were then lysed in lysis buffer, and protein content was determined by using the BCA protein assay kit. Equal amounts of total cell protein were precipitated with streptavidin-agarose beads (Sigma Chemical) diluted in lysis buffer by incubation overnight at 4°C. The beads were then washed three times with lysis buffer, twice with high-salt buffer (5 mM EDTA, 50 mM Tris · HCl, pH 7.4, and 500 mM NaCl), and once with 10 mM Tris · HCl (pH 7.4). Proteins were eluted from the beads by incubation of the biotinylated protein-streptavidin-agarose beads for 10 min in 50 µl of SDS-containing buffer (5.6% SDS, 240 mM Tris · HCl, pH 7.5, 6% {beta}-mercaptoethanol, 16% glycerol, and 0.008% bromphenol blue) and analyzed by Western blotting. The higher SDS concentration disrupts the avidin-biotin complexes. The amount of protein was expressed as densitometry in arbitrary units.

Statistics. Values are means ± SD of a minimum of three experiments. Comparisons involving three or more groups were analyzed by ANOVA and post hoc pairwise comparisons. Differences between means were considered significant at P < 0.05, adjusted for the number of comparisons.


    RESULTS
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 DISCLOSURES
 REFERENCES
 
T3 augments alveolar epithelial Na-K-ATPase enzyme activity. Na-K-ATPase is required for active Na+ transport across the alveolar epithelia (43). To understand the mechanism of T3 stimulation of alveolar fluid clearance in adult rat lung and identify the effect of T3 on Na-K-ATPase activity in adult lung epithelial cells, we used two adult rat AEC lines, MP48 and RLE-6TN, and adult rat primary ATII cells to investigate the effects of T3 on the ATP hydrolytic activity of Na-KATPase from physiological to pharmacological concentrations (10-9-10-4 M). We also assessed whether different cell culture conditions affected the response of Na-K-ATPase activity to T3.

MP48 cells were cultured for 24 h in media with 10% stripped FBS and then exposed to T3. T3 increased the enzyme activity in a dose-dependent fashion when presented in 1% stripped FBS or 1% FBS culture conditions (Fig. 1A); activity increased by >40% (P < 0.05) at concentrations as low as 10-9 M at 6 h of incubation. The T3-induced maximal increase was ~2.2-fold at 10-5 M in the presence of stripped 1% FBS medium. The magnitude of the increase in Na-K-ATPase activity at every T3 dose in 1% FBS was somewhat less than that in 1% stripped FBS. Smaller increases in Na-K-ATPase activity occurred when T3 was presented in 10% FBS (data not shown).



View larger version (18K):
[in this window]
[in a new window]
 
Fig. 1. 3,3',5-Triiodo-L-thyronine (T3) increases Na-K-ATPase hydrolytic activity in a dose-dependent manner in alveolar type II (ATII) cells. Ouabain-sensitive hydrolytic activity of Na-K-ATPase was measured after permeabilization of cell membrane. *P < 0.05; **P < 0.01 vs. control. A: MP48 cells were grown to ~80% confluence in Waymouth's medium containing 10% FBS. Open bars, cells cultured in medium supplemented with 10% stripped FBS for 24 h and then exposed to T3 for 6 h at 37°C in medium with 1% stripped FBS (ST); solid bars, cells cultured to confluence in Waymouth's medium containing 10% FBS and then exposed to T3 for 6 h at 37°C in medium with 1% FBS. Values are means ± SD of 5 independent experiments. B: RLE-6TN cells were cultured to confluence in DMEM-Ham's F-12 medium with 10% FBS. Cells were then exposed to T3 or 3,3',5'-triiodo-L-thyronine [reverse T3 (RT3)] for 3 (open bars) or 6 h (solid bars) in 10% FBS, respectively. Values are means ± SD of 5 independent experiments. C: primary ATII cells were T3 starved for 24 h by culture in DMEM-Ham's F-12 medium with 5% stripped FBS. Then cells were incubated with 10-8 M T3 for 6 h or 10-5 M T3 for 3 h in 1% stripped FBS with DMEM. Values are means ± SD of 3 independent experiments.

 

Similarly, in RLE-6TN cells, T3 increased Na-KATPase activity at 3 and 6 h and when presented in 1% stripped or 10% FBS (Fig. 1B and data not shown). At 3 h, T3 concentrations as low as 10-8 M increased Na+ pump activity in a dose-dependent fashion. The magnitude of the increase was greater at supraphysiological T3 levels but was statistically significant (30% increase, P < 0.05) at 10-8 M. In contrast to T3, 6 h of exposure to the metabolically inactive analog of T3 (50), reverse T3, at the same concentrations had no effect on Na-K-ATPase activity, indicating that the effect of T3 is specific.

Because the cell lines may not reflect the response of the primary rat ATII cells, we also assessed the effect of T3 on Na-K-ATPase activity in primary ATII isolates at 3 days of culture. The cells were T3 starved by exposure to 5% stripped FBS for 24 h before addition of T3 in 1% stripped FBS. T3 increased the Na-K-ATPase activity at 6 h of 10-8 MT3 by 131.7% and at 3 h of 10-5 M T3 by 149.3% (Fig. 1C). Because the responses to T3 were similar to those of AEC, complete dose-response curves were not determined.

The time course of the T3 effect on Na-K-ATPase activity in MP48 cells is shown in Fig. 2. Surprisingly, the T3-induced increase in Na-K-ATPase activity occurred rapidly at 10-5 or 10-6 M T3 in 1% stripped FBS. The increase in Na-K-ATPase activity was significant at 1 h of T3 exposure and peaked at 6 h (Fig. 2A). Pharmacological levels of T3 (10-4 M) also rapidly increased Na-K-ATPase activity in RLE-6TN cells in 10% FBS (Fig. 2B), with the maximal effect at 6 h. The rapid T3-induced increase in Na-K-ATPase activity suggested that the T3-induced increase may not require transcription or translation of Na-K-ATPase.



View larger version (20K):
[in this window]
[in a new window]
 
Fig. 2. Time course of effect of T3 on Na-K-ATPase hydrolytic activity in MP48 and RLE-6TN cells. Values are means ± SD of 4 independent experiments. *P < 0.05; **P < 0.01 vs. control. A: MP48 cells were exposed to 10-6 or 10-5 M T3 in medium with 1% stripped FBS after cells were T3 starved for 24 h by culture in 10% stripped FBS. B: RLE-6TN cells that were cultured in 10% FBS were incubated with 10-4 M T3 in medium supplied with 10% FBS.

 

T3 does not alter expression of Na-K-ATPase {alpha}1- and {beta}1-subunit mRNA and total cellular protein. The principal mechanism for action of T3 on Na-K-ATPase usually is transcriptional (7, 55), but in our study, the rapid increase in Na-K-ATPase activity observed within 0.5-1 h of exposure suggested that T3 stimulation was not transcriptional. To test this hypothesis, the change of Na-K-ATPase mRNA and protein levels was determined after cells were subjected to T3 treatments for various periods. Although the ATII cells expressed some quantities of the Na-K-ATPase {alpha}1-, {alpha}2-, {alpha}3-, {beta}1-, and {beta}3-isoforms, the predominant Na-K-ATPase isoforms that we detected in adult rat primary ATII cells and in the cell lines are {alpha}1, {beta}1, and {beta}3 (data not shown). Therefore, we investigated the effect of T3 on {alpha}1- and {beta}1-subunit mRNA and proteins to identify whether T3 affects the Na-K-ATPase activity in a fast manner by regulating the gene expression of {alpha}1- and {beta}1-subunits in adult alveolar cells.

In MP48 cells, T3 did not significantly alter the steady-state contents of Na-K-ATPase {alpha}1- and {beta}1-subunit mRNA at 6 h of exposure (P > 0.05; Fig. 3). The Northern blot for the {beta}1-subunit mRNA revealed two bands just below the 18S rRNA marker, with the signal much stronger for the lower band.



View larger version (53K):
[in this window]
[in a new window]
 
Fig. 3. T3 does not change steady-state mRNA levels of Na-K-ATPase {alpha}1- and {beta}1-subunits in MP48 cells. MP48 cells were cultured as described in Fig. 1. Cells were incubated with T3 for 6 h in 1% stripped FBS medium. Total RNA was isolated, and Northern blot analysis was performed with 10-20 µg of total RNA and 32P-labeled full-length rat cDNA probes for Na-K-ATPase {alpha}1- and {beta}1-subunit mRNA and mouse cDNA probes for 18S rRNA. RNA bands were visualized with standard autoradiography. Integrated optical density of RNA bands was determined with Densitometry Image software (Molecular Analyst). All RNA densitometric values were normalized to 18S rRNA. A and B: representative Northern blots for Na-K-ATPase {alpha}1- and {beta}1-subunit mRNA. Lane 1, control (untreated sample); lanes 2, 3, 4, 5, and 6, treatment with 10-9, 10-8, 10-7, 10-6, and 10-5 M T3, respectively. {beta}1 Northern blot revealed two bands, with the lower band having sufficiently greater intensity; the 2 bands are difficult to distinguish in this image. C: quantitation of effect of T3 on mRNA levels of Na-K-ATPase {alpha}1- and {beta}1-subunits at 6 h. Values are means ± SD of 4 independent experiments.

 

T3 also did not significantly change the protein levels of {alpha}1- and {beta}1-subunits at 10-9-10-5 M T3 at 6 h of exposure (Fig. 4, A and B) and at 10-5 M at 1-6 h of exposure (Fig. 4, C and D).



View larger version (54K):
[in this window]
[in a new window]
 
Fig. 4. T3 does not alter the quantity of total cell Na-K-ATPase {alpha}1- and {beta}1-subunit proteins in MP48 cells. MP48 cells were cultured as described in Fig. 1. Cells were exposed to T3 for 1 or 6 h in medium with 1% stripped FBS at 37°C. Total cell protein was collected and analyzed by Western blotting. Na-KATPase {alpha}1- and {beta}1-subunits and actin were detected by specific monoclonal antibodies against {alpha}1- and {beta}1-subunits and actin. All {alpha}1- and {beta}1-subunit protein densitometric values were normalized to actin protein. A: representative Western blot. MP48 cells were exposed to T3 for 6 h. B: densitometric values. Values are means ± SD from 4 different experiments. C: representative Western blot of Na-K-ATPase proteins from MP48 cells exposed to 10-5 M T3 for 1 and 6 h. D: densitometric values. Values are means ± SD from 6 and 9 different experiments for 1 and 6 h of exposure, respectively.

 

In RLE-6TN cells, Northern blot quantification of the steady-state mRNA levels of the {alpha}1-, {alpha}2-, {alpha}3-, {beta}1-, and {beta}3-subunits was performed to determine whether T3 induced a shift in Na-K-ATPase isoform expression. At 6 h of T3 exposure, there was no significant change in {alpha}1- and {beta}1-subunit levels in each of three experiments (data not shown). At 24 h, 10-8-10-6 M T3 did not increase the levels of any of the subunits (n = 3, mean change <10% for each subunit at 10-4 M T3). High T3 concentrations (10-5 M) led to small increases in the mRNA {alpha}1-subunit (mean 30% increase, n = 3) but not of other mRNA subunit levels during 24 h of exposure. Thus T3 did not increase the total cellular mRNA and protein levels of Na-K-ATPase subunits during the period with significantly increased Na-KATPase activity. These data suggested that the T3-induced increase in Na-K-ATPase activity was independent of transcription.

Actinomycin D does not block the T3-induced increase in Na-K-ATPase activity. Because the principal action of T3 is transcription and T3 could affect transcription of other Na-K-ATPase genes beyond the {alpha}1- and {beta}1-subunits, we wished to test more directly whether transcription was required for T3 stimulation of Na-KATPase. The general inhibitor of transcription, actinomycin D, was used to determine the role of transcription in T3 stimulation. MP48 cells were T3 starved for 24 h in 10% stripped FBS medium, and then cells were exposed to 10-6 M T3 with or without 10 µg/ml actinomycin D for 6 h in 1% stripped FBS. T3 stimulation of Na-K-ATPase activity was not diminished by actinomycin D (Fig. 5A). Similarly, actinomycin D did not inhibit T3 stimulation in MP48 cells at 3 or 6 h in the presence of 1% FBS (data not shown). Similarly, in RLE-6TN cells, actinomycin D did not inhibit T3 stimulation of Na+ pump activity at 3 or 6 h, even at very high T3 concentrations (10-4 M; Fig. 5B). Actinomycin D alone had no significant effect on Na-K-ATPase activity in MP48 and RLE-6TN cells. It is not surprising that actinomycin D also had little inhibitory effect on Na-K-ATPase, because the half-life of the Na+ pump protein is long in primary ATII cells and Madin-Darby canine kidney cells (unpublished data). Thus these data strongly support the concept that T3 stimulation of Na-K-ATPase activity is principally through a nontranscriptional mechanism.



View larger version (20K):
[in this window]
[in a new window]
 
Fig. 5. Actinomycin D (ActD) does not block T3-induced Na-K-ATPase activity in MP48 (A) or RLE-6TN (B) cells. Values are means ± SD of 4 independent experiments. *P < 0.05; **P < 0.01 vs. control. A: MP48 cells were incubated for 6 h in the presence or absence of 10-6 M T3 and 10 µg/ml actinomycin D in medium with 1% stripped FBS. B: RLE-6TN cells were maintained for 3 or 6 h in the presence or absence of 10-4 M T3 and 10 µg/ml actinomycin D in medium with 10% FBS.

 

T3 increases Na-K-ATPase subunit protein levels in plasma membrane of MP48 cells. The short-term posttranscriptional regulation of Na-K-ATPase activity by hormones may involve direct effects on the kinetic behavior of the enzyme or translocation of the enzyme between the plasma membrane and intracellular stores (52). On the basis of the minimal effect of T3 on mRNA and protein expression of Na-K-ATPase subunits in MP48 and RLE-6TN cells, we hypothesized that T3 stimulates rapid recruitment of Na-K-ATPase proteins to the plasma membrane in MP48 and RLE-6TN cells. To investigate the effect of T3 on cell surface expression of Na-K-ATPase, intact cells treated with T3 were reacted with a membrane-impermeable biotinylation reagent (sulfobiotin-X-NHS, water soluble, cleavable) that labels proteins exposed on the cell surface but not proteins in intracellular membranes. T3 increased the amount of Na-K-ATPase {alpha}1- and {beta}1-subunit proteins at the cell surface in MP48 cells (Fig. 6). T3 at 10-5 M stimulated a significant increase in {alpha}1-and {beta}1-subunit proteins at the cell surface after 6 h of T3 exposure; the maximal increase was 1.7-fold for the {alpha}1-subunit and 2-fold for the {beta}1-subunit. Significant augmentation (P < 0.05) of {alpha}1-subunit (133.9% of control, n = 4) and {beta}1-subunit (131.5% of control, n = 3) proteins at the plasma membrane also was induced at 1 h of T3 exposure (Fig. 6). T3 did not significantly change the total cell protein level of the Na-K-ATPase {alpha}1-subunit at 6 h of T3 treatment (Fig. 4); however, it markedly increased the amount of Na-K-ATPase {alpha}1-and {beta}1-subunit protein at the cell surface. Taken together with the prior findings, these data suggest that the T3-induced increase in Na-K-ATPase activity and protein at the membrane is due to increased plasma membrane recruitment of this enzyme and not increased de novo protein synthesis.



View larger version (25K):
[in this window]
[in a new window]
 
Fig. 6. T3 increases protein amount of Na-KATPase {alpha}1- and {beta}1-subunits at plasma membrane in MP48 cells. Cells were cultured as described in Fig. 1. Cells were exposed to 10-5 M T3 for 1 and 6 h in medium with 1% stripped FBS at 37°C, respectively. Cell surface proteins were biotinylated and lysed. Labeled proteins were immunoprecipitated by streptavidin-agarose beads. Na-K-ATPase {alpha}1- and {beta}1-subunits were detected by Western blotting using specific monoclonal antibodies against {alpha}1- and {beta}1-subunits. Values are means ± SD of 4 independent experiments. *P < 0.05; **P < 0.01 vs. control. A and C: representative Western blots for Na-KATPase {alpha}1- and {beta}1-subunits, respectively. B and D: densitometric values (means ± SD) from 4 ({alpha}1, 1 h), 8 ({alpha}1, 6 h), 3 ({beta}1, 1 h), and 6 ({beta}1, 6 h) different experiments.

 

Brefeldin A decreases T3-induced plasma membrane expression and activity of Na-K-ATPase. Brefeldin A disassembles and redistributes the Golgi complex into the endoplasmic reticulum within minutes of application (8) to disrupt cellular protein trafficking. Brefeldin A inhibits protein transport at various concentrations. Brefeldin A at 1-10 µg/ml completely inhibited protein secretion in cultured rat hepatocytes (42), and at 20 µg/ml, it prevented the dibutyryl-cAMP-dependent increase in cell surface expression and activity of Na-KATPase in renal epithelia (24). The effect of 10 µg/ml brefeldin A on the T3-induced increase in protein content of Na-K-ATPase in the plasma membrane was determined. Figure 7, A and B, shows that brefeldin A alone did not significantly modify the basal quantity of cell surface Na-K-ATPase {alpha}1- and {beta}1-subunit proteins but completely abolished the T3-induced increment in plasma membrane expression of Na-K-ATPase {alpha}1- and {beta}1-subunits in MP48 cells during 6 h of exposure. Thus T3 increased cell surface expression through a brefeldin A-dependent process in MP48 cells, suggesting that T3 stimulation of Na-K-ATPase trafficking is related to the Golgi complex. As expected, 10 µg/µl brefeldin A also completely blocked the T3-induced increment in Na-K-ATPase activity but did not alter basal Na-KATPase activity in MP48 cells (Fig. 7C). Abolition of the T3-induced plasma membrane expression and activity of Na-K-ATPase by brefeldin A established that T3-dependent translocation of Na-K-ATPase is responsible for upregulation of the enzyme activity.



View larger version (25K):
[in this window]
[in a new window]
 
Fig. 7. Brefeldin A eliminates T3-induced cell surface expression of Na-K-ATPase {alpha}1-subunit protein (A and B) and T3-induced activity (C) of Na-K-ATPase in MP48 cells. MP48 cells were incubated for 6 h in the presence or absence of 10-5 M T3 and 10 µg/µl brefeldin A in 1% stripped FBS medium. Protein on cell surface was measured as described in Fig. 6 legend, and activity of Na-K-ATPase was detected as described in Fig. 1 legend. **P < 0.01 vs. control. A: representative Western blots. B: densitometric values (means ± SD) from 3 different experiments. C: Na-K-ATPase activity data (means ± SD) from 5 different experiments.

 


    DISCUSSION
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 DISCLOSURES
 REFERENCES
 
Alveolar fluid clearance is critical for oxygenation and for recovery from alveolar flooding. Active lung fluid resorption requires alveolar epithelial Na-KATPase. In a recent study, T3 administered parenterally for 48 h stimulated alveolar fluid clearance in rat lungs in vivo (22). The present study establishes that T3 acts directly on AEC using specific and rapid pathways to increase Na-K-ATPase activity, primarily through stimulation of translocation of Na+ pump {alpha}1-and {beta}1-subunit protein to the plasma membrane, rather than through increased gene transcription.

T3 specifically stimulates Na-K-ATPase activity in primary ATII cells and two alveolar cell lines. This finding is important for several reasons. Although Na-K-ATPase is a ubiquitous and essential cell enzyme that has been studied intensively, tissue-specific regulation of Na-K-ATPase and the underlying mechanisms are not completely understood (52). Specifically, little is known about the T3 effect on Na-K-ATPase function in the lung. Because Na-K-ATPase is necessary for active Na+ and fluid transport across the alveolar epithelia (43) and T3 stimulates the alveolar fluid in adult rat lung (22), we hypothesized that T3 would stimulate AEC Na-K-ATPase. However, no prior studies have investigated whether the thyroid hormone activates AEC Na-K-ATPase. In the present study, we investigated T3 effects on Na-K-ATPase in primary ATII cells and two adult rat AEC lines, MP48 and RLE-6TN. We demonstrated that T3 stimulated alveolar epithelial Na-K-ATPase activity in a dose- and time-dependent fashion, as T3 does in rat liver cells (27). The inactive isomer reverse T3 had no significant effect on Na-K-ATPase activity in RLE-6TN cells, indicating that the T3 effect is specific.

In contrast to many other tissues or cell types, the stimulatory effect of T3 on Na-K-ATPase activity in AEC is not derived from transcriptional augmentation. Our data show that T3 stimulation of Na-K-ATPase activity was detected within 60 min in MP48 cells and within 30 min in RLE-6TN cells, with maximal stimulation at 6 h. Similarly, T3 rapidly activates Na-KATPase in liver of the Anabas fish (49). The mechanism underlying the short-term effect of T3 on Na-K-ATPase remains to be clarified. T3 is a well-known transcriptional regulator for Na-K-ATPase genes in an isoformspecific fashion (17, 29). There are positive thyroid hormone response elements in the 5'-flanking region of the Na-K-ATPase {beta}1-subunit (20) and negative thyroid hormone response elements in the 5'-flanking region of Na-K-ATPase {alpha}2- and {alpha}3-subunits (29). T3 stimulates Na-K-ATPase activity and isoform mRNA levels in various mammalian tissues (17, 23, 29). However, in our AEC, T3 did not change the steady-state levels of {alpha}1- and {beta}1-subunit mRNAs (Fig. 3). Moreover, the T3-induced Na-K-ATPase activity was not sensitive to a general inhibitor of gene transcription, actinomycin D. The failure of actinomycin D to block the T3-induced Na-K-ATPase activity directly demonstrated that T3 stimulated the Na-K-ATPase activity in a transcription-independent manner. Virtually all thyroid hormone effects previously were believed to be transcriptional; however, nongenomic effects of thyroid hormone are gaining some recognition (13, 18, 55).

The mechanisms by which thyroid hormone regulates Na-K-ATPase vary among tissues. T3 increases the protein level of Na-K-ATPase in kidney, heart, and skeletal muscle tissues (17); however, the effects of T3 on lung Na-K-ATPase subunit protein have not been reported. Our data indicate that the T3-induced increase in Na-K-ATPase activity was not accompanied by an increase in total cell Na-K-ATPase {alpha}1- and {beta}1-subunit protein, suggesting that T3 also does not affect translation of Na-K-ATPase mRNA at 6 h. However, the detailed effects of T3 on translation of Na-KATPase subunits and rate of translation need further investigation.

T3 increased plasma membrane Na-K-ATPase enzyme in AEC, likely because of stimulation of translocation. T3 previously has been reported to affect sorting and trafficking of proteins. T3 stimulates the translocation of Trip230, a coactivator of thyroid hormone receptor, from the Golgi complex to the nucleus (10), of choline phosphotransferase from cytosol to mitochondria (9), and of type II iodothyronine 5'-deiodinase from plasma membrane to endosomes (19). In our study, T3 did not change the total cell content of Na-K-ATPase {alpha}1- and {beta}1-subunit proteins but increased the plasma membrane expression. The T3-induced increase of Na-K-ATPase protein at the cell surface was abolished by brefeldin A, a potent inhibitor of translocation that disassembles the Golgi complex and redistributes it into the endoplasmic reticulum (10). Because brefeldin A also may alter recycling of plasma membrane proteins, these data strongly suggest, but do not prove, that T3 stimulates delivery of Na-KATPase to the cell surface via the endoplasmic reticulum-Golgi complex constitutive pathway in AEC. It is somewhat surprising that brefeldin A did not affect basal Na-K-ATPase activity or membrane protein quantity. The effects of T3 on internalization of Na-KATPase and the mechanism(s) underlying the control of T3 on trafficking of Na-K-ATPase remain to be determined.

The T3-stimulated translocation of Na-K-ATPase enzyme to the cell surface is necessary for T3-induced Na-K-ATPase activity in both AEC lines. Lo and Edelman (38) reported that the T3-induced increase in Na-K-ATPase activity in rat kidney in vivo appeared to occur through stimulation of enzyme synthesis, as they measured incorporation of labeled methionine into the protein in a membrane-rich fraction. However, because their assayed proteins came from the membrane-rich fraction, the observed increase in enzyme protein may have resulted from translocation of Na-K-ATPase enzyme units to the membrane, rather than increased synthesis. Indeed, their subsequent study also verified that T3 augmented the number of membrane-bound Na-K-ATPase units in rat kidney (39). Thus we propose that the T3-induced rapid increase of Na-KATPase activity depends on translocation of Na-KATPase to the cell plasma membrane but cannot exclude effects on recycling.

This rapid increase in plasma membrane Na-KATPase is similar to the augmentation of ATII cell Na-K-ATPase activity by dopamine and {beta}-adrenergic agonists reported by Sznajder and colleagues (4, 26, 45, 46). In their studies, increased Na+ pump activity resulted from a rapid translocation of Na-K-ATPase to the plasma membrane from a late endosomal compartment and from the slower effects of a mitogen-activated protein kinase pathway. Protein kinase C and phosphoinositide 3-kinase also regulate Na-K-ATPase protein trafficking, as occurs with dopamine in the kidney (56). In our studies of T3, it is likely that other processes, such as phosphorylation of Na-K-ATPase, and other T3-activated signals are required for this short-term stimulation by T3 along with translocation to the plasma membrane. For example, thyroid hormone stimulates the mitogen-activated protein kinase in 293T cells, CV-1 cells, and HeLa cells (14, 35). Therefore, T3 stimulation of Na-K-ATPase function may also be linked to T3 activation of other kinases, such as phosphoinositide 3-kinase or mitogen-activated protein kinase.

In summary, T3 directly stimulates ATII cell and AEC Na+ pump activity through posttranscriptional activation, likely by increasing translocation of Na+ pump molecules to plasma membrane. Because T3 augments alveolar fluid clearance in vivo (22), we hypothesize that increased activity of Na-K-ATPase is at least part of the in vivo stimulatory mechanism. However, the effects of T3 on apical proteins involved in Na+ transport, such as the epithelial Na+ channel, have not been defined.


    DISCLOSURES
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 DISCLOSURES
 REFERENCES
 
This work was supported by grants from the Minnesota Medical Foundation, the Knoll Pharmaceuticals Thyroid Research Council, and the American Lung Association (S. Nowbar) and by National Heart, Lung, and Blood Institute Grant P50-HL-50152.


    FOOTNOTES
 

Address for reprint requests and other correspondence: D. H. Ingbar, Pulmonary, Allergy, and Critical Care Div., MMC276, 420 Delaware St. SE, Minneapolis, MN 55455 (E-mail: ingba001{at}umn.edu).

The costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked "advertisement" in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.


    REFERENCES
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 DISCLOSURES
 REFERENCES
 

  1. Arumanayagam M and Swaminathan R. Stimulation of the Na,K-ATPase activity of K562 human erythroleukemia cells by triiodothyronine. Life Sci 51: 1913-1920, 1992.[ISI][Medline]
  2. Barker PM, Walters DV, Markiewicz M, and Strang LB. Development of the lung liquid reabsorptive mechanism in fetal sheep: synergism of triiodothyronine and hydrocortisone. J Physiol 433: 435-449, 1991.[Abstract]
  3. Béguin P, Hasler U, Beggah A, Horisberger JD, and Greeing K. Membrane integration of Na,K-ATPase {alpha}-subunits and {beta}-subunit assembly. J Biol Chem 273: 24921-24931, 1998.[Abstract/Free Full Text]
  4. Bertorello AM, Ridge KM, Chibalin AV, Katz AI, and Sznajder JI. Isoproterenol increases Na,K-ATPase activity by membrane insertion of {alpha}-subunits in lung alveolar cells. Am J Physiol Lung Cell Mol Physiol 276: L20-L27, 1999.[Abstract/Free Full Text]
  5. Blanco G and Mercer RW. Isozymes of the Na,K-ATPase: heterogeneity in structure, diversity in function. Am J Physiol Renal Physiol 275: F633-F650, 1998.[Abstract/Free Full Text]
  6. Book CBS, Sun XW, and Ng YC. Developmental changes in regulation of the Na,K-ATPase {alpha}3 isoform by thyroid hormone in ferret heart. Biochim Biophys Acta 1358: 172-180, 1997.[ISI][Medline]
  7. Brent GA. The molecular basis of thyroid hormone action. N Engl J Med 331: 847-853, 1994.[Free Full Text]
  8. Chardin P and McCormick F. Brefeldin A: the advantage of being uncompetitive. Cell 97: 153-155, 1999.[ISI][Medline]
  9. Chatterjee D, Mukherjee S, and Das SK. Regulation of choline phosphotransferase by thyroid hormone. Biochem Biophys Res Commun 282: 861-864, 2001.[ISI][Medline]
  10. Chen Y, Chen PL, Chen CF, Sharp DZ, and Lee WH. Thyroid hormone, T3-dependent phosphorylation and translocation of Trip230 from the Golgi complex to the nucleus. Proc Natl Acad Sci USA 96: 4443-4448, 1999.[Abstract/Free Full Text]
  11. Chibalin AV, Ogimoto G, Pedemonte CH, Pressley T, Katz A, Feraille E, Berggren PO, and Bertorello AM. Dopamine-induced endocytosis of Na,K-ATPase is initiated by phosphorylation of Ser-18 in the rat {alpha}-subunit and is responsible for the decreased activity in epithelial cells. J Biol Chem 274: 1920-1927, 1999.[Abstract/Free Full Text]
  12. Chow DC and Forte J. Functional significance of the {beta}-subunit for heterodimeric P-type ATPases. J Exp Biol 198: 1-17, 1995.[Abstract/Free Full Text]
  13. Davis PJ and Davis FB. Nongenomic actions of thyroid hormone on the heart. Thyroid 12: 459-466, 2002.[ISI][Medline]
  14. Davis PJ, Shih A, Lin HY, Martino LJ, and Davis FB. Thyroxine promotes association of mitogen-activated protein kinase and nuclear thyroid hormone receptor (TR) and causes serine phosphorylation of TR. J Biol Chem 275: 38032-38039, 2000.[Abstract/Free Full Text]
  15. Driscoll KE, Carter JM, Iype PT, Kumari HL, Crosby LL, Aardema MJ, Isfort RJ, Cody D, Chestnut MH, Burns JL, et al. Establishment of immortalized alveolar type II epithelial cell lines from adult rats. In Vitro Cell Dev Biol Anim 31: 516-527, 1995.[ISI][Medline]
  16. Effors RM, Mason GR, Hukkanen J, and Silverman P. New evidence for active sodium transport from fluid-filled rat lungs. J Appl Physiol 66: 906-919, 1989.[Abstract/Free Full Text]
  17. Ewart HS and Klip A. Hormonal regulation of the Na,KATPase: mechanisms underlying rapid and sustained changes in pump activity. Am J Physiol Cell Physiol 269: C295-C311, 1995.[Abstract/Free Full Text]
  18. Falkenstein E, Tillmann HC, Christ M, Feuring M, and Wehling M. Multiple actions of steroid hormones—a focus on rapid, nongenomic effects. Pharmacol Rev 52: 513-556, 2000.[Abstract/Free Full Text]
  19. Farwell AP, DiBenedetto DJ, and Leonard JL. Thyroxine targets different pathways of internalization of type II iodothyronine 5'-deiodinase in astrocytes. J Biol Chem 268: 5055-5062, 1993.[Abstract/Free Full Text]
  20. Feng J, Orlowski J, and Lingrel JB. Identification of a functional thyroid hormone response element in the upstream flanking region of the human Na,K-ATPase {beta}1 gene. Nucleic Acids Res 21: 2619-2626, 1993.[Abstract]
  21. Féraille E and Doucet A. Sodium-potassium-adenosinetriphosphatase-dependent sodium transport in the kidney: hormonal control. Physiol Rev 81: 345-418, 2001.[Abstract/Free Full Text]
  22. Folkesson HG, Norlin A, Wang Y, Abedinpour P, and Matthay MA. Dexamethasone and thyroid hormone pretreatment upregulate alveolar epithelial fluid clearance in adult rats. J Appl Physiol 88: 416-424, 2000.[Abstract/Free Full Text]
  23. Gick GG, Melikian J, and Ismail-Beigi F. Thyroidal enhancement of rat myocardial Na,K-ATPase: preferential expression of {alpha}2 activity and mRNA abundance. J Membr Biol 115: 273-282, 1990.[ISI][Medline]
  24. Gonin S, Deschênes G, Roger F, Bens M, Martin PY, Carpentier JL, Vandewalle A, Doucet A, and Fêraille E. Cyclic AMP increases cell surface expression of functional Na,KATPase units in mammalian cortical collecting duct principal cells. Mol Biol Cell 13: 255-264, 2001.
  25. Gottardi CJ, Dunbar LA, and Caplan MJ. Biotinylation and assessment of membrane polarity: caveats and methodological concerns. Am J Physiol Renal Fluid Electrolyte Physiol 268: F285-F295, 1995.[Abstract/Free Full Text]
  26. Guerrero C, Lecuona E, Pesce L, Ridge KM, and Sznajder JI. Dopamine regulates Na,K-ATPase in alveolar epithelial cells via MAPK-ERK-dependent mechanisms. Am J Physiol Lung Cell Mol Physiol 281: L79-L85, 2001.[Abstract/Free Full Text]
  27. Haber RS, Ismail-Beigi F, and Loeb JN. Time course of Na,K-ATPase and other metabolic responses to thyroid hormone in clone 9 cells. Endocrinology 123: 238-247, 1988.[Abstract]
  28. Horowitz B, Hensley CB, Quintero M, Azuma KK, Putnam D, and McDonough AA. Differential regulation of Na,K-ATPase {alpha}1, {alpha}2, and {beta} subunit mRNA and protein levels by thyroid hormone. J Biol Chem 265: 14308-14314, 1990.[Abstract/Free Full Text]
  29. Huang F, He H, and Gick G. Thyroid hormone regulation of Na,K-ATPase {alpha}2 gene expression in cardiac myocytes. Cell Mol Biol Res 40: 41-52, 1994.[ISI][Medline]
  30. Ingbar DH, Duvick S, Savick KS, Schellhase DE, Detterding R, Jamieson JD, and Shannon J. Developmental changes of fetal rat lung Na,K-ATPase after maternal treatment with dexamethasone. Am J Physiol Lung Cell Mol Physiol 272: L665-L672, 1997.[Abstract/Free Full Text]
  31. Ingbar DH, Weeks CB, Gilmore-Hebert M, Jacobsen E, Duvick S, Dowin R, Savik SK, and Jamieson JD. Developmental regulation of Na,K-ATPase in rat lung. Am J Physiol Lung Cell Mol Physiol 270: L619-L629, 1996.[Abstract/Free Full Text]
  32. Jiang X, Ingbar DH, and O'Grady SM. Adrenergic regulation of ion transport across adult alveolar epithelial cells: effects on Cl- channel activation and transport function in cultures with an apical air interface. J Membr Biol 181: 195-204, 2001.[ISI][Medline]
  33. Jørgensen PL and Skou JC. Preparation of highly active (Na+ + K+)-ATPase from the outer medulla of rabbit kidney. Biochem Biophys Res Commun 37: 39-46, 1969.[ISI][Medline]
  34. Kurihara K, Tajima Y, Kawasaki M, and Ueha T. Modulation of thyroid hormone-dependent Na-K-ATPase induction in cultured human submandibular gland cell lines, HSG cells. Oral Oncol 35: 368-374, 1999.[ISI][Medline]
  35. Lin HY, Davis FB, Gordinier JK, Martino LJ, and Davis PJ. Thyroid hormone induces activation of mitogen-activated protein kinase in cultured cells. Am J Physiol Cell Physiol 276: C1014-C1024, 1999.[Abstract/Free Full Text]
  36. Lin MH and Akera T. Increased (Na+ + K+)-ATPase concentrations in various tissues of rats caused by thyroid hormone treatment. J Biol Chem 253: 723-726, 1978.[Abstract]
  37. Lo CS, August TR, Liberman UA, and Edelman IS. Dependence of renal (Na+ + K+)-adenosine triphosphatase activity on thyroid status. J Biol Chem 251: 7826-7833, 1976.[Abstract]
  38. Lo CS and Edelman IS. Effect of triiodothyronine on the synthesis and degradation of renal cortical (Na+ + K+)-adenosine triphosphatase. J Biol Chem 251: 7834-7840, 1976.[Abstract]
  39. Lo CS and Lo TN. Triiodothyronine augments the number of membrane-bound (Na+-K+)-adenosine triphosphatase units, but does not affect the sedimentation properties of plasma membrane components. Endocrinology 109: 1473-1478, 1981.[Abstract]
  40. Matthay MA, Folkesson HG, and Verkman AS. Salt and water transport across alveolar and distal airway epithelia in the adult lung. Am J Physiol Lung Cell Mol Physiol 270: L487-L503, 1996.[Abstract/Free Full Text]
  41. Minor NT, Sha Q, Nichols CG, and Mercer RW. The {gamma}-subunit of the Na,K-ATPase induces cation channel activity. Proc Natl Acad Sci USA 95: 6521-6525, 1998.[Abstract/Free Full Text]
  42. Misumi Y, Misumi Y, Miki K, Takatsuki A, Tamura G, and Ikehara Y. Novel blockade by brefeldin A of intracellular transport of secretory proteins in cultured rat hepatocytes. J Biol Chem 261: 11398-11403, 1986.[Abstract/Free Full Text]
  43. Olivera WG, Ridge KM, Wood LDH, and Sznajder JI. Active sodium transport and alveolar epithelial Na,K-ATPase increase during subacute hyperoxia in rats. Am J Physiol Lung Cell Mol Physiol 266: L577-L584, 1994.[Abstract/Free Full Text]
  44. Pasternack M, Floerchinger CS, and Hunninghake LW. E1A-induced immortalization of rat type II alveolar epithelial cells. Exp Lung Res 22: 525-539, 1996.[ISI][Medline]
  45. Pesce L, Guerrero C, Comellas A, Ridge KM, and Sznajder JI. {beta}-Adrenergic agonists regulate Na,K-ATPase via novel MAPK/ERK and rapamycin-sensitive pathways. FEBS Lett 486: 310-314, 2000.[ISI][Medline]
  46. Ridge KM, Dada L, Lecuona E, Bertorello AM, Katz AI, Mochly-Rosen D, and Sznajder JI. Dopamine-induced exocytosis of Na,K-ATPase is dependent on activation of protein kinase C-{epsilon} and -{delta}. Mol Biol Cell 13: 1381-1389, 2002.[Abstract/Free Full Text]
  47. Samuels HH, Stanley F, and Casanova J. Depletion of L-3,5,3'-triiodothyronine and L-thyroxine in euthyroid calf serum for use in cell culture studies of the action of thyroid hormone. Endocrinology 105: 80-85, 1979.[Abstract]
  48. Sarkar PK and Ray AK. Synaptosomal action of thyroid hormone: changes in Na,K-ATPase activity in adult rat cerebral cortex. Horm Metab Res 25: 1-3, 1993.[ISI][Medline]
  49. Shameena B, Varghese S, Leena S, and Oommen OV. 3,5,3'-Triidothyronine (T3) and 3',5'-diiodothyronine (T2) have short-term effects on lipid metabolism in a teleost Anabas testudineus (Bloch): evidence from enzyme activities. Endocr Res 26: 431-444, 2000.[ISI][Medline]
  50. Shulkin BL and Utiger RD. Reverse triiodothyronine does not alter pituitary-thyroid function in normal subjects. J Clin Endocrinol Metab 58: 1184-1187, 1984.[Abstract]
  51. Suzuki S, Zuege D, and Berthiaume Y. Sodium-independent modulation of Na,K-ATPase activity by {beta}-adrenergic agonist in alveolar type II cells. Am J Physiol Lung Cell Mol Physiol 268: L983-L990, 1995.[Abstract/Free Full Text]
  52. Therien AG and Blostein R. Mechanism of sodium pump regulation. Am J Physiol Cell Physiol 279: C541-C566, 2000.[Abstract/Free Full Text]
  53. Therien AG, Karlish SJ, and Blostein R. Expression and functional role of the {gamma}-subunit of the Na,K-ATPase in mammalian cells. J Biol Chem 274: 12252-12256, 1999.[Abstract/Free Full Text]
  54. Wu Y and Koenig RJ. Gene regulation by thyroid hormone. Trends Endocrinol Metab 11: 207-211, 2000.[ISI][Medline]
  55. Yen PM. Physiological and molecular basis of thyroid hormone action. Physiol Rev 81: 1097-1142, 2001.[Abstract/Free Full Text]
  56. Yudowski GA, Efendiev R, Pedemonte CH, Katz AI, Berggren PO, and Bertorello AM. Phosphoinositide-3-kinase binds to a proline-rich motif in the Na,K-ATPase {alpha}-subunit and regulates its trafficking. Proc Natl Acad Sci USA 97: 6556-6561, 2000.[Abstract/Free Full Text]