Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 288(6): 3929-37, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23264624

ABSTRACT

Scaffolding proteins often bring kinases together with their substrates to facilitate cell signaling. This arrangement is critical for the phosphorylation and regulation of the transient receptor potential vanilloid 1 (TRPV1) channel, a key target of inflammatory mediators such as prostaglandins. The protein kinase A anchoring protein AKAP79/150 organizes a multiprotein complex to position protein kinase A (PKA) and protein kinase C (PKC) in the immediate proximity of TRPV1 channels to enhance phosphorylation efficiency. This arrangement suggests that regulators upstream of the kinases must also be present in the signalosome. Here, we show that AKAP79/150 facilitates a complex containing TPRV1 and adenylyl cyclase (AC). The anchoring of AC to this complex generates local pools of cAMP, shifting the concentration of forskolin required to attenuate capsaicin-dependent TRPV1 desensitization by ∼100-fold. Anchoring of AC to the complex also sensitizes the channel to activation by ß-adrenergic receptor agonists. Significant AC activity is found associated with TRPV1 in dorsal root ganglia. The dissociation of AC from an AKAP150-TRPV1 complex in dorsal root ganglia neurons abolishes sensitization of TRPV1 induced by forskolin and prostaglandin E(2). Thus, the direct anchoring of both PKA and AC to TRPV1 by AKAP79/150 facilitates the response to inflammatory mediators and may be critical in the pathogenesis of thermal hyperalgesia.


Subject(s)
A Kinase Anchor Proteins/metabolism , Adenylyl Cyclases/metabolism , Ganglia, Spinal/metabolism , Multiprotein Complexes/metabolism , Nerve Tissue Proteins/metabolism , TRPV Cation Channels/metabolism , A Kinase Anchor Proteins/genetics , Adenylyl Cyclases/genetics , Adrenergic beta-Agonists/pharmacology , Animals , Capsaicin/pharmacology , Colforsin/pharmacology , Cyclic AMP/genetics , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Dinoprostone/genetics , Dinoprostone/metabolism , Ganglia, Spinal/pathology , HEK293 Cells , Humans , Hyperalgesia/genetics , Hyperalgesia/metabolism , Hyperalgesia/pathology , Mice , Multiprotein Complexes/genetics , Nerve Tissue Proteins/genetics , Protein Kinase C/genetics , Protein Kinase C/metabolism , Sensory System Agents/pharmacology , TRPV Cation Channels/genetics
2.
J Cardiovasc Pharmacol ; 58(4): 339-44, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21978991

ABSTRACT

3'-5'-Cyclic adenosine monophosphate (cAMP), generated by adenylyl cyclase (AC), serves as a second messenger in signaling pathways regulating many aspects of cardiac physiology, including contraction rate and action potential duration, and in the pathophysiology of hypertrophy and heart failure. A kinase-anchoring proteins localize the effect of cAMP in space and time by organizing receptors, AC, protein kinase A, and other components of the cAMP cascade into multiprotein complexes. In this review, we discuss how the interaction of A kinase-anchoring proteins with distinct AC isoforms affects cardiovascular physiology.


Subject(s)
A Kinase Anchor Proteins/metabolism , Adenylyl Cyclases/metabolism , Cyclic AMP/metabolism , Action Potentials , Adenylyl Cyclases/chemistry , Animals , Cardiovascular Diseases/physiopathology , Cardiovascular System/metabolism , Humans , Isoenzymes , Signal Transduction/physiology , Time Factors
3.
J Biol Chem ; 285(19): 14450-8, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20231277

ABSTRACT

Spatiotemporal specificity of cAMP action is best explained by targeting protein kinase A (PKA) to its substrates by A-kinase-anchoring proteins (AKAPs). At synapses in the brain, AKAP79/150 incorporates PKA and other regulatory enzymes into signal transduction networks that include beta-adrenergic receptors, alpha-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA), and N-methyl-d-aspartic acid receptors. We previously showed that AKAP79/150 clusters PKA with type 5 adenylyl cyclase (AC5) to assemble a negative feedback loop in which the anchored kinase phosphorylates AC5 to dynamically suppress cAMP synthesis. We now show that AKAP79 can associate with multiple AC isoforms. The N-terminal regions of AC5, -6, and -9 mediate this protein-protein interaction. Mapping studies located a reciprocal binding surface between residues 77-108 of AKAP79. Intensity- and lifetime-based fluorescence resonance energy transfer demonstrated that deletion of AKAP79(77-108) region abolished AC5-AKAP79 interaction in living cells. The addition of the AKAP79(77-153) polypeptide fragment uncouples AC5/6 interactions with the anchoring protein and prevents PKA-mediated inhibition of AC activity in membranes. Use of the AKAP79(77-153) polypeptide fragment in brain extracts from wild-type and AKAP150(-/-) mice reveals that loss of the anchoring protein results in decreased AMPA receptor-associated AC activity. Thus, we propose that AKAP79/150 mediates protein-protein interactions that place AC5 in proximity to synaptic AMPA receptors.


Subject(s)
A Kinase Anchor Proteins/metabolism , Adenylyl Cyclases/metabolism , Hippocampus/metabolism , Isoenzymes/metabolism , Neurons/metabolism , Receptors, AMPA/metabolism , Animals , Blotting, Western , Cell Membrane/metabolism , Cells, Cultured , Humans , Immunoenzyme Techniques , Immunoprecipitation , Kidney/metabolism , Mice
4.
J Biol Chem ; 284(35): 23540-6, 2009 Aug 28.
Article in English | MEDLINE | ID: mdl-19574217

ABSTRACT

Protein kinase A-anchoring proteins (AKAPs) play important roles in the compartmentation of cAMP signaling, anchoring protein kinase A (PKA) to specific cellular organelles and serving as scaffolds that assemble localized signaling cascades. Although AKAPs have been recently shown to bind adenylyl cyclase (AC), the functional significance of this association has not been studied. In cardiac myocytes, the muscle protein kinase A-anchoring protein beta (mAKAPbeta) coordinates cAMP-dependent, calcium, and MAP kinase pathways and is important for cellular hypertrophy. We now show that mAKAPbeta selectively binds type 5 AC in the heart and that mAKAPbeta-associated AC activity is absent in AC5 knock-out hearts. Consistent with its known inhibition by PKA phosphorylation, AC5 is inhibited by association with mAKAPbeta-PKA complexes. AC5 binds to a unique N-terminal site on mAKAP-(245-340), and expression of this peptide disrupts endogenous mAKAPbeta-AC association. Accordingly, disruption of mAKAPbeta-AC5 complexes in neonatal cardiac myocytes results in increased cAMP and hypertrophy in the absence of agonist stimulation. Taken together, these results show that the association of AC5 with the mAKAPbeta complex is required for the regulation of cAMP second messenger controlling cardiac myocyte hypertrophy.


Subject(s)
A Kinase Anchor Proteins/metabolism , Adenylyl Cyclases/metabolism , Cyclic AMP/metabolism , Isoenzymes/metabolism , Myocytes, Cardiac/enzymology , Signal Transduction , A Kinase Anchor Proteins/genetics , Adenylyl Cyclases/genetics , Animals , Cells, Cultured , Isoenzymes/genetics , Mice , Mice, Knockout , Myocytes, Cardiac/metabolism , Protein Binding , Rats , Rats, Sprague-Dawley
5.
Am J Physiol Renal Physiol ; 295(4): F1117-25, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18701625

ABSTRACT

Most of the transepithelial transport of sodium in proximal tubules occurs through the coordinated action of the apical sodium/proton exchanger and the basolateral Na-K-ATPase. Hormones that regulate proximal tubule sodium excretion regulate the activities of these proteins. We have previously demonstrated that the level of intracellular sodium concentration modulates the regulation of Na-K-ATPase activity by angiotensin II and dopamine. An increase of a few millimolars in intracellular sodium concentration leads to increased Na-K-ATPase activity without a statistically significant increase in the number of plasma membrane Na-K-ATPase molecules, as determined by cell surface protein biotinylation. Using total internal reflection fluorescence, we detected an increased number of Na-K-ATPase molecules in cytosolic compartments adjacent to the plasma membrane, suggesting that the increased intracellular sodium concentration induces a movement of Na-K-ATPase molecules toward the plasma membrane. While intracellular compartments containing Na-K-ATPase molecules are very close to the plasma membrane, compartments containing type 1 dopamine receptors (D1Rs) are distributed in different parts of the cell cytosol. Fluorescence determinations indicate that an increased intracellular sodium concentration induces the increased colocalization of dopamine receptors with Na-K-ATPase molecules in the region of the plasma membrane. We propose that under in vivo conditions, in response to a sodium load in the lumen of proximal tubules, an increased level of intracellular sodium in epithelial cells is an early event that triggers the cellular response that leads to dopamine inhibition of proximal tubule sodium reabsorption.


Subject(s)
Epithelial Cells/enzymology , Kidney Tubules, Proximal/cytology , Receptors, Dopamine D1/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Sodium/metabolism , Animals , Cell Compartmentation/physiology , Cell Line , Cell Membrane/enzymology , Endocytosis/physiology , Epithelial Cells/cytology , Green Fluorescent Proteins/genetics , Microscopy, Fluorescence , Opossums , Phosphorylation , Rats , Rubidium/pharmacokinetics , Sodium-Potassium-Exchanging ATPase/genetics , Transfection
6.
J Biol Chem ; 283(25): 17561-7, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18420589

ABSTRACT

Motion of integral membrane proteins to the plasma membrane in response to G-protein-coupled receptor signals requires selective cargo recognition motifs that bind adaptor protein 1 and clathrin. Angiotensin II, through the activation of AT1 receptors, promotes the recruitment to the plasma membrane of Na,K-ATPase molecules from intracellular compartments. We present evidence to demonstrate that a tyrosine-based sequence (IVVY-255) present within the Na,K-ATPase alpha1-subunit is involved in the binding of adaptor protein 1. Mutation of Tyr-255 to a phenylalanine residue in the Na,K-ATPase alpha1-subunit greatly reduces the angiotensin II-dependent activation of Na,K-ATPase, recruitment of Na,K-ATPase molecules to the plasma membrane, and association of adaptor protein 1 with Na,K-ATPase alpha1-subunit molecules. To determine protein-protein interaction, we used fluorescence resonance energy transfer between fluorophores attached to the Na,K-ATPase alpha1-subunit and adaptor protein 1. Although angiotensin II activation of AT1 receptors induces a significant increase in the level of fluorescence resonance energy transfer between the two molecules, this effect was blunted in cells expressing the Tyr-255 mutant. Thus, results from different methods and techniques suggest that the Tyr-255-based sequence within the NKA alpha1-subunit is the site of adaptor protein 1 binding in response to the G-protein-coupled receptor signals produced by angiotensin II binding to AT1 receptors.


Subject(s)
Adaptor Protein Complex 1/metabolism , Cell Membrane/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Tyrosine/chemistry , Angiotensin II/chemistry , Animals , Cell Line , Enzyme Activation , Mutation , Opossums , Phenylalanine/chemistry , Protein Binding , Protein Conformation , Rats , Transfection
7.
FEBS Lett ; 580(21): 5067-70, 2006 Sep 18.
Article in English | MEDLINE | ID: mdl-16949583

ABSTRACT

Dopamine is a major regulator of sodium reabsorption in proximal tubule epithelia. It induces the endocytosis of plasma membrane Na,K-ATPase molecules, and this results in a reduced capacity of the cells to transport sodium. Dopamine induces the phosphorylation of Ser-18 in the alpha1-subunit of Na,K-ATPase. Fluorescence resonance energy transfer analysis of cells expressing YFP-alpha1 and beta1-CFP reveals that treatment of the cells with dopamine increases energy transfer between CFP and YFP. This is consistent with a protein conformational change that results in the N-terminal end of alpha1 moving closer to the internal face of the plasma membrane.


Subject(s)
Endocytosis , Fluorescence Resonance Energy Transfer , Protein Subunits/chemistry , Receptors, G-Protein-Coupled/metabolism , Sodium-Potassium-Exchanging ATPase/chemistry , Sodium-Potassium-Exchanging ATPase/metabolism , Alkaloids , Androstadienes/pharmacology , Animals , Benzophenanthridines , Cells, Cultured , Dopamine/pharmacology , Endocytosis/drug effects , Opossums , Phenanthridines/pharmacology , Protein Conformation/drug effects , Protein Subunits/metabolism , Rats , Wortmannin
8.
Am J Respir Cell Mol Biol ; 35(1): 127-32, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16498080

ABSTRACT

Activation of G protein-coupled receptor by dopamine and hypoxia-generated reactive oxygen species promote Na+,K+-ATPase endocytosis. This effect is clathrin dependent and involves the activation of protein kinase C (PKC)-zeta and phosphorylation of the Na+,K+-ATPase alpha-subunit. Because the incorporation of cargo into clathrin vesicles requires association with adaptor proteins, we studied whether phosphorylation of adaptor protein (AP)-2 plays a role in its binding to the Na+,K+-ATPase alpha-subunit and thereby in its endocytosis. Dopamine induces a time-dependent phosphorylation of the AP-2 mu2 subunit. Using specific inhibitors and dominant-negative mutants, we establish that this effect was mediated by activation of the adaptor associated kinase 1/PKC-zeta isoform. Expression of the AP-2 mu2 bearing a mutation in its phosphorylation site (T156A) prevented Na+,K+-ATPase endocytosis and changes in activity induced by dopamine. Similarly, in lung alveolar epithelial cells, hypoxia-induced endocytosis of Na+,K+-ATPase requires the binding of AP-2 to the tyrosine-based motif (Tyr-537) located in the Na+,K+-ATPase alpha-subunit, and this effect requires phosphorylation of the AP-2 mu2 subunit. We conclude that phosphorylation of AP-2 mu2 subunit is essential for Na+,K+-ATPase endocytosis in response to a variety of signals, such as dopamine or reactive oxygen species.


Subject(s)
Adaptor Protein Complex 2/metabolism , Adaptor Protein Complex mu Subunits/metabolism , Endocytosis/drug effects , Reactive Oxygen Species/pharmacology , Receptors, G-Protein-Coupled/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Cell Hypoxia/drug effects , Cells, Cultured , Dopamine/pharmacology , Humans , Models, Biological , Mutation/genetics , Opossums , Phosphorylation/drug effects , Protein Binding , Tyrosine/metabolism
9.
J Am Soc Nephrol ; 17(1): 31-8, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16338965

ABSTRACT

Renal sodium homeostasis is a major determinant of BP and is regulated by several natriuretic and antinatriuretic hormones. These hormones, acting through intracellular secondary messengers, either activate or inhibit proximal tubule Na,K-ATPase. It was shown previously that phorbol esters and angiotensin II and serotonin induce the phosphorylation of both Ser-11 and Ser-18 of the Na,K-ATPase alpha-subunit. This results in the recruitment of Na,K-ATPase molecules to the plasma membrane and an increased capacity to transport sodium ions. Treatment of the same cells with dopamine leads to phosphorylation of the Na,K-ATPase alpha-subunit Ser-18. The subsequent internalization of Na,K-ATPase molecules results in a reduced capacity to transport sodium ions. These effects are observed in cells that express the rat-type Na,K-ATPase. However, the Na,K-ATPase alpha1-subunit of several species, such as human, pig, and mouse, does not have a Ser-18 in their N-terminal region. Therefore, the possibility exists that, in those species, the Na,K-ATPase is not regulated by the hormones that regulate natriuresis. This study presents evidence that in cells that express the human-type Na,K-ATPase, dopamine inhibits and phorbol esters activate the Na,K-ATPase-mediated transport. These opposite effects are mediated by the phosphorylation of the same amino acid residue, Ser-11 of Na,K-ATPase alpha1, and the presence of alpha1 Ser-18 is not essential for the hormonal regulation of Na,K-ATPase activity in LLCPK1 cells. It was observed that, whereas the regulatory stimulation of Na,K-ATPase is mediated by protein kinase Cbeta, the regulatory inhibition is mediated by protein kinase Czeta. This is similar to what was demonstrated previously in cells that express the rat-type Na,K-ATPase.


Subject(s)
Dopamine/pharmacology , Sodium-Potassium-Exchanging ATPase/metabolism , Amino Acid Sequence , Animals , Humans , Mice , Molecular Sequence Data , Phosphorylation , Protein Kinase C/physiology , Rats , Sodium-Potassium-Exchanging ATPase/chemistry , Swine , Tetradecanoylphorbol Acetate/pharmacology
10.
Semin Nephrol ; 25(5): 322-7, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16139687

ABSTRACT

In the current report we review the results that lay grounds for the model of intracellular sodium-mediated dopamine-induced endocytosis of Na,K-ATPase. Under conditions of a high salt diet, dopamine activates PKCzeta, which phosphorylates NKA alpha1 Ser-18. The phosphorylation produces a conformational change of alpha1 NH2-terminus, which through interaction with other domains of alpha1 exposes PI3K- and AP-2-binding domains. PI3K bound to the NKA alpha1 induces the recruitment and activation of other proteins involved in endocytosis, and PI3K-generated 3-phosphoinositides affect the local cytoskeleton and modify the biophysical conditions of the membrane for development of clathrin-coated pits. Plasma membrane phosphorylated NKA is internalized to specialized intracellular compartments where the NKA will be dephosphorylated. The NKA internalization results in a reduced Na+ transport by proximal tubule epithelial cells.


Subject(s)
Cardiotonic Agents/pharmacology , Dopamine/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Kidney Tubules, Proximal/cytology , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/drug effects , Animals , Blood Pressure/drug effects , Endocytosis/drug effects , Humans , Multiprotein Complexes/drug effects , Multiprotein Complexes/metabolism , Phosphorylation/drug effects , Protein Kinase C/drug effects , Protein Kinase C/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Vesicular Transport Proteins/drug effects , Vesicular Transport Proteins/metabolism
11.
J Biol Chem ; 280(16): 16272-7, 2005 Apr 22.
Article in English | MEDLINE | ID: mdl-15722354

ABSTRACT

Clathrin-dependent endocytosis of Na(+),K(+)-ATPase molecules in response to G protein-coupled receptor signals is triggered by phosphorylation of the alpha-subunit and the binding of phosphoinositide 3-kinase. In this study, we describe a molecular mechanism linking phosphorylation of Na(+),K(+)-ATPase alpha-subunit to binding and activation of phosphoinositide 3-kinase. Co-immunoprecipitation studies, as well as experiments using confocal microscopy, revealed that dopamine favored the association of 14-3-3 protein with the basolateral plasma membrane and its co-localization with the Na(+),K(+)-ATPase alpha-subunit. The functional relevance of this interaction was established in opossum kidney cells expressing a 14-3-3 dominant negative mutant, where dopamine failed to decrease Na(+),K(+)-ATPase activity and to promote its endocytosis. The phosphorylated Ser-18 residue within the alpha-subunit N terminus is critical for 14-3-3 binding. Activation of phosphoinositide 3-kinase by dopamine during Na(+),K(+)-ATPase endocytosis requires the binding of the kinase to a proline-rich domain within the alpha-subunit, and this effect was blocked by the presence of a 14-3-3 dominant negative mutant. Thus, the 14-3-3 protein represents a critical linking mechanism for recruiting phosphoinositide 3-kinase to the site of Na(+),K(+)-ATPase endocytosis.


Subject(s)
14-3-3 Proteins/metabolism , Endocytosis/physiology , Phosphatidylinositol 3-Kinases/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , 14-3-3 Proteins/genetics , Amino Acid Motifs , Animals , Binding Sites , Gene Transfer Techniques , Phosphorylation , Rats , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology
12.
Circ Res ; 95(11): 1100-8, 2004 Nov 26.
Article in English | MEDLINE | ID: mdl-15528469

ABSTRACT

Alpha-adducin polymorphism in humans is associated with abnormal renal sodium handling and high blood pressure. The mechanisms by which mutations in adducin affect the renal set point for sodium excretion are not known. Decreases in Na+,K+-ATPase activity attributable to endocytosis of active units in renal tubule cells by dopamine regulates sodium excretion during high-salt diet. Milan rats carrying the hypertensive adducin phenotype have a higher renal tubule Na+,K+-ATPase activity, and their Na+,K+-ATPase molecules do not undergo endocytosis in response to dopamine as do those of the normotensive strain. Dopamine fails to promote the interaction between adaptins and the Na+,K+-ATPase because of adaptin-mu2 subunit hyperphosphorylation. Expression of the hypertensive rat or human variant of adducin into normal renal epithelial cells recreates the hypertensive phenotype with higher Na+,K+-ATPase activity, mu2-subunit hyperphosphorylation, and impaired Na+,K+-ATPase endocytosis. Thus, increased renal Na+,K+-ATPase activity and altered sodium reabsorption in certain forms of hypertension could be attributed to a mutant form of adducin that impairs the dynamic regulation of renal Na+,K+-ATPase endocytosis in response to natriuretic signals.


Subject(s)
Adaptor Protein Complex 2/metabolism , Adaptor Protein Complex mu Subunits/metabolism , Cytoskeletal Proteins/physiology , Hypertension/genetics , Kidney Tubules/enzymology , Microfilament Proteins/physiology , Natriuresis/physiology , Sodium-Potassium-Exchanging ATPase/metabolism , Adaptor Protein Complex 2/chemistry , Adaptor Protein Complex mu Subunits/chemistry , Amino Acid Substitution , Animals , Blood Pressure/genetics , Blood Pressure/physiology , Cell Line/drug effects , Cell Line/enzymology , Cytoskeletal Proteins/genetics , Dopamine/pharmacology , Endocytosis/drug effects , Endosomes/enzymology , Epithelium/enzymology , Humans , Hypertension/enzymology , Hypertension/physiopathology , Kidney Tubules/drug effects , Microfilament Proteins/genetics , Mutagenesis, Site-Directed , Natriuresis/drug effects , Natriuresis/genetics , Opossums , Phosphoprotein Phosphatases/metabolism , Protein Interaction Mapping , Protein Subunits , Rats , Rats, Mutant Strains , Recombinant Fusion Proteins/physiology , Structure-Activity Relationship , Transfection
13.
J Biol Chem ; 278(31): 28719-26, 2003 Aug 01.
Article in English | MEDLINE | ID: mdl-12759348

ABSTRACT

The balance and cross-talk between natruretic and antinatruretic hormone receptors plays a critical role in the regulation of renal Na+ homeostasis, which is a major determinant of blood pressure. Dopamine and angiotensin II have antagonistic effects on renal Na+ and water excretion, which involves regulation of the Na+,K+-ATPase activity. Herein we demonstrate that angiotensin II (Ang II) stimulation of AT1 receptors in proximal tubule cells induces the recruitment of Na+,K+-ATPase molecules to the plasmalemma, in a process mediated by protein kinase Cbeta and interaction of the Na+,K+-ATPase with adaptor protein 1. Ang II stimulation led to phosphorylation of the alpha subunit Ser-11 and Ser-18 residues, and substitution of these amino acids with alanine residues completely abolished the Ang II-induced stimulation of Na+,K+-ATPase-mediated Rb+ transport. Thus, for Ang II-dependent stimulation of Na+,K+-ATPase activity, phosphorylation of these serine residues is essential and may constitute a triggering signal for recruitment of Na+,K+-ATPase molecules to the plasma membrane. When cells were treated simultaneously with saturating concentrations of dopamine and Ang II, either activation or inhibition of the Na+,K+-ATPase activity was produced dependent on the intracellular Na+ concentration, which was varied in a very narrow physiological range (9-19 mm). A small increase in intracellular Na+ concentrations induces the recruitment of D1 receptors to the plasma membrane and a reduction in plasma membrane AT1 receptors. Thus, one or more proteins may act as an intracellular Na+ concentration sensor and play a major regulatory role on the effect of hormones that regulate proximal tubule Na+ reabsorption.


Subject(s)
Homeostasis , Kidney Tubules, Proximal/metabolism , Receptors, Angiotensin/metabolism , Receptors, Dopamine D1/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Sodium/metabolism , Absorption , Angiotensin II/pharmacology , Animals , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Dopamine/pharmacology , Epithelial Cells , Kidney , Kidney Tubules, Proximal/drug effects , Opossums , Phosphorylation , Protein Kinase C/metabolism , Protein Kinase C beta , Receptor, Angiotensin, Type 1 , Rubidium/metabolism , Serine/metabolism , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology , Transfection
14.
Mol Biol Cell ; 14(3): 1149-57, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12631730

ABSTRACT

Dopamine (DA) increases Na(+),K(+)-ATPase activity in lung alveolar epithelial cells. This effect is associated with an increase in Na(+),K(+)-ATPase molecules within the plasma membrane (). Analysis of Na(+),K(+)-ATPase motion was performed in real-time in alveolar cells stably expressing Na(+),K(+)-ATPase molecules carrying a fluorescent tag (green fluorescent protein) in the alpha-subunit. The data demonstrate a distinct (random walk) pattern of basal movement of Na(+),K(+)-ATPase-containing vesicles in nontreated cells. DA increased the directional movement (by 3.5 fold) of the vesicles and an increase in their velocity (by 25%) that consequently promoted the incorporation of vesicles into the plasma membrane. The movement of Na(+),K(+)-ATPase-containing vesicles and incorporation into the plasma membrane were microtubule dependent, and disruption of this network perturbed vesicle motion toward the plasma membrane and prevented the increase in the Na(+),K(+)-ATPase activity induced by DA. Thus, recruitment of new Na(+),K(+)-ATPase molecules into the plasma membrane appears to be a major mechanism by which dopamine increases total cell Na(+),K(+)-ATPase activity.


Subject(s)
Cell Membrane/metabolism , Dopamine/pharmacology , GTP-Binding Proteins/metabolism , Receptors, Cell Surface/metabolism , Respiratory Mucosa/drug effects , Sodium-Potassium-Exchanging ATPase/metabolism , Actins/metabolism , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line , Cell Polarity , Cytoplasmic Vesicles/metabolism , Green Fluorescent Proteins , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microtubules/metabolism , Paclitaxel/pharmacology , Protein Transport/physiology , Pulmonary Alveoli/cytology , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism , Rats , Recombinant Fusion Proteins/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , Signal Transduction/physiology , Sodium-Potassium-Exchanging ATPase/genetics , Thiazoles/pharmacology , Thiazolidines
15.
Br J Pharmacol ; 137(8): 1380-6, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12466249

ABSTRACT

1. The present study demonstrates that stimulation of hormonal receptors of proximal tubule cells with the serotonin-agonist 8-hydroxy-2-(di-n-propylamino) tetraline (8-OH-DPAT) induces an augmentation of Na(+),K(+)-ATPase activity that results from the recruitment of enzyme molecules to the plasmalemma. 2. Cells expressing the rodent wild-type Na(+),K(+)-ATPase alpha-subunit had the same basal Na(+),K(+)-ATPase activity as cells expressing the alpha-subunit S11A or S18A mutants, but stimulation of Na(+),K(+)-ATPase activity was completely abolished in either mutant. 3. 8-OH-DPAT treatment of OK cells led to PKC(beta)-dependent phosphorylation of the alpha-subunit Ser-11 and Ser-18 residues, and determination of enzyme activity with the S11A and S18A mutants indicated that both residues are essential for the agonist-dependent stimulation of Na(+),K(+)-ATPase activity. 4. When cells were treated with both dopamine and 8-OH-DPAT, an activation of Na(+),K(+)-ATPase was observed at basal intracellular sodium concentration (approximately 9 mM), and this activation was gradually reduced and became a significant inhibition as the concentration of intracellular sodium gradually increased from 9 to 19 mM. Thus, besides the antagonistic effects of dopamine and 8-OH-DPAT, intracellular sodium modulates whether an activation or an inhibition of Na(+),K(+)-ATPase is produced.


Subject(s)
Intracellular Fluid/metabolism , Protein Kinase C/metabolism , Receptors, Cell Surface/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Sodium/physiology , Animals , Cell Membrane/drug effects , Cell Membrane/enzymology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Intracellular Fluid/enzymology , Kidney/cytology , Kidney/drug effects , Kidney/enzymology , Opossums , Protein Kinase C/antagonists & inhibitors , Protein Kinase C beta , Receptors, Serotonin/metabolism , Sodium/metabolism , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors
16.
J Biol Chem ; 277(46): 44108-14, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12205083

ABSTRACT

Clathrin-dependent endocytosis of Na(+),K(+)-ATPase in response to dopamine regulates its catalytic activity in intact cells. Because fission of clathrin-coated pits requires dynamin, we examined the mechanisms by which dopamine receptor signals promote dynamin-2 recruitment and assembly at the site of Na(+),K(+)-ATPase endocytosis. Western blotting revealed that dopamine increased the association of dynamin-2 with the plasma membrane and with phosphatidylinositol 3-kinase. Dopamine inhibited Na(+),K(+)-ATPase activity in OK cells and in those overexpressing wild type dynamin-2 but not in cells expressing a dominant-negative mutant. Dephosphorylation of dynamin is important for its assembly. Dopamine increased protein phosphatase 2A activity and dephosphorylated dynamin-2. In cells expressing a dominant-negative mutant of protein phosphatase 2A, dopamine failed to dephosphorylate dynamin-2 and to reduce Na(+),K(+)-ATPase activity. Dynamin-2 is phosphorylated at Ser(848), and expression of the S848A mutant significantly blocked the inhibitory effect of dopamine. These results demonstrate a distinct signaling network originating from the dopamine receptor that regulates the state of dynamin-2 phosphorylation and that promotes its location (by interaction with phosphatidylinositol 3-kinase) at the site of Na(+),K(+)-ATPase endocytosis.


Subject(s)
Cell Membrane/metabolism , Dopamine/metabolism , Dynamin II/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Blotting, Western , Cell Line , Cells, Cultured , Clathrin/metabolism , Dynamins/metabolism , Electrophoresis, Polyacrylamide Gel , Endocytosis , Green Fluorescent Proteins , Kinetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Models, Biological , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Plasmids/metabolism , Precipitin Tests , Protein Binding , Rats , Time Factors , Transfection
17.
J Biol Chem ; 277(19): 17108-11, 2002 May 10.
Article in English | MEDLINE | ID: mdl-11859087

ABSTRACT

In renal epithelial cells endocytosis of Na(+),K(+)-ATPase molecules is initiated by phosphorylation of its alpha(1)-subunit, leading to activation of phosphoinositide 3-kinase and adaptor protein-2 (AP-2)/clathrin recruitment. The present study was performed to establish the identity of the AP-2 recognition domain(s) within the Na(+),K(+)-ATPase alpha(1)-subunit. We identified a conserved sequence (Y(537)LEL) within the alpha(1)-subunit that represents an AP-2 binding site. Binding of AP-2 to the Na(+),K(+)-ATPase alpha(1)-subunit in response to dopamine (DA) was increased in OK cells stably expressing the wild type rodent alpha-subunit (OK-WT), but not in cells expressing the Y537A mutant (OK-Y537A). DA treatment was associated with increased alpha(1)-subunit abundance in clathrin vesicles from OK-WT but not from OK-Y537A cells. In addition, this mutation also impaired the ability of DA to inhibit Na(+),K(+)-ATPase activity. Because phorbol esters increase Na(+),K(+)-ATPase activity in OK cells, and this effect was not affected by the Y537A mutation, the present results suggest that the identified motif is specifically required for DA-induced AP-2 binding and Na(+),K(+)-ATPase endocytosis.


Subject(s)
Carrier Proteins/metabolism , Clathrin/metabolism , Endocytosis , Membrane Proteins/metabolism , Sodium-Potassium-Exchanging ATPase/chemistry , Tyrosine/chemistry , Adaptor Proteins, Vesicular Transport , Animals , Cell Line , Dopamine/pharmacology , Microscopy, Confocal , Models, Chemical , Mutation , Opossums , Phorbol Esters/pharmacology , Plasmids/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Transfection , Tyrosine/metabolism
18.
J Biol Chem ; 277(13): 11489-96, 2002 Mar 29.
Article in English | MEDLINE | ID: mdl-11796710

ABSTRACT

We tested the hypothesis that the level of intracellular sodium modulates the hormonal regulation of the Na(+),K(+)-ATPase activity in proximal tubule cells. By using digital imaging fluorescence microscopy of a sodium-sensitive dye, we determined that the sodium ionophore monensin induced a dose-specific increase of intracellular sodium. A correspondence between the elevation of intracellular sodium and the level of dopamine-induced inhibition of Na(+),K(+)-ATPase activity was determined. At basal intracellular sodium concentration, stimulation of cellular protein kinase C by phorbol 12-myristate 13-acetate (PMA) promoted a significant increase in Na(+),K(+)-ATPase activity; however, this activation was gradually reduced as the concentration of intracellular sodium was increased to become a significant inhibition at concentrations of intracellular sodium higher than 16 mm. Under these conditions, PMA and dopamine share the same signaling pathway to inhibit the Na(+),K(+)-ATPase. The effects of PMA and dopamine on the Na(+),K(+)-ATPase activity and the modulation of these effects by different intracellular sodium concentrations were not modified when extracellular and intracellular calcium were almost eliminated. These results suggest that the level of intracellular sodium modulates whether hormones stimulate, inhibit, or have no effect on the Na(+),K(+)-ATPase activity leading to a tight control of sodium reabsorption.


Subject(s)
Kidney/enzymology , Sodium-Potassium-Exchanging ATPase/metabolism , Sodium/metabolism , Animals , Calcium/metabolism , Cell Line , Dopamine/pharmacology , Enzyme Activation , Monensin/pharmacology , Opossums , Osmolar Concentration , Protein Kinase C/metabolism , Signal Transduction , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Tetradecanoylphorbol Acetate/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...