Open access peer-reviewed chapter

The Na/K-ATPase Signaling Regulates Natriuresis in Renal Proximal Tubule

Written By

Jiang Liu, Yanling Yan and Joseph I. Shapiro

Submitted: 13 January 2020 Reviewed: 21 May 2020 Published: 30 September 2020

DOI: 10.5772/intechopen.92968

From the Edited Volume

Biomarkers and Bioanalysis Overview

Edited by Ane Claudia Fernandes Nunes

Chapter metrics overview

704 Chapter Downloads

View Full Metrics

Abstract

For decades, the Na/K-ATPase has been proposed and recognized as one of the targets for the regulation of renal salt handling. While direct inhibition of the Na/K-ATPase ion transport activity and sodium reabsorption was the focus, the underlying mechanism is not well understood since decreases in basolateral Na/K-ATPase activity alone do not appear sufficient to decrease net sodium reabsorption across the renal tubular epithelium. The newly appreciated signaling function of Na/K-ATPase, which can be regulated by Na/K-ATPase ligands (cardiotonic steroids (CTS)) and reactive oxygen species (ROS), has been widely confirmed and provides a mechanistic framework for natriuresis regulation in renal proximal tubule (RPT). The focus of this review aims to understand, in renal proximal tubule, how the activation of Na/K-ATPase signaling function, either by CTS or ROS, stimulates a coordinated reduction of cell surface Na/K-ATPase and sodium/hydrogen exchanger isoform 3 (NHE3) that leads to ultimately decreases in net transcellular sodium transport/reabsorption.

Keywords

  • cardiotonic steroids
  • natriuresis
  • renal proximal tubule
  • Na/K-ATPase
  • NHE3
  • signaling
  • ROS

1. Introduction

Since J.C. Skou’s discovery in 1957 [1], the energy-transducing Na/K-ATPase has been extensively studied for its ion-pumping function and, later on, its signaling function. While the signaling function was first demonstrated in cardiac myocyte primary culture, the phenomenon has been confirmed in different cell types and animal models. The roles of Na/K-ATPase signaling in renal proximal tubule (RPT) sodium handling and oxidative modification of the Na/K-ATPase α1 subunit in Na/K-ATPase signaling were explored both in vitro and in vivo. The findings may explain certain mechanism(s) related to the Na/K-ATPase signaling-ROS amplification loop and subsequent regulation of salt sensitivity.

The RPT mediates over 60% of the filtered Na+ reabsorption [2, 3]. There are two Na+ reabsorption pathways in RPTs. One is through the transcellular pathway, mainly through the apical Na+ entry mainly via NHE3 (and other apical Na+-coupled transporters like Na+-glucose cotransporters 1 and 2, to a lesser extent) and basolateral Na+ extrusion through the Na/K-ATPase [2, 3]. A coordinated and coupled regulation of sodium/hydrogen exchanger isoform 3 (NHE3, SLC9A3) and the Na/K-ATPase is critical in maintaining intracellular Na+ homeostasis and extracellular fluid volume. The other one is the paracellular Na+ reabsorption pathway through a tight junction (TJ), which depends on the transepithelial electrochemical force and tight junction permeability. Claudin-2 forms paracellular channels with other protein that are selective for small cations like Na+ and K+, small anion like Cl, as well as water [4, 5, 6]. Interestingly, the Na/K-ATPase signaling function is able to regulate the apical/basolateral polarity of the Na/K-ATPase as well as the tight junctions’ components like claudins in distal tubule MDCK cells [7, 8].

The Na/K-ATPase belongs to the P-type ATPase family and consists of two non-covalently linked α- and β-subunits. Several α- and β-isoforms, expressed in a tissue-specific manner, have been identified and functionally characterized [9, 10, 11, 12]. In RPTs, the γ-subunit (γa and γb, also known as FXYD2, one of the small type I single-span membrane FXYD protein families) also interacts with the α1 subunit to regulate the Na/K-ATPase activity [13, 14, 15]. There is also a fifth member of the β-subunit family, named βm coded by an ATP1B4 gene, that is predominantly expressed in skeletal muscle. Interestingly, the βm is not associated with the α1 subunit like other β-subunits, but accumulated in the nuclear membrane and associated with transcriptional coregulator Ski-interacting protein, which led to the regulation of TGF-β-responsive reporter Smad7 [16]. The α1 subunit contains multiple structural motifs that interact with soluble, membrane, and structural proteins. Binding to these proteins not only regulates the ion-pumping function of the enzyme, but it also conveys signal-transducing functions to the Na/K-ATPase [17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32]. NHE3 belongs to a family of electroneutral mammalian Na+/H+ exchangers [33, 34, 35]. In RPT, NHE3 resides in the apical membrane of S1 and S2 segments, mediating transcellular reabsorption of Na+ and HCO3 and fluid reabsorption [36, 37]. In the kidney, more than 85% of the filtered NaHCO3 is reabsorbed in the RPTs, and NHE3 contributes up to ∼60% of the total reabsorption of this segment [38]. RPT NHE3 secrets the largest portion of net H+ to the lumen and interacts with HCO3 to form H2O and CO2 which can freely translocate into RPT cytosol. In cytosol, H2O and CO2 form H+ and HCO3 through carbonic anhydrase catalyzation. Finally, the newly formed cytosolic H+ will be secreted to the lumen, and HCO3 will be moved to the blood through the basolateral-resided Na+/HCO3 cotransporter (NBCe1-A, SLC4A4). This cycling carbonic anhydrase-controlled CO2-HCO3 system links the NHE3-mediated H+ secretion to HCO3 reabsorption, to achieve an acid-base equilibrium [39, 40]. Moreover, vesicular NHE3 activity also regulates endosomal pH and consequently affects receptor-mediated endocytosis as well as endocytic vesicle fusion [41, 42]. Under normal conditions, the Na/K-ATPase resides at the basolateral surface, providing the driving force for the vectorial transport of Na+ from the tubular lumen to the vascular compartment, while the NHE3 resides at the apical surface providing a rate-limiting Na+ entry into cells.

Advertisement

2. The concept of endogenous cardiotonic steroids (CTS) as natriuretic hormones

CTS (also known as endogenous digitalis-like substances) are specific ligands and inhibitors of the Na/K-ATPase, which include plant-derived glycosides such as digoxin and ouabain and vertebrate-derived aglycones such as bufalin and marinobufagenin (MBG). Although the production and secretion of endogenous CTS are not completely understood, both ouabain and MBG have been identified as endogenous steroid hormones whose production and secretion can be regulated by multiple stimuli including angiotensin II and adrenocorticotropic hormone (ACTH) [30, 43, 44, 45, 46, 47, 48]. Endogenous CTS are present in measurable amounts under normal physiological conditions and are markedly increased under a number of pathological conditions such as sodium imbalance, chronic renal failure, hyperaldosteronism, hypertension, congestive heart failure, acute plasma volume expansion, and preeclampsia [46, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59].

Even though digitalis-like drugs have been used to treat heart failure patients for over 200 years, studies have also revealed many extra-cardiac actions of these compounds, such as in response to salt loading in both animal models and human hypertensive patients [29, 57, 60, 61, 62]. In addition, low doses of CTS not only induced hypertension in rats but also caused a significant cardiovascular remodeling independent of their effect on blood pressure (BP) [63, 64, 65, 66].

Bricker was the first to propose the existence of “the third factor” (named after the glomerular filtration rate as the first factor and the aldosterone as the second factor), and Dahl proposed the existence of a hormonal natriuretic factor that might cause a sustained increase in BP in salt-sensitive hypertensive rats [67, 68]. Subsequently, Bricker, de Wardener, and others proposed that this hormonal natriuretic factor inhibits the Na/K-ATPase, and Blaustein described how an increase in endogenous Na/K-ATPase inhibitors might cause a vascular contractility change and then a rise in BP [67, 69, 70, 71, 72]. In 1980, de Wardener and MacGregor summarized the state of research at the time and proposed an insightful scheme explaining how the Na/K-ATPase inhibitor works as a natriuretic hormone [73]. In essence, it was contended that the Na/K-ATPase inhibitor (endogenous CTS) will rise in response to either a defect in renal Na+ excretion or high salt intake. This increase, while returning Na+ balance toward normal by increasing renal Na+ excretion, also causes hypertension through acting on the vascular Na/K-ATPase. With the advances in the field over the decades, much has been learned. The first unequivocal demonstration of ouabain-like substance in the human plasma was reported decades ago [46]. Blaustein and Hamlyn’s laboratory has demonstrated how increases in endogenous CTS change vascular contractility and its effect on BP [74]. However, the pathophysiological significance of endogenous CTS (e.g., as a natriuretic hormone) has been a subject of debate since it was first proposed until Lingrel’s laboratory reported their gene replacement in vivo studies, which unequivocally demonstrated that endogenous CTS play an important role in the regulation of renal Na+ excretion and BP through the Na/K-ATPase [75, 76, 77]. Specifically, Lingrel’s group generated several lines of mice in which the mouse endogenous ouabain-insensitive α1 subunit is replaced by a mutant that alters the ouabain sensitivity of the Na/K-ATPase. For example, they generated a line of “humanized” α1S/S mice where the endogenous ouabain-insensitive α1 is replaced by an ouabain-sensitive (human like) α1-mutant and used these mice to explore the role of endogenous CTS in the regulation of renal function and BP. Should endogenous CTS be important for these regulations, an increased CTS sensitivity in α1S/S mice would make these mice more sensitive to conditions that raise circulating CTS. Indeed, when ACTH was administered to raise endogenous CTS, it caused much severe hypertension in α1S/S mice than their control littermates. Moreover, expression of the ouabain-sensitive α1-mutant significantly increased renal Na+ excretion, confirming the natriuretic function of endogenous CTS as proposed by the pioneers of the field [67, 68, 70, 71, 72, 73]. More evidences indicate that increases in endogenous CTS regulate both renal Na+ excretion and BP through the Na/K-ATPase [74, 75, 76, 78, 79].

Advertisement

3. The Na/K-ATPase signaling by specific ligands and ROS in RPTs

Ouabain-stimulated protein-protein interaction and subsequent Na/K-ATPase signaling function were first demonstrated in rat neonatal myocytes, which were further confirmed and developed in porcine LLC-PK1 cells (an immobilized RPT cell line) and other cell types. CTS-stimulated Na/K-ATPase signaling has been reviewed everywhere [22, 31, 32, 47, 80, 81, 82, 83].

In LLC-PK1 cells, ouabain-stimulated Na/K-ATPase signaling increases ROS generation. Other than ouabain, exogenous H2O2 and glucose oxidase-induced H2O2 also activate Na/K-ATPase signaling pathways including phosphorylation of c-Src and ERK1/2, as well as protein carbonylation modification of Na/K-ATPase (direct carbonylation of two amino acid residues, Pro222 and Thr224, in the actuator domain of the α1 subunit) [84, 85, 86, 87]. Pretreatment with antioxidant N-acetyl-l-cysteine (NAC) or disruption of the Na/K-ATPase/c-Src signaling complex attenuated ouabain- and glucose oxidase-stimulated Na/K-ATPase/c-Src signaling, protein carbonylation, redistribution of Na/K-ATPase, and inhibition of active transepithelial 22Na+ transport. A basal level of ROS is critical in initiating ouabain-stimulated Na/K-ATPase/c-Src signaling, and carbonylation modification of the α1 subunit is involved in a feed-forward mechanism of the regulation of ouabain-mediated Na/K-ATPase signal function and subsequent Na+ transport. Furthermore, a stable overexpression of rat α1-mutant Pro224/Ala (Pro224 of rat α1 is the same as the Pro222 of pig α1) prevented ouabain-stimulated signal function of Na/K-ATPase, protein carbonylation, Na/K-ATPase endocytosis, and ouabain-induced inhibition of active transepithelial 22Na+ transport [79, 86, 87]. Taken together, in LLC-PK1 cells, there is a positive-feedback amplification loop of Na/K-ATPase signaling and ROS generation, in which carbonylation of the Pro222 of the α1 subunit is critical. In this working model, both Na/K-ATPase-specific ligands (such as ouabain) and ROS increases (induced by other stimuli like exogenous added glucose oxidase) could activate the Na/K-ATPase signaling, and the Na/K-ATPase/c-Src complex can function as a “receptor” of ROS signaling. This Na/K-ATPase signaling-ROS axis may explain the role of Na/K-ATPase signaling in the development of different pathophysiological conditions, including RPT sodium handling.

Advertisement

4. Endocytosis of Na/K-ATPase

Endocytosis is involved in many important cellular functions. Ouabain-induced endocytosis of the Na/K-ATPase was first observed by the laboratories of Cook and Lamb, which demonstrated that [3H]-ouabain (bound to the Na/K-ATPase) was translocated from the plasmalemmal membrane surface to intracellular compartments (lysosomes) in HeLa cells, chick embryo heart cells, and Girardi heart cells [88, 89, 90, 91, 92].

4.1 Dopamine and PTH

One of the best-studied paradigms of hormonal natriuresis is the renal dopamine system [93, 94, 95, 96]. Renal dopamine release increases in response to high salt intake or volume expansion. The activation of D1-like dopamine receptors stimulates PLC-γ and cAMP-PKA pathways and increases intracellular Ca2+. These pathways work in concert and produce the coordinated downregulation of NHE3 and the Na/K-ATPase and consequently natriuresis [93, 94, 95, 97, 98]. While Aperia’s laboratory first revealed the pathways involved in dopamine-induced regulation of Na/K-ATPase activity [99, 100, 101] that is related to endocytosis of the Na/K-ATPase [102], Moe and others have mapped the pathways of NHE3 phosphorylation and trafficking [103, 104, 105]. In RPT, dopamine alters sodium handling by inducing Na/K-ATPase and NHE3 endocytosis. In RPT primary culture of Sprague-Dawley rats, dopamine-induced clathrin-dependent endocytosis of the rat Na/K-ATPase α1 subunit is triggered by activation of PI3K and subsequently phosphorylation of Ser-18 of rat α1 subunit [24, 106, 107, 108, 109]. The activation of PI3K also stimulated phosphorylation of the Tyr537 of the α1 subunit that facilitates its binding with adaptor protein-2 (AP-2), providing the inclusion of the Na/K-ATPase into clathrin-coated pits (CCP) [24, 108]. However, Ser-18 is found only in rat α1 subunit and is not present in pig and dog α1 subunits [110]. Depending on the type of renal tubular epithelium, dopamine-induced endocytosis of the Na/K-ATPase may be mediated through PKC- or PKA-dependent mechanisms [108, 111, 112, 113]. Parathyroid hormone (PTH)-induced inhibition and endocytosis of the Na/K-ATPase were also demonstrated in opossum kidney (OK) cells, which is clathrin-mediated and requires ERK-dependent phosphorylation of Ser-11 of the α1 subunit [114].

4.2 Ouabain-induced endocytosis of Na/K-ATPase through Na/K-ATPase signaling

In LLC-PK1 cells, at the doses used, ouabain has no discernable effects on cell morphology, viability, transepithelial electrical resistance, tight junction integrity, and intracellular [Na+] [115]. However, ouabain causes decreases in membrane-bound Na/K-ATPase without significantly affecting intracellular [Na+] [116, 117]. As a specific ligand, nontoxic ouabain (~1/10th–1/20th of acute IC50) caused a dose- and time-dependent decrease in Na/K-ATPase ion-pumping activity (ouabain-sensitive 86Rb uptake), which is attributed to ouabain-stimulated clathrin-dependent endocytosis of the α1/β1-subunits, demonstrated by a decrease in cell surface biotinylated α1 subunit and a concomitant accumulation of α1/β1-subunit and c-Src in early endosome (EE)/late endosome (LE) fractions. This leads to a net decrease in abundance of Na/K-ATPase in the plasma membrane and total ion-pumping activity of Na/K-ATPase and transcellular 22Na+ transport. This phenomenon was only observed when ouabain was applied to the basolateral, but not apical, aspect of Costar Transwell with membrane support for 12 hours, which indicates that this ouabain-induced endocytosis of the Na/K-ATPase is initiated by activating the receptor Na/K-ATPase/Src complex involving phosphorylation of c-Src and PI3K. The endocytosed [3H]-ouabain/Na/K-ATPase/c-Src/EGFR complex can be detected in both EE and LE fractions.

To understand the molecular mechanism(s) involved in this process, studies were performed with LLC-PK1 as well as SYF and SYF + c-Src cells. SYF cells are triple Src kinase (c-Src, Yes, Fyn)-null mouse fibroblast cells, and SYF + c-Src are c-Src-rescued SYF cells. This pair of cells was used to determine the role of c-Src activation in ouabain-induced Na/K-ATPase signaling and endocytosis. While ouabain accumulates Na/K-ATPase α1 subunit content in clathrin-coated pits and EE/LE fractions, it also causes a translocation of the α1 subunit to nuclear fraction. Interestingly, the effects of ouabain are fully reversible in terms of ion-pumping activity, transepithelial 22Na+ flux, and cell surface Na/K-ATPase within 24 hours following the removal of ouabain with a fresh culture medium, suggesting a reversible process. Immunofluorescence showed that the Na/K-ATPase α1 subunit co-localized with clathrin both before and after ouabain treatment, and immunoprecipitation experiments indicated that ouabain stimulated interactions among the α1 subunit, AP-2, and clathrin heavy chain (CHC). Disruption and/or arresting of clathrin-coated pit formation (by potassium depletion with hypotonic shock [118] and chlorpromazine treatment [119]) significantly attenuated this ouabain-induced endocytosis, suggesting the involvement of a clathrin-coated pit. Inhibition of the ouabain-activated signaling with PP2 (a specific c-Src kinase inhibitor) or wortmannin (a specific PI3K inhibitor) also significantly attenuated ouabain-induced endocytosis. Experiments performed in SYF cells and SYF + c-Src demonstrated that ouabain induces the endocytosis of the Na/K-ATPase in SYF + c-Src cells, but not in the SYF, indicating that ouabain-induced endocytosis of the Na/K-ATPase is c-Src-dependent.

Ouabain-stimulated Na/K-ATPase signaling also requires caveolin-1 (Cav-1) (a structural protein of caveolae, a subset of membrane lipid rafts) that functions as an anchoring protein for attracting the Na/K-ATPase α1 subunit into caveolae [120]. Accordingly, depletion of cholesterol (by methyl-β-cyclodextrin (Mβ-CD)) or caveolin-1 (by siRNA) blocked ouabain-induced endocytosis of the Na/K-ATPase, compartmentalization of signaling molecules in clathrin-coated pits, and early endosome. In addition, depletion of caveolin-1 also significantly reduced the protein-protein interactions among α1 subunit, AP-2, PI3K, and clathrin heavy chain, suggesting that caveolin-1 is involved in both ouabain-induced endocytosis of Na/K-ATPase and signal transduction [117].

These data demonstrate that ouabain stimulates a clathrin- and caveolin-1-dependent endocytosis of the Na/K-ATPase, a phenomenon requiring ouabain-induced Na/K-ATPase signaling function. Taken together, it is most likely that clathrin- and/or caveola-/lipid raft-mediated endocytosis of the Na/K-ATPase is a common phenomenon, but the mechanism and the relationship between the endocytosis of the Na/K-ATPase and signal transduction are still not fully understood. This is the first time to demonstrate that ligand-modulated endocytosis of the Na/K-ATPase is a mechanism by which RPT sodium transport is altered in a physiologically meaningful manner (Figure 1).

Figure 1.

Illustration of activation of the Na/K-ATPase signaling-mediated endocytosis of the Na/K-ATPase. Both CTS and ROS can activate Na/K-ATPase signaling, which leads to translocation of cell surface Na/K-ATPase (α1- and β1-subunits), along with EGFR, c-Src, and ERK1/2, into clathrin-coated pits and early and late endosomes. This process is independent of change in intracellular Na+ and Ca2+, but is dependent on activation of c-Src and PI3K, and the presence of caveolin-1. The activation of the Na/K-ATPase signaling also stimulates ROS generation which further activates the signaling. In LLC-PK1 cells, ouabain has no significant effect on recycling of endocytosed α1 subunit. AP-2, adaptor protein-2; Cav-1, caveolin-1; CCP, clathrin-coated pits; CHC, clathrin heavy chain; CTS, cardiotonic steroids; EE, early endosome; LE, late endosome; Na+/X, Na+-dependent antitransporter; Na+/Y, Na+-dependent cotransporter; NKA, Na/K-ATPase; TJ, tight junction.

Advertisement

5. The Na/K-ATPase signaling regulates NHE3 trafficking and activity

5.1 NHE3 regulation

In RPT, NHE3 resides in the apical membrane of S1 and S2 segments, mediating transcellular reabsorption of Na+ and HCO3 and fluid reabsorption [36, 37]. Moreover, vesicular NHE3 activity regulates endosomal pH and consequently affects receptor-mediated endocytosis as well as endocytic vesicle fusion [41, 42]. Consistent with its cellular function, upregulation of NHE3 activity and expression is associated with the development of hypertension [121, 122, 123, 124]. Conversely, the reduction of NHE3 surface expression or NHE3 activity occurs during pressure natriuresis in rats [125, 126, 127, 128]. As expected, NHE3-deficient mice are hypotensive [129, 130, 131] because of reduced Na+ reabsorption and increased Na+ excretion. Interestingly, NHE3-deficient mice also develop acidosis since the blunted H+ secretion through NHE3, which links to greatly reduced RPT HCO3 reabsorption (please see Introduction for the linkage of NHE3 H+ secretion and HCO3 reabsorption), could not be compensated by H+-ATPase and AE1 (anion exchanger-1, SLC4A1) Cl/HCO3 exchanger, compared with wild-type mice [131, 132]. These observations put renal Na+ reabsorption through NHE3 in a central position in the development and control of salt loading- and volume expansion-mediated hypertension. Structurally, NHE3 has a predicted N-terminal hydrophobic ion-translocating domain and a variable C-terminal hydrophilic domain which contains regulatory sequences [133].

The NHE3 activity is regulated at various levels through different mechanisms, mainly via phosphorylation, trafficking, and transcriptional regulation [34, 35, 103]. The surface expression of NHE3 is mainly regulated by changes in endocytosis/exocytosis and is the primary regulatory mechanism of NHE3 activity. NHE3 has been found to traffic between the plasma membrane and EE/LE fractions via a clathrin- and PI3K-dependent pathway [41, 134, 135, 136, 137, 138, 139, 140, 141]. The NHE3 activity can be stimulated by exocytosis [141, 142, 143] or inhibited by endocytosis [105, 125, 144]. The activation of c-Src, PKA, and PKC and increase in intracellular Ca2+ are involved in the regulation of NHE3 trafficking.

NHE3 has been shown to be redistributed under a hypertensive state, accompanying reversible downregulation of the Na/K-ATPase activity in the renal cortex [125, 127, 145]. This raised the possibility that the basolateral-localized Na/K-ATPase and apically localized NHE3 work in concert to regulate renal sodium handling in response to the Na/K-ATPase signaling. The coordinated regulation of NHE3 and the Na/K-ATPase is critical in maintaining intracellular Na+ homeostasis and extracellular fluid volume. It is believed that the apical Na+ entry through NHE3 is the rate-limiting step because the functional reserve of the Na/K-ATPase in the nephron is more than sufficient even under some pathological conditions.

5.2 Chronic NHE3 regulation by Na/K-ATPase signaling

In LLC-PK1 cells, chronic, low-concentration ouabain (50 and 100 nM, 24 hours) treatment in the basolateral aspect, but not in apical aspect, did not change intracellular [Na+] but decreased apical NHE3-mediated Na+ absorption, NHE3 promoter activity, and NHE3 protein and mRNA abundance. Pretreatment with specific inhibitors against c-Src and PI3K attenuates ouabain-induced downregulation of NHE3 activity and NHE3 mRNA [146]. In caveolin-1 knockdown LLC-PK1 cells, ouabain failed to reduce NHE3 mRNA and NHE3 promoter activity, in which ouabain-induced Na/K-ATPase signaling reduced Sp1 and TR DNA binding activity and consequently decreased NHE3 expression and activity [146]. These effects are abolished by inhibition of either c-Src or PI3K. Promoter mapping identified that ouabain-response elements reside in a region between −450 and −1194 nt and that ouabain reduces the binding of transcriptional factor Sp1 to its cognate cis-element.

5.3 Acute NHE3 regulation by Na/K-ATPase signaling

Acute application of ouabain (1 hour) in the basolateral, but not apical, aspect significantly reduced NHE3 activity (22Na+ uptake) and active transepithelial 22Na+ transport. This is accompanied by a reduced NHE3 content on cell surface and an increased NHE3 content in EE/LE fractions, as seen in the case of the Na/K-ATPase α1 subunit. These changes are independent of change in the integrity of tight junctions and the intracellular Na+ concentration [115]. Ouabain-induced NHE3 trafficking was abolished by either PI3K or c-Src inhibition. Disruption of caveolae/lipid rafts by cholesterol depletion prevented ouabain-induced accumulation of NHE3 and Na/K-ATPase α1 in early endosomes, and cholesterol repletion restored the ouabain-induced endosomal accumulation of NHE3 and Na/K-ATPase α1. Moreover, pretreatment of cells with the intracellular Ca2+ chelator BAPTA-AM attenuated ouabain-induced NHE3 trafficking, suggesting Ca2+ might link the Na/K-ATPase signaling to NHE3 regulation which is in agreement with observations that intracellular Ca2+ can regulate NHE3 activity and trafficking [147, 148]. These changes indicate that ouabain acutely stimulates NHE3 trafficking, like Na/K-ATPase, by activating the basolateral Na/K-ATPase signaling complex [115]. In RPT cell lines (human HK-2, porcine LLC-PK1, and AAC-19 originated from LLC-PK1 in which the pig α1 was replaced by ouabain-resistant rat α1), results further indicate that ouabain-induced inhibition of transcellular 22Na+ transport as well as trafficking of the α1 subunit and NHE3 is not a species-specific phenomenon. Furthermore, in LLC-PK1 cells, ouabain inhibited the endocytic recycling of internalized NHE3, but has no significant effect on recycling of endocytosed α1 subunit [149].

Taken together, by activating the basolateral receptor Na/K-ATPase/c-Src complex, ouabain can simultaneously and coordinately regulate trafficking of basolateral Na/K-ATPase and apical NHE3, leading to inhibition of transepithelial Na+ transport. This mechanism may be important to RPT Na+ handling during conditions associated with increases in circulating endogenous CTS. However, it remains to be established whether ouabain-induced regulation of NHE3 trafficking comes from the endocytosed Na/K-ATPase/c-Src complex or directly from the plasma membrane, since ouabain still binds to endocytosed Na/K-ATPase (Figure 2).

Figure 2.

Illustration of activation of the Na/K-ATPase signaling-mediated endocytosis of NHE3. Activation of the Na/K-ATPase signaling leads to intracellular Na+-independent NHE3 endocytosis. However, like Na/K-ATPase signaling-mediated Na/K-ATPase endocytosis, the NHE3 endocytosis is dependent on intracellular Ca2+, activation of c-Src and PI3K, and caveolin-1. In LLC-PK1 cells, ouabain inhibits the endocytic recycling of endocytosed NHE3. Since the Na/K-ATPase and NHE3 reside on basolateral and apical membrane in monolayer, respectively, it is still unclear how the basolateral Na/K-ATPase signaling is transmitted to NHE3 regulation. There are several possible pathways as illustrated, as proposed in the text (please see Figure 1 for abbreviations).

Advertisement

6. Ouabain-induced regulation of Na/K-ATPase α1 subunit and NHE3 is independent of intracellular [Na+]

High concentrations of ouabain are known to increase intracellular [Na+], depolarize the proximal tubule, and affect the tight junction of epithelial cells. In LLC-PK1 cells, ouabain (up to 100 nM) has no acute effect on intracellular [Na+], transepithelial electrical resistance, and tight junction integrity, suggesting that in the concentration, ouabain is not likely to increase passive Na+ transport by depolarizing LLC-PK1 monolayers [115]. To further define whether the effects of ouabain on the Na/K-ATPase and NHE3 are independent of intracellular [Na+], the change in intracellular transporters after the equilibrium of intracellular [Na+] with extracellular [Na+] was achieved by using conventional “Na+-clamping” methods [150]. LLC-PK1 cells (both control and ouabain-treated) are pretreated either with 20 μM monensin or with 10 μM monensin plus 5 μM gramicidin for 30 min. Both “clamping” methods raise basal levels of α1 and NHE3 in EE/LE fractions (monensin is known to accumulate proteins in intracellular compartments). However, ouabain is still able to further accumulate more α1 and NHE3 in EE/LE. These observations indicate that ouabain-induced trafficking of α1 and NHE3 can be independent of intracellular [Na+] change [115].

Advertisement

7. Coordinated and coupled regulation of Na/K-ATPase and NHE3 by Na/K-ATPase signaling

Although the mechanisms are still being elucidated, accumulating evidence supports the notion that the expression and activity of the basolateral Na/K-ATPase and apical NHE3 are coordinated and coupled under certain circumstances. For example, McDonough’s laboratory has shown that, during pressure natriuresis and salt loading, the surface expression and activity of both NHE3 and the Na/K-ATPase are simultaneously downregulated to remove Na+ from the body [125, 127, 145, 151]. During the development of hypertension in spontaneous hypertensive rat (SHR), the expression and activity of both the Na/K-ATPase and NHE3 are elevated in comparison with the normotensive control rats [121, 152, 153, 154, 155].

Activation of Na/K-ATPase signaling, by either ouabain or a high-salt diet, is also capable of stimulating a coordinated and coupled downregulation of apical NHE3 and basolateral Na/K-ATPase to inhibit active transepithelial Na+ transport in cultured or isolated RPTs [79, 115, 116, 117, 149]. This coordinated regulation depends on activation of the Na/K-ATPase signaling function, but not on acute inhibition of the Na/K-ATPase activity since it requires the activation of Src and PI3K and increase in intracellular Ca2+. Moreover, MBG infusion also induced endocytosis of RPT Na/K-ATPase in rats, which could be prevented by an antibody-mediated neutralization of infused MBG [156].

A high salt intake or volume expansion increases both dopamine and CTS. It has been shown that dopamine-induced regulation of RPT Na/K-ATPase of Dahl S rats was defective because of an apparent decoupling between the binding of dopamine to its D1 receptor and activation of GPCRs [157, 158, 159, 160, 161]. In response to salt loading, Dahl S rats have a similar diuretic, but much less CTS-related natriuretic response than that seen in Dahl R rats [162]. Both dopamine and CTS can regulate the activity and trafficking of RPT Na/K-ATPase and NHE3. Even though the initiating steps and signaling pathways might be different, they share some signaling steps such as the activation of PLC/PKC and calcium signaling. It will be of interest to further assess whether there is a crosstalk between CTS- and dopamine-activated signaling pathways in the regulation of renal Na+ handling.

In vivo studies suggest the essential role of CTS in modulating renal sodium excretion and BP with different approaches. First, the administration of some (e.g., ouabain) but not all CTS induces natriuresis [163, 164]. Second, in transgenic mice expressing ouabain-sensitive Na/K-ATPase α1 subunit, both acute salt load and ouabain infusion augment natriuretic responses, which were prevented by administration of an anti-digoxin antibody fragment [75, 76]. Third, immune neutralization of endogenous CTS prevents CTS-mediated natriuretic and vasoconstrictor effects [55, 59, 78, 80]. Fourth, the administration of the ouabain antagonist, rostafuroxin (also known as PST 2238), prevents not only ouabain-induced Na/K-ATPase signaling but also ouabain-induced increase in BP [64]. Finally, in humans, a high salt intake increases circulating endogenous CTS [57, 80, 165]. An increased CTS excretion is directly linked to an enhanced RPT-mediated fractional Na+ excretion, but inversely related to age and to age-dependent increase in salt sensitivity [165].

Although the historical focus has largely been on the direct inhibition of CTS on the Na/K-ATPase ion-pumping activity and sodium reabsorption in RPT as well as vascular tone/contractility, decreases in basolateral Na/K-ATPase activity alone do not appear to be sufficient to reduce net RPT sodium reabsorption since the apical NHE3, but not the Na/K-ATPase, is the rate-limiting step.

In contrast, the newly appreciated signaling function of Na/K-ATPase has been widely confirmed and provides a realistic, mechanistic framework that the renal Na/K-ATPase and its signaling play a key role in regulating renal sodium handling. In porcine RPT LLC-PK1 cells, ouabain activates the Na/K-ATPase signaling pathways and consequently redistributes the basolateral Na/K-ATPase and the apical NHE3 in a coordinated manner; this leads to a symmetrical reduction of cell surface Na/K-ATPase and NHE3 content and ultimately decreased net transcellular sodium transport [86, 87, 115, 116, 117]. No significant acute change in intracellular Na+ concentration was observed [115], further suggesting the coordination of the downregulation of both apical and basolateral sodium transporters. This Na/K-ATPase signaling-mediated regulation of renal tubular epithelial ion transporters was further confirmed in in vivo studies [79, 156]. It has been shown that endocytosis of signaling molecules could be a way to terminate or propagate the signaling and could further regulate endocytosis itself [166, 167, 168, 169, 170, 171]. In this regard, it is possible that ouabain- and ROS-induced endocytosis could be an effective way to terminate Na/K-ATPase signaling-mediated oxidant amplification loop by the degradation of carbonylated Na/K-ATPase, to maintain a certain basal level of ROS and carbonylated protein [172].

Advertisement

8. Endocytosis and signaling transduction

The clathrin-dependent endocytosis is the main endocytosis pathway for many membrane proteins in mammalian cells [166, 167, 173, 174, 175]. Apart from its endocytic function, the clathrin-coated pits also represent a specialized microdomain, where proteins are assembled into active signaling complexes before internalization of some or all of their components [176]. Some molecules involved in transmembrane signaling, such as β-arrestin, RGS-GAIP (a GTPase-activating protein for Gαi heterotrimeric G proteins) [177], GIPC (a PDZ domain-containing protein) [178], and Src family kinases [179], have been localized to clathrin-coated pits, suggesting that the interaction with the components of the pit machinery may facilitate some signaling functions of transmembrane receptors.

Caveolae/lipid rafts play a central role in transcytosis and endocytosis [180, 181, 182, 183, 184]. Many signaling molecules and membrane receptors are dynamically associated with caveolae, such as the Src family kinases, Ras, PKC, ERK, insulin receptor, platelet-derived growth factor receptor (PDGFR), EGFR, and some entire signaling modules like PDGFR-Ras-ERK, mainly through their interactions with caveolins [182, 185, 186]. Caveolins stabilize caveolae and modulate signal transduction by attracting signaling molecules to caveolae and regulating their activities [186]. There is also evidence that caveolins modulate endocytosis through their interactions with clathrin [187, 188, 189, 190]. Interestingly, both caveolin and clathrin heavy chain are substrates of Src kinase [169, 184].

The Na/K-ATPase α-subunit, c-Src, and caveolin are present in caveolae isolated by a detergent-free method, in adult rat cardiac myocytes, human embryonic kidney (HEK)-293 cells, and LLC-PK1 cells. In adult rat cardiac myocytes, ouabain not only recruits α-subunit and c-Src to caveolae but also activates caveolar ERK1/2 [191]. Furthermore, some signaling molecules, such as EGFR and c-Src, are also concentrated in clathrin-coated pits and endosomes in response to ouabain [116], suggesting that both clathrin-coated pits and caveolae are involved in ouabain-mediated Na/K-ATPase signal transduction and endocytosis.

The receptor-mediated endocytosis has been shown not only to attenuate ligand-activated signaling but also to continue the signaling on the endocytic pathway, especially from endosomes [166, 167, 192, 193, 194]. While endocytosis is important in the activation and propagation of signaling pathways [168, 195, 196], signal transduction can also regulate endocytosis [169, 197]. Endocytic receptor tyrosine kinase (RTK) receptors could control the magnitude of the original signaling responses (generated at the cell surface) or initiate distinct signaling cascades (qualitatively different from that generated at the cell surface) [170]. In polarized epithelial cells, the distribution of RTK substrates could affect cellular responses [118]. The endosomal signaling appears to be dependent on both the receptor and cell type.

In LLC-PK1 cells, ouabain not only induced compartmentalization of Na/K-ATPase, c-Src, EGF receptor, and ERK in early endosomes but also bound to Na/K-ATPase along the endocytic route [116]. Interestingly, caveolin-1 is also present in early or late endosomes. These facts make it possible that endosomal ouabain-Na/K-ATPase/c-Src might be able to propagate its original signaling or to initiate distinct signaling cascades. This is supported by the findings that ouabain-induced NHE3 regulation is mediated by the activation of the receptor function of Na/K-ATPase. Furthermore, endocytosis is required for ouabain to remove basolateral Na/K-ATPase, which induces a significant inhibition of the pumping activity. Moreover, blockade of Na/K-ATPase signaling/endocytosis appears to be sufficient to abolish ouabain-induced trafficking and transcriptional regulation of NHE3.

Although the mechanisms that involved ouabain-initiated endocytosis of the Na/K-ATPase and NHE3 (and expression) are not fully understood, endocytosis of the Na/K-ATPase may play an important role in renal sodium handling. This is because if ouabain induces a significant depletion of plasmalemmal Na/K-ATPase in proximal tubule type cells (rat proximal tubule primary culture, LLC-PK1) but not in distal tubule type cells (rat distal tubule primary culture, MDCK), it will make physiological “sense” in terms of allowing bulk sodium transport (primarily in the proximal tubule) to be altered and leaving fine-tuning (distal tubule) sodium handling intact.

Advertisement

9. ROS and the Na/K-ATPase signaling: the possible link from CTS-stimulated signaling to NHE3 regulation

It is well established that both oxidative stress and high BP are a cause and consequence of each other. The increase in oxidative stress occurs in many forms of experimental models of hypertension, including Dahl salt-sensitive hypertension [198, 199, 200, 201, 202, 203, 204]. Increases in ROS can regulate physiological processes including renal tubular ion transport, fluid reabsorption, and sodium excretion [79, 205, 206, 207, 208, 209, 210]. In particular, increases in ROS regulate the activity and cellular distribution of the basolateral Na/K-ATPase as well as the apical NHE3 and sodium/glucose cotransporter, at least under normal circumstances [79, 151, 208, 211, 212, 213, 214, 215, 216]. Oxidative modification can affect the Na/K-ATPase activity through different mechanisms. For example, S-glutathionylation cysteine residue(s) of the Na/K-ATPase α-subunit can block the intracellular ATP-binding site [217], and S-glutathionylation of cysteine of the Na/K-ATPase β1-subunit can affect the Na/K-ATPase conformational poise [218, 219]. Oxidant and oxidative modification of the Na/K-ATPase can lead to degradation, functional changes, and formation of Na/K-ATPase oligomeric structure [74, 84, 85, 86, 87, 217, 219, 220, 221, 222, 223, 224, 225, 226, 227, 228, 229, 230]. In LLC-PK1 cells, increase in ROS generation, induced by either ouabain or glucose oxidase, is critical in the activation of Na/K-ATPase signaling which mediates trafficking of the Na/K-ATPase and NHE3 and transcellular Na+ transport [86, 87]. Pretreatment with higher doses, but not a low dose, of NAC attenuated the effect of ouabain on c-Src activation and transcellular 22Na+ flux, suggesting a role of basal physiological redox status in the initiation of ouabain-induced Na/K-ATPase signaling. While CTS stimulates ROS generation and Na/K-ATPase signaling in different in vitro and in vivo models [63, 85, 231, 232, 233], an increase in ROS alone (without the presence of ouabain) by extracellularly added glucose oxidase is also able to activate Na/K-ATPase signaling, indicating that activation of Na/K-ATPase signaling can be achieved by general stimuli like ROS, other than its specific ligands. Glucose oxidase-induced H2O2 alone also stimulates Na/K-ATPase endocytosis and inhibits active transcellular 22Na+ transport [85, 86]. The phenomenon of redox sensitivity of the Na/K-ATPase has been demonstrated in different cell types, tissues, and animal species.

In LLC-PK1 cells, both ouabain and glucose oxidase-induced H2O2 stimulate Na/K-ATPase signaling as well as direct protein carbonylation of Pro222 and Thr224 residues of the Na/K-ATPase α1 subunit (α1-carbonylation) [86]. The Pro222 and Thr224 are located in peptide 211VDNSSLTGESEPQTR225 [UniProtKB/Swiss-Prot No P05024 (AT1A1_PIG)]. While the α1 subunit is highly conserved among humans, pigs, rats, and mice (the homology is over 98.5%), the identified peptide is 100% identical among these four species. This peptide is located in the actuator (A) domain of α1 subunit, and Pro222/Thr224 are highly exposed and facing the nucleotide binding (N) domain of the α1 subunit. Upon ouabain binding, Na/K-ATPase undergoes conformational changes, in which the A domain is rotated to the N domain favoring an E2-P conformation. The structure-function analysis indicates that these conformational changes may affect binding of the α1 subunit to signaling molecules such as c-Src and PI3K [234]. In addition, the peptide also contains the TGES motif that is the anchor of A domain rotation [234].

Biologically, ROS can oxidize various types of biological molecules including proteins, leading to their functional changes. Through Fenton’s reaction, H2O2 is reduced to HO by coupling oxidation of reduced ferrous ion (Fe2+) to ferric ion (Fe3+). This metal-catalyzed oxidation (MCO) process oxidizes proteins by introducing carbonyl groups (such as aldehydes, ketones, or lactams) into the side chains of certain amino acids (such as proline, arginine, lysine, and threonine) that named direct (primary) carbonylation that have been implied in various conditions like chronic renal failure [235, 236, 237, 238, 239, 240]. Since Fenton’s reaction involves the conversion of H2O2 to HO, any specie of ROS with H2O2 as an intermediate and/or end product may stimulate the reaction.

Protein carbonylation is reversible (decarbonylation) and may function as a regulatory mechanism of cell signaling [241, 242, 243, 244]. We also observed an undefined decarbonylation mechanism, which apparently reverses the carbonylation of the Na/K-ATPase α1 subunit induced by ouabain [86]. The removal of ouabain from the culture medium reverses ouabain-mediated carbonylation, as seen in the reversed Na/K-ATPase ion-pumping activity [116]. Moreover, inhibition of de novo protein synthesis as well as degradation pathway through lysosome and proteasome does not affect this decarbonylation, which is still poorly understood. It is possible that carbonylation modification might stabilize the Na/K-ATPase in a certain conformational status favoring ouabain binding to the Na/K-ATPase α1 subunit and ouabain-Na/K-ATPase signaling. Nevertheless, the underlying mechanism might be physiologically significant since the carbonylation/decarbonylation process could be an important regulator of the RPT Na/K-ATPase signaling and sodium handling.

It is reasonable to propose that carbonylation modification of RPT Na/K-ATPase α1 subunit has biphasic effects. On one hand, physiological and controllable α1-carbonylation stimulates Na/K-ATPase signaling and sodium excretion, rendering salt resistance, whereas on the other hand, prolonged exposure to oxidant stress leads to overstimulated α1-carbonylation and desensitized Na/K-ATPase signaling, increasing salt sensitivity. First, Dahl S rats show considerably higher basal levels of oxidative stress than R rats, and high-salt diets increase renal oxidative stresses that contribute to salt-sensitive hypertension [202, 203, 204]. Second, while high-salt diets increase circulating CTS, a high-salt diet (HS, 2% NaCl for 7 days) stimulates the Na/K-ATPase signaling in isolated RPT from Dahl salt-resistant (R) but not salt-sensitive (S) rats (i.e., impaired Na/K-ATPase signaling in S rats) [79]. Third, CTS- and H2O2-mediated redox-sensitive Na/K-ATPase signaling and α1-carbonylation are involved in this signaling process, in a feed-forward mechanism [86]. Fourth, high but not low concentration of NAC is able to prevent α1-carbonylation and Na/K-ATPase signaling [86]. Even though it is still not clear of the carbonylation/decarbonylation process, this could be another new regulatory mechanism of Na/K-ATPase signaling. It is reasonable to postulate that prolonged excessive α1-carbonylation (by CTS and/or other factors) might overcome the decarbonylation capacity, leading to the desensitization or termination of the Na/K-ATPase signaling function. This is reminiscent of the observations in clinical trials using antioxidant supplements. The beneficial effect of antioxidant supplements is controversial and not seen in most clinical trials with administration of antioxidant supplements [200, 245]. Low doses of antioxidant supplementation may be ineffective, but high doses may be even dangerous since excess antioxidants might become prooxidants if they cannot promptly be reduced in the antioxidant chain [246]. It appears that the balance of the redox status, within a physiological range, may be critical in order to maintain beneficial ROS signaling.

Advertisement

10. Endocytosis of Na/K-ATPase and NHE3 in salt sensitivity

In male Sprague-Dawley rats, compared to a normal salt (0.4% NaCl, 7 days) diet, a high-salt (4% NaCl, 7 days) diet increased urinary sodium and MBG excretion. In isolated proximal tubules, a high-salt diet inhibits the Na/K-ATPase ion-exchange activity and enzymatic activity, which is accompanied by a decreased Na/K-ATPase α1 content in heavy membrane fraction and an increased Na/K-ATPase α1 content in both early and late endosomes. These high-salt diet-mediated changes were ameliorated by administration of an antibody against MBG [156]. Results indicate that a high-salt diet increased MBG production, activated RPT Na/K-ATPase signaling, and induced endocytosis of Na/K-ATPase.

The Dahl R and S rat strains were developed from Sprague-Dawley rats by selective breeding, depending on the resistance or susceptibility to the hypertensive effects of high dietary sodium [247]. In these two strains, the RPT sodium handling is an essential determinant of their different BP responses [248, 249, 250, 251]. At the cost of elevated systolic BP, Dahl S rats get rid of excess sodium primarily via pressure natriuresis. In contrast, Dahl R rats get rid of excess sodium primarily via a significant reduction of renal sodium reabsorption without increasing the BP. In vivo study indicates that impaired RPT Na/K-ATPase signaling appears to be causative of experimental Dahl salt sensitivity [79]. In vivo studies with Dahl R and S rats (Jr strains) demonstrated that impairment of RPT Na/K-ATPase signaling is a causative factor of experimental Dahl salt sensitivity [79]. In Dahl R but not S rats, a high-salt (2% NaCl, 1 week) diet activated RPT Na/K-ATPase signaling and stimulated coordinated redistribution of the Na/K-ATPase and NHE3, leading to increased total and fractional urinary sodium excretion as well as normal BP. However, there are still questions about the underlying mechanism(s) that need to be further investigated, such as the difference of Na/K-ATPase signaling function between Dahl R and S rats, as well as the translation of Na/K-ATPase signaling to NHE3 regulation. Furthermore, low concentration of ouabain causes hypertrophic response both in the heart and kidney, by concentrating the Na/K-ATPase, Src, EGFR, and MAPKs within rat caveolae, and activates the Na/K-ATPase/Src/MAPK signaling pathway [64]. However, there is no simple explanation for this occurrence. First, the α1 subunit is essentially the only α isoform expressed in RPT, and genes coding α1 subunit and NHE3 (in rat chromosomes 1 and 2, respectively) are not located in identified and/or proposed BP quantitative trait loci [252]. Second, there is no difference in α1 gene (Atp1a1) coding [251], α1 ouabain sensitivity [253], and α1 expression [79] between these two strains. Third, acute salt loading increases circulating CTS (ouabain and MBG) in both S and R rats [162]. These observations suggest that there must be resistance to CTS signaling in the Dahl S rat, a phenomenon that we only partially understand. As discussed above, the carbonylation/decarbonylation process could be another new regulatory mechanism of Na/K-ATPase signaling. It is reasonable to postulate that prolonged excessive α1-carbonylation in Dahl salt-sensitive rats might overcome the decarbonylation capacity, leading to desensitization or termination of the Na/K-ATPase signaling function.

11. Perspective

As pointed out by Guyton many years ago [254], the kidney is the most important organ in the regulation of Na+ handling and BP. Dietary salt intake vs. renal sodium handling is a key determinant of long-term BP regulation and plays an important role in the pathogenesis of hypertension, with more pronounced effects seen in salt-sensitive patients. Consequently, modest restriction of dietary salt and diuretic therapy are often recommended for the treatment of resistant hypertension, particularly with the salt-sensitive subgroup [254, 255, 256, 257, 258].

Although the relationships among CTS, renal Na+ handling, and hypertension were proposed many years ago, there has been an explosion of reports supporting this idea. As discussed, reports from Lingrel’s laboratory clearly demonstrated a specific role of the isoforms of the Na/K-ATPase and its interaction with endogenous CTS in the regulation of Na+ excretion and BP in intact animals [75, 76, 77]. From the ligand perspective, studies have demonstrated that CTS are present in measurable amounts under normal physiological conditions and that several disease states are associated with elevations in the circulating levels of CTS. The new concept that the Na/K-ATPase has an ion-pumping-independent receptor function (induced by both CTS and ROS) that can confer the agonist-like effects of CTS on intracellular signal transduction is a new mechanism for RPT sodium handling. Moreover, this newly discovered signaling mechanism operates in intact animals in response to CTS stimulation. The Na/K-ATPase has recently emerged as a therapeutic target [259, 260]. A clearer understanding of the mechanisms, in which a CTS-ROS-Na/K-ATPase signaling axis counterbalancing salt retention, would not only have major pathophysiological and therapeutic implications, but also further explain the progressive impairment of renal sodium handling under excessive oxidative stresses such as hypertension, aging, obesity, and diabetes.

Funding

This work was supported by NIH R15 1R15DK106666 (to J. Liu) and NIH RO1 HL071556 (to J.I. Shapiro).

Abbreviations

BPblood pressure
CTScardiotonic steroids
NHE3sodium/hydrogen exchanger isoform 3
ROSreactive oxygen species
RPTrenal proximal tubule

References

  1. 1. Skou JC. The influence of some cations on an adenosine triphosphatase from peripheral nerves. Biochimica et Biophysica Acta. 1957;23:394-401
  2. 2. Curthoys NP, Moe OW. Proximal tubule function and response to acidosis. Clinical Journal of the American Society of Nephrology. 2014;9:1627-1638
  3. 3. Boron WF, Boulpaep EL. Medical Physiology: A Cellular and Molecular Approach, Updated Second Edition. Philadelphia, PA: Saunders/Elsevier; 2012
  4. 4. Muto S, Hata M, Taniguchi J, Tsuruoka S, Moriwaki K, Saitou M, et al. Claudin-2–deficient mice are defective in the leaky and cation-selective paracellular permeability properties of renal proximal tubules. Proceedings of the National Academy of Sciences. 2010;107:8011-8016
  5. 5. Fromm M, Piontek J, Rosenthal R, Günzel D, Krug SM. Tight junctions of the proximal tubule and their channel proteins. Pflügers Archiv - European Journal of Physiology. 2017;469:877-887
  6. 6. Muto S, Furuse M, Kusano E. Claudins and renal salt transport. Clinical and Experimental Nephrology. 2012;16:61-67
  7. 7. Cereijido M, Contreras RG, Shoshani L, Larre I. The Na+-K+-ATPase as self-adhesion molecule and hormone receptor. American Journal of Physiology-Cell Physiology. 2012;302:C473-C481
  8. 8. Rajasekaran AK, Rajasekaran SA. Role of Na-K-ATPase in the assembly of tight junctions. American Journal of Physiology. Renal Physiology. 2003;285:F388-F396
  9. 9. Sweadner KJ. Isozymes of the Na+/K+-ATPase. Biochimica et Biophysica Acta. 1989;988:185-220
  10. 10. Kaplan JH. Biochemistry of Na,K-ATPase. Annual Review of Biochemistry. 2002;71:511-535
  11. 11. Blanco G, Mercer RW. Isozymes of the Na-K-ATPase: Heterogeneity in structure, diversity in function. The American Journal of Physiology. 1998;275:F633-F650
  12. 12. Sanchez G, Nguyen AN, Timmerberg B, Tash JS, Blanco G. The Na,K-ATPase alpha4 isoform from humans has distinct enzymatic properties and is important for sperm motility. Molecular Human Reproduction. 2006;12:565-576
  13. 13. Geering K. FXYD proteins: New regulators of Na-K-ATPase. American Journal of Physiology. Renal Physiology. 2006;290:F241-F250
  14. 14. Arystarkhova E, Wetzel RK, Sweadner KJ. Distribution and oligomeric association of splice forms of Na+-K+-ATPase regulatory γ-subunit in rat kidney. American Journal of Physiology. Renal Physiology. 2002;282:F393-F407
  15. 15. Mercer RW, Biemesderfer D, Bliss DP Jr, Collins JH, Forbush B 3rd. Molecular cloning and immunological characterization of the gamma polypeptide, a small protein associated with the Na,K-ATPase. Journal of Cell Biology. 1993;121:579-586
  16. 16. Pestov NB, Ahmad N, Korneenko TV, Zhao H, Radkov R, Schaer D, et al. Evolution of Na,K-ATPase βm-subunit into a coregulator of transcription in placental mammals. Proceedings of the National Academy of Sciences. 2007;104:11215-11220
  17. 17. Barwe SP, Anilkumar G, Moon SY, Zheng Y, Whitelegge JP, Rajasekaran SA, et al. Novel role for Na,K-ATPase in phosphatidylinositol 3-kinase signaling and suppression of cell motility. Molecular Biology of the Cell. 2005;16:1082-1094
  18. 18. Beggah AT, Geering K. Alpha and beta subunits of Na,K-ATPase interact with BiP and calnexin. Annals of the New York Academy of Sciences. 1997;834:537-539
  19. 19. Feschenko MS, Wetzel RK, Sweadner KJ. Phosphorylation of Na,K-ATPase by protein kinases. Sites, susceptibility, and consequences. Annals of the New York Academy of Sciences. 1997;834:479-488
  20. 20. Jordan C, Puschel B, Koob R, Drenckhahn D. Identification of a binding motif for ankyrin on the alpha-subunit of Na+,K(+)-ATPase. The Journal of Biological Chemistry. 1995;270:29971-29975
  21. 21. Lee K, Jung J, Kim M, Guidotti G. Interaction of the alpha subunit of Na,K-ATPase with cofilin. The Biochemical Journal. 2001;353:377-385
  22. 22. Xie Z, Cai T. Na+-K+--ATPase-mediated signal transduction: From protein interaction to cellular function. Molecular Interventions. 2003;3:157-168
  23. 23. Tian J, Cai T, Yuan Z, Wang H, Liu L, Haas M, et al. Binding of Src to Na+/K+-ATPase forms a functional signaling complex. Molecular Biology of the Cell. 2006;17:317-326
  24. 24. Yudowski GA, Efendiev R, Pedemonte CH, Katz AI, Berggren PO, Bertorello AM. Phosphoinositide-3 kinase binds to a proline-rich motif in the Na+, K+-ATPase alpha subunit and regulates its trafficking. Proceedings of the National Academy of Sciences of the United States of America. 2000;97:6556-6561
  25. 25. Zhang Z, Devarajan P, Dorfman AL, Morrow JS. Structure of the ankyrin-binding domain of alpha-Na,K-ATPase. The Journal of Biological Chemistry. 1998;273:18681-18684
  26. 26. Song H, Lee MY, Kinsey SP, Weber DJ, Blaustein MP. An N-terminal sequence targets and tethers Na+ pump alpha2 subunits to specialized plasma membrane microdomains. The Journal of Biological Chemistry. 2006;281:12929-12940
  27. 27. Zhang S, Malmersjo S, Li J, Ando H, Aizman O, Uhlen P, et al. Distinct role of the N-terminal tail of the Na,K-ATPase catalytic subunit as a signal transducer. The Journal of Biological Chemistry. 2006;281:21954-21962
  28. 28. Kaplan JH. A moving new role for the sodium pump in epithelial cells and carcinomas. Science’s STKE. 2005, 2005:pe31
  29. 29. Kaunitz JD. Membrane transport proteins: not just for transport anymore. American Journal of Physiology. Renal Physiology. 2006;290:F995-F996
  30. 30. Schoner W, Scheiner-Bobis G. Endogenous and exogenous cardiac glycosides: Their roles in hypertension, salt metabolism, and cell growth. American Journal of Physiology. Cell Physiology. 2007;293:C509-C536
  31. 31. Li Z, Xie Z. The Na/K-ATPase/Src complex and cardiotonic steroid-activated protein kinase cascades. Pflügers Archiv. 2009;457:635-644
  32. 32. Xie Z, Askari A. Na(+)/K(+)-ATPase as a signal transducer. European Journal of Biochemistry. 2002;269:2434-2439
  33. 33. Donowitz M, Li X. Regulatory binding partners and complexes of NHE3. Physiological Reviews. 2007;87:825-872
  34. 34. Alexander RT, Grinstein S. Tethering, recycling and activation of the epithelial sodium-proton exchanger, NHE3. The Journal of Experimental Biology. 2009;212:1630-1637
  35. 35. Bobulescu IA, Moe OW. Luminal Na(+)/H (+) exchange in the proximal tubule. Pflügers Archiv. 2009;458:5-21
  36. 36. Amemiya M, Loffing J, Lotscher M, Kaissling B, Alpern RJ, Moe OW. Expression of NHE-3 in the apical membrane of rat renal proximal tubule and thick ascending limb. Kidney International. 1995;48:1206-1215
  37. 37. Biemesderfer D, Pizzonia J, Abu-Alfa A, Exner M, Reilly R, Igarashi P, et al. NHE3: A Na+/H+ exchanger isoform of renal brush border. The American Journal of Physiology. 1993;265:F736-F742
  38. 38. Alpern RJ. Cell mechanisms of proximal tubule acidification. Physiological Reviews. 1990;70:79-114
  39. 39. Aronson PS. Mechanisms of active H+ secretion in the proximal tubule. American Journal of Physiology. Renal Physiology. 1983;245:F647-F659
  40. 40. Hamm LL, Nakhoul N, Hering-Smith KS. Acid-Base Homeostasis. Clinical Journal of the American Society of Nephrology. 2015;10:2232-2242
  41. 41. D’Souza S, Garcia-Cabado A, Yu F, Teter K, Lukacs G, Skorecki K, et al. The epithelial sodium-hydrogen antiporter Na+/H+ exchanger 3 accumulates and is functional in recycling endosomes. The Journal of Biological Chemistry. 1998;273:2035-2043
  42. 42. Gekle M, Freudinger R, Mildenberger S. Inhibition of Na+-H+ exchanger-3 interferes with apical receptor-mediated endocytosis via vesicle fusion. The Journal of Physiology. 2001;531:619-629
  43. 43. Schoner W. Endogenous cardiac glycosides, a new class of steroid hormones. European Journal of Biochemistry. 2002;269:2440-2448
  44. 44. Schoner W, Scheiner-Bobis G. Endogenous and exogenous cardiac glycosides and their mechanisms of action. American Journal of Cardiovascular Drugs. 2007;7:173-189
  45. 45. Schoner W, Scheiner-Bobis G. Role of endogenous cardiotonic steroids in sodium homeostasis. Nephrology Dialysis Transplantation. 2008;23:2723-2729
  46. 46. Hamlyn JM, Blaustein MP, Bova S, DuCharme DW, Harris DW, Mandel F, et al. Identification and characterization of a ouabain-like compound from human plasma. Proceedings of the National Academy of Sciences of the United States of America. 1991;88:6259-6263
  47. 47. Bagrov AY, Shapiro JI, Fedorova OV. Endogenous cardiotonic steroids: Physiology, pharmacology, and novel therapeutic targets. Pharmacological Reviews. 2009;61:9-38
  48. 48. Laredo J, Shah JR, Lu ZR, Hamilton BP, Hamlyn JM. Angiotensin II stimulates secretion of endogenous ouabain from bovine adrenocortical cells via angiotensin type 2 receptors. Hypertension. 1997;29:401-407
  49. 49. Blaustein MP. Physiological effects of endogenous ouabain: Control of intracellular Ca2+ stores and cell responsiveness. The American Journal of Physiology. 1993;264:C1367-C1387
  50. 50. Hamlyn JM, Lu ZR, Manunta P, Ludens JH, Kimura K, Shah JR, et al. Observations on the nature, biosynthesis, secretion and significance of endogenous ouabain. Clinical and Experimental Hypertension. 1998;20:523-533
  51. 51. Gottlieb SS, Rogowski AC, Weinberg M, Krichten CM, Hamilton BP, Hamlyn JM. Elevated concentrations of endogenous ouabain in patients with congestive heart failure. Circulation. 1992;86:420-425
  52. 52. Hasegawa T, Masugi F, Ogihara T, Kumahara Y. Increase in plasma ouabainlike inhibitor of Na+, K+-ATPase with high sodium intake in patients with essential hypertension. Journal of Clinical Hypertension. 1987;3:419-429
  53. 53. Fedorova OV, Doris PA, Bagrov AY. Endogenous marinobufagenin-like factor In acute plasma volume expansion. Clinical and Experimental Hypertension. 1998;20:581-591
  54. 54. Rossi G, Manunta P, Hamlyn JM, Pavan E, De Toni R, Semplicini A, et al. Immunoreactive endogenous ouabain in primary aldosteronism and essential hypertension: Relationship with plasma renin, aldosterone and blood pressure levels. Journal of Hypertension. 1995;13:1181-1191
  55. 55. Fedorova OV, Talan MI, Agalakova NI, Lakatta EG, Bagrov AY. Endogenous ligand of alpha(1) sodium pump, marinobufagenin, is a novel mediator of sodium chloride--dependent hypertension. Circulation. 2002;105:1122-1127
  56. 56. Komiyama Y, Dong XH, Nishimura N, Masaki H, Yoshika M, Masuda M, et al. A novel endogenous digitalis, telocinobufagin, exhibits elevated plasma levels in patients with terminal renal failure. Clinical Biochemistry. 2005;38:36-45
  57. 57. Manunta P, Hamilton BP, Hamlyn JM. Salt intake and depletion increase circulating levels of endogenous ouabain in normal men. American Journal of Physiology. Regulatory, Integrative and Comparative Physiology. 2006;290:R553-R559
  58. 58. Manunta P, Stella P, Rivera R, Ciurlino D, Cusi D, Ferrandi M, et al. Left ventricular mass, stroke volume, and ouabain-like factor in essential hypertension. Hypertension. 1999;34:450-456
  59. 59. Fedorova OV, Kolodkin NI, Agalakova NI, Lakatta EG, Bagrov AY. Marinobufagenin, an endogenous alpha-1 sodium pump ligand, in hypertensive Dahl salt-sensitive rats. Hypertension. 2001;37:462-466
  60. 60. Fedorova OV, Agalakova NI, Talan MI, Lakatta EG, Bagrov AY. Brain ouabain stimulates peripheral marinobufagenin via angiotensin II signalling in NaCl-loaded Dahl-S rats. Journal of Hypertension. 2005;23:1515-1523
  61. 61. Haddy FJ, Pamnani MB. Role of ouabain-like factors and Na-K-ATPase inhibitors in hypertension--some old and recent findings. Clinical and Experimental Hypertension. 1998;20:499-508
  62. 62. Ferrari P, Ferrandi M, Valentini G, Bianchi G. Rostafuroxin: An ouabain antagonist that corrects renal and vascular Na+-K+- ATPase alterations in ouabain and adducin-dependent hypertension. American Journal of Physiology. Regulatory, Integrative and Comparative Physiology. 2006;290:R529-R535
  63. 63. Kennedy DJ, Vetteth S, Periyasamy SM, Kanj M, Fedorova L, Khouri S, et al. Central role for the cardiotonic steroid marinobufagenin in the pathogenesis of experimental uremic cardiomyopathy. Hypertension. 2006;47:488-495
  64. 64. Ferrandi M, Molinari I, Barassi P, Minotti E, Bianchi G, Ferrari P. Organ hypertrophic signaling within caveolae membrane subdomains triggered by ouabain and antagonized by PST 2238. The Journal of Biological Chemistry. 2004;279:33306-33314
  65. 65. Jiang X, Ren YP, Lv ZR. Ouabain induces cardiac remodeling in rats independent of blood pressure. Acta Pharmacologica Sinica. 2007;28:344-352
  66. 66. Skoumal R, Szokodi I, Aro J, Foldes G, Gooz M, Seres L, et al. Involvement of endogenous ouabain-like compound in the cardiac hypertrophic process in vivo. Life Sciences. 2007;80:1303-1310
  67. 67. Bricker NS. The control of sodium excretion with normal and reduced nephron populations. The pre-eminence of third factor. The American Journal of Medicine. 1967;43:313-321
  68. 68. Dahl LK, Knudsen KD, Iwai J. Humoral transmission of hypertension: Evidence from parabiosis. Circulation Research. 1969;24:21-33
  69. 69. Bricker NS, Klahr S, Lubowitz H, Slatopolsky E. The pathophysiology of renal insufficiency. On the functional transformations in the residual nephrons with advancing disease. Pediatric Clinics of North America. 1971;18:595-611
  70. 70. Haddy FJ, Overbeck HW. The role of humoral agents in volume expanded hypertension. Life Sciences. 1976;19:935-947
  71. 71. De Wardener HE. Natriuretic hormone. Clinical Science and Molecular Medicine. 1977;53:1-8
  72. 72. Blaustein MP. Sodium ions, calcium ions, blood pressure regulation, and hypertension: A reassessment and a hypothesis. The American Journal of Physiology. 1977;232:C165-C173
  73. 73. de Wardener HE, MacGregor GA. Dahl’s hypothesis that a saluretic substance may be responsible for a sustained rise in arterial pressure: Its possible role in essential hypertension. Kidney International. 1980;18:1-9
  74. 74. Blaustein MP, Zhang J, Chen L, Song H, Raina H, Kinsey SP, et al. The pump, the exchanger, and endogenous ouabain: Signaling mechanisms that link salt retention to hypertension. Hypertension. 2009;53:291-298
  75. 75. Dostanic-Larson I, Van Huysse JW, Lorenz JN, Lingrel JB. The highly conserved cardiac glycoside binding site of Na,K-ATPase plays a role in blood pressure regulation. Proceedings of the National Academy of Sciences of the United States of America. 2005;102:15845-15850
  76. 76. Loreaux EL, Kaul B, Lorenz JN, Lingrel JB. Ouabain-sensitive alpha1 Na,K-ATPase enhances natriuretic response to saline load. Journal of the American Society of Nephrology. 2008;19:1947-1954
  77. 77. Dostanic I, Paul RJ, Lorenz JN, Theriault S, Van Huysse JW, Lingrel JB. The alpha2-isoform of Na-K-ATPase mediates ouabain-induced hypertension in mice and increased vascular contractility in vitro. American Journal of Physiology. Heart and Circulatory Physiology. 2005;288:H477-H485
  78. 78. Nesher M, Dvela M, Igbokwe VU, Rosen H, Lichtstein D. Physiological roles of endogenous ouabain in normal rats. American Journal of Physiology. Heart and Circulatory Physiology. 2009;297:H2026-H2034
  79. 79. Liu J, Yan Y, Liu L, Xie Z, Malhotra D, Joe B, et al. Impairment of Na/K-ATPase signaling in renal proximal tubule contributes to Dahl salt-sensitive hypertension. The Journal of Biological Chemistry. 2011;286:22806-22813
  80. 80. Bagrov AY, Shapiro JI. Endogenous digitalis: Pathophysiologic roles and therapeutic applications. Nature Clinical Practice. Nephrology. 2008;4:378-392
  81. 81. Liu J, Xie ZJ. The sodium pump and cardiotonic steroids-induced signal transduction protein kinases and calcium-signaling microdomain in regulation of transporter trafficking. Biochimica et Biophysica Acta. 2010;1802:1237-1245
  82. 82. Tian J, Xie ZJ. The Na-K-ATPase and calcium-signaling microdomains. Physiology (Bethesda). 2008;23:205-211
  83. 83. Cui X, Xie Z. Protein interaction and Na/K-ATPase-mediated signal transduction. Molecules. 2017;22(6):990
  84. 84. Wang Y, Ye Q , Liu C, Xie JX, Yan Y, Lai F, et al. Involvement of Na/K-ATPase in hydrogen peroxide-induced activation of the Src/ERK pathway in LLC-PK1 cells. Free Radical Biology & Medicine. 2014;71:415-426
  85. 85. Liu L, Li J, Liu J, Yuan Z, Pierre SV, Qu W, et al. Involvement of Na+/K+-ATPase in hydrogen peroxide-induced hypertrophy in cardiac myocytes. Free Radical Biology & Medicine. 2006;41:1548-1556
  86. 86. Yan Y, Shapiro AP, Haller S, Katragadda V, Liu L, Tian J, et al. Involvement of reactive oxygen species in a feed-forward mechanism of Na/K-ATPase-mediated signaling transduction. The Journal of Biological Chemistry. 2013;288:34249-34258
  87. 87. Yan Y, Shapiro AP, Mopidevi BR, Chaudhry MA, Maxwell K, Haller ST, et al. Protein carbonylation of an amino acid residue of the Na/K-ATPase alpha1 subunit determines Na/K-ATPase signaling and sodium transport in renal proximal tubular cells. Journal of the American Heart Association. 2016;5(9):e003675
  88. 88. Cook JS, Tate EH, Shaffer C. Uptake of [3H]ouabain from the cell surface into the lysosomal compartment of HeLa cells. Journal of Cellular Physiology. 1982;110:84-92
  89. 89. Algharably N, Owler D, Lamb JF. The rate of uptake of cardiac glycosides into human cultured cells and the effects of chloroquine on it. Biochemical Pharmacology. 1986;35:3571-3581
  90. 90. Lamb JF, McCall D. Uptake of (3H)ouabain and Na pump turnover rates in monolayer cultures of Girardi heart cells. The Journal of Physiology. 1971;213:57P-58P
  91. 91. Lamb JF, Ogden P. Internalization of ouabain and replacement of sodium pumps in the plasma membranes of HeLa cells following block with cardiac glycosides. Quarterly Journal of Experimental Physiology. 1982;67:105-119
  92. 92. Aiton JF, Lamb JF, Ogden P. Down-regulation of the sodium pump following chronic exposure of HeLa cells and chick embryo heart cells to ouabain. British Journal of Pharmacology. 1981;73:333-340
  93. 93. Aperia AC. Intrarenal dopamine: A key signal in the interactive regulation of sodium metabolism. Annual Review of Physiology. 2000;62:621-647
  94. 94. Zeng C, Sanada H, Watanabe H, Eisner GM, Felder RA, Jose PA. Functional genomics of the dopaminergic system in hypertension. Physiological Genomics. 2004;19:233-246
  95. 95. Hussain T, Lokhandwala MF. Renal dopamine receptors and hypertension. Experimental Biology and Medicine (Maywood, N.J.). 2003;228:134-142
  96. 96. McDonald RH Jr, Goldberg LI, McNay JL, Tuttle EP Jr. Effect of dopamine in man: Augmentation of sodium excretion, glomerular filtration rate, and renal plasma flow. The Journal of Clinical Investigation. 1964;43:1116-1124
  97. 97. Jose PA, Eisner GM, Felder RA. Role of dopamine receptors in the kidney in the regulation of blood pressure. Current Opinion in Nephrology and Hypertension. 2002;11:87-92
  98. 98. Wang X, Villar VA, Armando I, Eisner GM, Felder RA, Jose PA. Dopamine, kidney, and hypertension: Studies in dopamine receptor knockout mice. Pediatric Nephrology. 2008;23:2131-2146
  99. 99. Bertorello A, Aperia A. Both DA1 and DA2 receptor agonists are necessary to inhibit NaKATPase activity in proximal tubules from rat kidney. Acta Physiologica Scandinavica. 1988;132:441-443
  100. 100. Bertorello A, Aperia A. Inhibition of proximal tubule Na(+)-K(+)-ATPase activity requires simultaneous activation of DA1 and DA2 receptors. The American Journal of Physiology. 1990;259:F924-F928
  101. 101. Bertorello A, Hokfelt T, Goldstein M, Aperia A. Proximal tubule Na+-K+-ATPase activity is inhibited during high-salt diet: Evidence for DA-mediated effect. The American Journal of Physiology. 1988;254:F795-F801
  102. 102. Chibalin AV, Katz AI, Berggren PO, Bertorello AM. Receptor-mediated inhibition of renal Na(+)-K(+)-ATPase is associated with endocytosis of its alpha- and beta-subunits. The American Journal of Physiology. 1997;273:C1458-C1465
  103. 103. Moe OW. Acute regulation of proximal tubule apical membrane Na/H exchanger NHE-3: Role of phosphorylation, protein trafficking, and regulatory factors. Journal of the American Society of Nephrology. 1999;10:2412-2425
  104. 104. Bacic D, Kaissling B, McLeroy P, Zou L, Baum M, Moe OW. Dopamine acutely decreases apical membrane Na/H exchanger NHE3 protein in mouse renal proximal tubule. Kidney International. 2003;64:2133-2141
  105. 105. Hu MC, Fan L, Crowder LA, Karim-Jimenez Z, Murer H, Moe OW. Dopamine acutely stimulates Na+/H+ exchanger (NHE3) endocytosis via clathrin-coated vesicles: Dependence on protein kinase A-mediated NHE3 phosphorylation. The Journal of Biological Chemistry. 2001;276:26906-26915
  106. 106. Chibalin AV, Ogimoto G, Pedemonte CH, Pressley TA, Katz AI, Feraille E, et al. 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. The Journal of Biological Chemistry. 1999;274:1920-1927
  107. 107. Chibalin AV, Zierath JR, Katz AI, Berggren P-O, Bertorello AM. Phosphatidylinositol 3-kinase-mediated endocytosis of renal Na+,K+-ATPase alpha subunit in response to dopamine. Molecular Biology of the Cell. 1998;9:1209-1220
  108. 108. Done SC, Leibiger IB, Efendiev R, Katz AI, Leibiger B, Berggren PO, et al. Tyrosine 537 within the Na+,K+-ATPase alpha-subunit is essential for AP-2 binding and clathrin-dependent endocytosis. The Journal of Biological Chemistry. 2002;277:17108-17111
  109. 109. Efendiev R, Bertorello AM, Pressley TA, Rousselot M, Feraille E, Pedemonte CH. Simultaneous phosphorylation of Ser11 and Ser18 in the alpha-subunit promotes the recruitment of Na(+),K(+)-ATPase molecules to the plasma membrane. Biochemistry. 2000;39:9884-9892
  110. 110. Feschenko MS, Sweadner KJ. Structural basis for species-specific differences in the phosphorylation of Na,K-ATPase by protein kinase C. The Journal of Biological Chemistry. 1995;270:14072-14077
  111. 111. Efendiev R, Bertorello AM, Pedemonte CH. PKC-beta and PKC-zeta mediate opposing effects on proximal tubule Na+,K+-ATPase activity. FEBS Letters. 1999;456:45-48
  112. 112. Pedemont CH, Bertorello AM. Short-term regulation of the proximal tubule Na+,K+-ATPase: Increased/decreased Na+,K+-ATPase activity mediated by protein kinase C isoforms. Journal of Bioenergetics and Biomembranes. 2001;33:439-447
  113. 113. Ridge KM, Dada L, Lecuona E, Bertorello AM, Katz AI, Mochly-Rosen D, et al. Dopamine-induced exocytosis of Na,K-ATPase is dependent on activation of protein kinase C-epsilon and -delta. Molecular Biology of the Cell. 2002;13:1381-1389
  114. 114. Khundmiri SJ, Bertorello AM, Delamere NA, Lederer ED. Clathrin-mediated endocytosis of Na+,K+-ATPase in response to parathyroid hormone requires ERK-dependent phosphorylation of Ser-11 within the alpha1-subunit. The Journal of Biological Chemistry. 2004;279:17418-17427
  115. 115. Cai H, Wu L, Qu W, Malhotra D, Xie Z, Shapiro JI, et al. Regulation of apical NHE3 trafficking by ouabain-induced activation of the basolateral Na+-K+-ATPase receptor complex. American Journal of Physiology. Cell Physiology. 2008;294:C555-C563
  116. 116. Liu J, Kesiry R, Periyasamy SM, Malhotra D, Xie Z, Shapiro JI. Ouabain induces endocytosis of plasmalemmal Na/K-ATPase in LLC-PK1 cells by a clathrin-dependent mechanism. Kidney International. 2004;66:227-241
  117. 117. Liu J, Liang M, Liu L, Malhotra D, Xie Z, Shapiro JI. Ouabain-induced endocytosis of the plasmalemmal Na/K-ATPase in LLC-PK1 cells requires caveolin-1. Kidney International. 2005;67:1844-1854
  118. 118. Larkin JM, Brown MS, Goldstein JL, Anderson RG. Depletion of intracellular potassium arrests coated pit formation and receptor-mediated endocytosis in fibroblasts. Cell. 1983;33:273-285
  119. 119. Wang LH, Rothberg KG, Anderson RG. Mis-assembly of clathrin lattices on endosomes reveals a regulatory switch for coated pit formation. The Journal of Cell Biology. 1993;123:1107-1117
  120. 120. Wang H, Haas M, Liang M, Cai T, Tian J, Li S, et al. Ouabain assembles signaling cascades through the Caveolar Na+/K+-ATPase. Journal of Biological Chemistry. 2004;279:17250-17259
  121. 121. LaPointe MS, Sodhi C, Sahai A, Batlle D. Na+/H+ exchange activity and NHE-3 expression in renal tubules from the spontaneously hypertensive rat. Kidney International. 2002;62:157-165
  122. 122. Li XX, Xu J, Zheng S, Albrecht FE, Robillard JE, Eisner GM, et al. D(1) dopamine receptor regulation of NHE3 during development in spontaneously hypertensive rats. American Journal of Physiology. Regulatory, Integrative and Comparative Physiology. 2001;280:R1650-R1656
  123. 123. Kelly MP, Quinn PA, Davies JE, Ng LL. Activity and expression of Na+-H+ exchanger isoforms 1 and 3 in kidney proximal tubules of hypertensive rats. Circulation Research. 1997;80:853-860
  124. 124. Hayashi M, Yoshida T, Monkawa T, Yamaji Y, Sato S, Saruta T. Na+/H+-exchanger 3 activity and its gene in the spontaneously hypertensive rat kidney. Journal of Hypertension. 1997;15:43-48
  125. 125. Zhang Y, Mircheff AK, Hensley CB, Magyar CE, Warnock DG, Chambrey R, et al. Rapid redistribution and inhibition of renal sodium transporters during acute pressure natriuresis. The American Journal of Physiology. 1996;270:F1004-F1014
  126. 126. McDonough AA, Leong PK, Yang LE. Mechanisms of pressure natriuresis: How blood pressure regulates renal sodium transport. Annals of the New York Academy of Sciences. 2003;986:669-677
  127. 127. Yang L, Leong PK, Chen JO, Patel N, Hamm-Alvarez SF, McDonough AA. Acute hypertension provokes internalization of proximal tubule NHE3 without inhibition of transport activity. American Journal of Physiology. Renal Physiology. 2002;282:F730-F740
  128. 128. Yang LE, Maunsbach AB, Leong PK, McDonough AA. Differential traffic of proximal tubule Na+ transporters during hypertension or PTH: NHE3 to base of microvilli vs. NaPi2 to endosomes. American Journal of Physiology. Renal Physiology. 2004;287:F896-F906
  129. 129. Lorenz JN, Schultheis PJ, Traynor T, Shull GE, Schnermann J. Micropuncture analysis of single-nephron function in NHE3-deficient mice. The American Journal of Physiology. 1999;277:F447-F453
  130. 130. Ledoussal C, Lorenz JN, Nieman ML, Soleimani M, Schultheis PJ, Shull GE. Renal salt wasting in mice lacking NHE3 Na+/H+ exchanger but not in mice lacking NHE2. American Journal of Physiology. Renal Physiology. 2001;281:F718-F727
  131. 131. Schultheis PJ, Clarke LL, Meneton P, Miller ML, Soleimani M, Gawenis LR, et al. Renal and intestinal absorptive defects in mice lacking the NHE3 Na+/H+ exchanger. Nature Genetics. 1998;19:282-285
  132. 132. Wang T, Yang C-L, Abbiati T, Schultheis PJ, Shull GE, Giebisch G, et al. Mechanism of proximal tubule bicarbonate absorption in NHE3 null mice. American Journal of Physiology. Renal Physiology. 1999;277:F298-F302
  133. 133. Orlowski J, Grinstein S. Diversity of the mammalian sodium/proton exchanger SLC9 gene family. Pflügers Archiv. 2004;447:549-565
  134. 134. Chow C-W, Khurana S, Woodside M, Grinstein S, Orlowski J. The epithelial Na+/H+ exchanger, NHE3, is internalized through a clathrin-mediated pathway. The Journal of Biological Chemistry. 1999;274:37551-37558
  135. 135. Li X, Galli T, Leu S, Wade JB, Weinman EJ, Leung G, et al. Na+-H+ exchanger 3 (NHE3) is present in lipid rafts in the rabbit ileal brush border: A role for rafts in trafficking and rapid stimulation of NHE3. The Journal of Physiology. 2001;537:537-552
  136. 136. Donowitz M, Janecki A, Akhter S, Cavet ME, Sanchez F, Lamprecht G, et al. Short-term regulation of NHE3 by EGF and protein kinase C but not protein kinase A involves vesicle trafficking in epithelial cells and fibroblasts. Annals of the New York Academy of Sciences. 2000;915:30-42
  137. 137. Fan L, Wiederkehr MR, Collazo R, Wang H, Crowder LA, Moe OW. Dual mechanisms of regulation of Na/H exchanger NHE-3 by parathyroid hormone in rat kidney. The Journal of Biological Chemistry. 1999;274:11289-11295
  138. 138. Kurashima K, Szabo EZ, Lukacs G, Orlowski J, Grinstein S. Endosomal recycling of the Na+/H+ exchanger NHE3 isoform is regulated by the phosphatidylinositol 3-kinase pathway. The Journal of Biological Chemistry. 1998;273:20828-20836
  139. 139. Li X, Zhang H, Cheong A, Leu S, Chen Y, Elowsky CG, et al. Carbachol regulation of rabbit ileal brush border Na+-H+ exchanger 3 (NHE3) occurs through changes in NHE3 trafficking and complex formation and is Src dependent. The Journal of Physiology. 2004;556:791-804
  140. 140. Tsuganezawa H, Preisig PA, Alpern RJ. Dominant negative c-Src inhibits angiotensin II induced activation of NHE3 in OKP cells. Kidney International. 1998;54:394-398
  141. 141. Janecki AJ, Janecki M, Akhter S, Donowitz M. Basic fibroblast growth factor stimulates surface expression and activity of Na(+)/H(+) exchanger NHE3 via mechanism involving phosphatidylinositol 3-kinase. The Journal of Biological Chemistry. 2000;275:8133-8142
  142. 142. Yang X, Amemiya M, Peng Y, Moe OW, Preisig PA, Alpern RJ. Acid incubation causes exocytic insertion of NHE3 in OKP cells. American Journal of Physiology. Cell Physiology. 2000;279:C410-C419
  143. 143. Lee-Kwon W, Johns DC, Cha B, Cavet M, Park J, Tsichlis P, et al. Constitutively active phosphatidylinositol 3-kinase and AKT are sufficient to stimulate the epithelial Na+/H+ exchanger 3. The Journal of Biological Chemistry. 2001;276:31296-31304
  144. 144. Collazo R, Fan L, Hu MC, Zhao H, Wiederkehr MR, Moe OW. Acute regulation of Na+/H+ exchanger NHE3 by parathyroid hormone via NHE3 phosphorylation and dynamin-dependent endocytosis. The Journal of Biological Chemistry. 2000;275:31601-31608
  145. 145. Zhang Y, Magyar CE, Norian JM, Holstein-Rathlou NH, Mircheff AK, McDonough AA. Reversible effects of acute hypertension on proximal tubule sodium transporters. The American Journal of Physiology. 1998;274:C1090-C1100
  146. 146. Oweis S, Wu L, Kiela PR, Zhao H, Malhotra D, Ghishan FK, et al. Cardiac glycoside downregulates NHE3 activity and expression in LLC-PK1 cells. American Journal of Physiology. Renal Physiology. 2006;290:F997-F1008
  147. 147. Di Sole F, Cerull R, Petzke S, Casavola V, Burckhardt G, Helmle-Kolb C. Bimodal acute effects of A1 adenosine receptor activation on Na+/H+ exchanger 3 in opossum kidney cells. Journal of the American Society of Nephrology. 2003;14:1720-1730
  148. 148. Lee-Kwon W, Kim JH, Choi JW, Kawano K, Cha B, Dartt DA, et al. Ca2+−dependent inhibition of NHE3 requires PKC alpha which binds to E3KARP to decrease surface NHE3 containing plasma membrane complexes. American Journal of Physiology. Cell Physiology. 2003;285:C1527-C1536
  149. 149. Yan Y, Haller S, Shapiro A, Malhotra N, Tian J, Xie Z, et al. Ouabain-stimulated trafficking regulation of the Na/K-ATPase and NHE3 in renal proximal tubule cells. Molecular and Cellular Biochemistry. 2012;367:175-183
  150. 150. Harootunian AT, Kao JP, Eckert BK, Tsien RY. Fluorescence ratio imaging of cytosolic free Na+ in individual fibroblasts and lymphocytes. The Journal of Biological Chemistry. 1989;264:19458-19467
  151. 151. Yang LE, Sandberg MB, Can AD, Pihakaski-Maunsbach K, McDonough AA. Effects of dietary salt on renal Na+ transporter subcellular distribution, abundance, and phosphorylation status. American Journal of Physiology. Renal Physiology. 2008;295:F1003-F1016
  152. 152. Pinho MJ, Serrao MP, Jose PA, Soares-da-Silva P. Organ specific underexpression renal of Na+−dependent B0AT1 in the SHR correlates positively with overexpression of NHE3 and salt intake. Molecular and Cellular Biochemistry. 2007;306:9-18
  153. 153. Pedrosa R, Gomes P, Zeng C, Hopfer U, Jose PA, Soares-da-Silva P. Dopamine D3 receptor-mediated inhibition of Na+/H+ exchanger activity in normotensive and spontaneously hypertensive rat proximal tubular epithelial cells. British Journal of Pharmacology. 2004;142:1343-1353
  154. 154. Garg LC, Narang N. Sodium-potassium-adenosine triphosphatase in nephron segments of spontaneously hypertensive rats. The Journal of Laboratory and Clinical Medicine. 1985;106:43-46
  155. 155. Hinojos CA, Doris PA. Altered subcellular distribution of Na+,K+-ATPase in proximal tubules in young spontaneously hypertensive rats. Hypertension. 2004;44:95-100
  156. 156. Periyasamy SM, Liu J, Tanta F, Kabak B, Wakefield B, Malhotra D, et al. Salt loading induces redistribution of the plasmalemmal Na/K-ATPase in proximal tubule cells. Kidney International. 2005;67:1868-1877
  157. 157. Nishi A, Bertorello AM, Aperia A. High salt diet down-regulates proximal tubule Na+, K(+)-ATPase activity in Dahl salt-resistant but not in Dahl salt-sensitive rats: Evidence of defective dopamine regulation. Acta Physiologica Scandinavica. 1992;144:263-267
  158. 158. Nishi A, Eklof AC, Bertorello AM, Aperia A. Dopamine regulation of renal Na+,K(+)-ATPase activity is lacking in Dahl salt-sensitive rats. Hypertension. 1993;21:767-771
  159. 159. Felder RA, Jose PA. Mechanisms of disease: The role of GRK4 in the etiology of essential hypertension and salt sensitivity. Nature Clinical Practice. Nephrology. 2006;2:637-650
  160. 160. Hussain T, Lokhandwala MF. Renal dopamine DA1 receptor coupling with G(S) and G(q/11) proteins in spontaneously hypertensive rats. The American Journal of Physiology. 1997;272:F339-F346
  161. 161. Felder RA, Sanada H, Xu J, Yu PY, Wang Z, Watanabe H, et al. G protein-coupled receptor kinase 4 gene variants in human essential hypertension. Proceedings of the National Academy of Sciences of the United States of America. 2002;99:3872-3877
  162. 162. Fedorova OV, Lakatta EG, Bagrov AY. Endogenous Na,K pump ligands are differentially regulated during acute NaCl loading of Dahl rats. Circulation. 2000;102:3009-3014
  163. 163. Foulkes R, Ferrario RG, Salvati P, Bianchi G. Differences in ouabain-induced natriuresis between isolated kidneys of Milan hypertensive and normotensive rats. Clinical Science (London, England). 1992;82:185-190
  164. 164. Yates NA, McDougall JG. Effect of volume expansion on the natriuretic response to ouabain infusion. Renal Physiology and Biochemistry. 1995;18:311-320
  165. 165. Anderson DE, Fedorova OV, Morrell CH, Longo DL, Kashkin VA, Metzler JD, et al. Endogenous sodium pump inhibitors and age-associated increases in salt sensitivity of blood pressure in normotensives. American Journal of Physiology - Regulatory, Integrative and Comparative Physiology. 2008;294:R1248-R1254
  166. 166. McPherson PS, Kay BK, Hussain NK. Signaling on the endocytic pathway. Traffic. 2001;2:375-384
  167. 167. Cavalli V, Corti M, Gruenberg J. Endocytosis and signaling cascades: A close encounter. FEBS Letters. 2001;498:190-196
  168. 168. Roy S, Wyse B, Hancock JF. H-Ras signaling and K-Ras signaling are differentially dependent on endocytosis. Molecular and Cellular Biology. 2002;22:5128-5140
  169. 169. Wilde A, Beattie EC, Lem L, Riethof DA, Liu SH, Mobley WC, et al. EGF receptor signaling stimulates SRC kinase phosphorylation of clathrin, influencing clathrin redistribution and EGF uptake. Cell. 1999;96:677-687
  170. 170. Wiley HS, Burke PM. Regulation of receptor tyrosine kinase signaling by endocytic trafficking. Traffic. 2001;2:12-18
  171. 171. Kuwada SK, Lund KA, Li XF, Cliften P, Amsler K, Opresko LK, et al. Differential signaling and regulation of apical vs. basolateral EGFR in polarized epithelial cells. The American Journal of Physiology. 1998;275:C1419-C1428
  172. 172. Liu J, Lilly MN, Shapiro JI. Targeting Na/K-ATPase signaling: A new approach to control oxidative stress. Current Pharmaceutical Design. 2018;24:359-364
  173. 173. Hinshaw JE. Dynamin and its role in membrane fission. Annual Review of Cell and Developmental Biology. 2000;16:483-519
  174. 174. Sever S. Dynamin and endocytosis. Current Opinion in Cell Biology. 2002;14:463-467
  175. 175. Sorkin A. The endocytosis machinery. Journal of Cell Science. 2000;113(Pt 24):4375-4376
  176. 176. Pol A, Calvo M, Enrich C. Isolated endosomes from quiescent rat liver contain the signal transduction machinery. Differential distribution of activated Raf-1 and Mek in the endocytic compartment. FEBS Letters. 1998;441:34-38
  177. 177. De Vries L, Elenko E, McCaffery JM, Fischer T, Hubler L, McQuistan T, et al. RGS-GAIP, a GTPase-activating protein for Galpha i Heterotrimeric G proteins, is located on clathrin-coated vesicles. Molecular Biology of the Cell. 1998;9:1123-1134
  178. 178. De Vries L, Lou X, Zhao G, Zheng B, Farquhar MG. GIPC, a PDZ domain containing protein, interacts specifically with the C terminus of RGS-GAIP. PNAS. 1998;95:12340-12345
  179. 179. Stenberg PE, Pestina TI, Barrie RJ, Jackson CW. The Src family kinases, Fgr, Fyn, Lck, and Lyn, colocalize with coated membranes in platelets. Blood. 1997;89:2384-2393
  180. 180. Anderson RG. Caveolae: Where incoming and outgoing messengers meet. Proceedings of the National Academy of Sciences of the United States of America. 1993;90:10909-10913
  181. 181. Bickel PE. Lipid rafts and insulin signaling. American Journal of Physiology. Endocrinology and Metabolism. 2002;282:E1-E10
  182. 182. Liu P, Rudick M, Anderson RG. Multiple functions of caveolin-1. The Journal of Biological Chemistry. 2002;277:41295-41298
  183. 183. Pelkmans L, Helenius A. Endocytosis via caveolae. Traffic. 2002;3:311-320
  184. 184. Schlegel A, Lisanti MP. The caveolin triad: Caveolae biogenesis, cholesterol trafficking, and signal transduction. Cytokine & Growth Factor Reviews. 2001;12:41-51
  185. 185. Galbiati F, Razani B, Lisanti MP. Emerging themes in lipid rafts and caveolae. Cell. 2001;106:403-411
  186. 186. Nabi IR, Le PU. Caveolae/raft-dependent endocytosis. The Journal of Cell Biology. 2003;161:673-677
  187. 187. Shigematsu S, Watson RT, Khan AH, Pessin JE. The adipocyte plasma membrane caveolin functional/structural organization is necessary for the efficient endocytosis of GLUT4. The Journal of Biological Chemistry. 2003;278:10683-10690
  188. 188. Sleight S, Wilson BA, Heimark DB, Larner J. G(q/11) is involved in insulin-stimulated inositol phosphoglycan putative mediator generation in rat liver membranes: Co-localization of G(q/11) with the insulin receptor in membrane vesicles. Biochemical and Biophysical Research Communications. 2002;295:561-569
  189. 189. Scherer PE, Lisanti MP, Baldini G, Sargiacomo M, Mastick CC, Lodish HF. Induction of caveolin during adipogenesis and association of GLUT4 with caveolin-rich vesicles. The Journal of Cell Biology. 1994;127:1233-1243
  190. 190. Stoddart A, Dykstra ML, Brown BK, Song W, Pierce SK, Brodsky FM. Lipid rafts unite signaling cascades with clathrin to regulate BCR internalization. Immunity. 2002;17:451-462
  191. 191. Liu L, Mohammadi K, Aynafshar B, Wang H, Li D, Liu J, et al. Role of caveolae in signal-transducing function of cardiac Na+/K+-ATPase. American Journal of Physiology. Cell Physiology. 2003;284:C1550-C1560
  192. 192. Felberbaum-Corti M, Gruenberg J. Signaling from the far side. Molecular Cell. 2002;10:1259-1260
  193. 193. Liu J, Shapiro JI. Endocytosis and signal transduction: Basic science update. Biological Research for Nursing. 2003;5:117-128
  194. 194. Sorkin A, Von Zastrow M. Signal transduction and endocytosis: Close encounters of many kinds. Nature Reviews. Molecular Cell Biology. 2002;3:600-614
  195. 195. Di Guglielmo GM, Baass PC, Ou WJ, Posner BI, Bergeron JJ. Compartmentalization of SHC, GRB2 and mSOS, and hyperphosphorylation of Raf-1 by EGF but not insulin in liver parenchyma. The EMBO Journal. 1994;13:4269-4277
  196. 196. Kranenburg O, Verlaan I, Moolenaar WH. Dynamin is required for the activation of mitogen-activated protein (MAP) kinase by MAP kinase kinase. The Journal of Biological Chemistry. 1999;274:35301-35304
  197. 197. Ware MF, Tice DA, Parsons SJ, Lauffenburger DA. Overexpression of cellular Src in fibroblasts enhances endocytic internalization of epidermal growth factor receptor. The Journal of Biological Chemistry. 1997;272:30185-30190
  198. 198. Vaziri ND, Rodriguez-Iturbe B. Mechanisms of disease: Oxidative stress and inflammation in the pathogenesis of hypertension. Nature Clinical Practice. Nephrology. 2006;2:582-593
  199. 199. Wilcox CS. Oxidative stress and nitric oxide deficiency in the kidney: A critical link to hypertension? American Journal of Physiology. Regulatory, Integrative and Comparative Physiology. 2005;289:R913-R935
  200. 200. Touyz RM. Reactive oxygen species, vascular oxidative stress, and redox signaling in hypertension: What is the clinical significance? Hypertension. 2004;44:248-252
  201. 201. Welch WJ. Intrarenal oxygen and hypertension. Clinical and Experimental Pharmacology & Physiology. 2006;33:1002-1005
  202. 202. Kitiyakara C, Chabrashvili T, Chen Y, Blau J, Karber A, Aslam S, et al. Salt intake, oxidative stress, and renal expression of NADPH oxidase and superoxide dismutase. Journal of the American Society of Nephrology. 2003;14:2775-2782
  203. 203. Meng S, Roberts LJ, Cason GW, Curry TS, Manning RD. Superoxide dismutase and oxidative stress in Dahl salt-sensitive and -resistant rats. American Journal of Physiology - Regulatory, Integrative and Comparative Physiology. 2002;283:R732-R738
  204. 204. Taylor NE, Glocka P, Liang M, Cowley AW. NADPH oxidase in the renal medulla causes oxidative stress and contributes to salt-sensitive hypertension in Dahl S rats. Hypertension. 2006;47:692-698
  205. 205. Garvin JL, Ortiz PA. The role of reactive oxygen species in the regulation of tubular function. Acta Physiologica Scandinavica. 2003;179:225-232
  206. 206. Schreck C, O’Connor PM. NAD(P)H oxidase and renal epithelial ion transport. American Journal of Physiology - Regulatory, Integrative and Comparative Physiology. 2011;300:R1023-R1029
  207. 207. Wang X, Armando I, Upadhyay K, Pascua A, Jose PA. The regulation of proximal tubular salt transport in hypertension: An update. Current Opinion in Nephrology and Hypertension. 2009;18:412-420
  208. 208. Panico C, Luo Z, Damiano S, Artigiano F, Gill P, Welch WJ. Renal proximal tubular reabsorption is reduced in adult spontaneously hypertensive rats: Roles of superoxide and Na+/H+ exchanger 3. Hypertension. 2009;54:1291-1297
  209. 209. Banday AA, Lokhandwala MF. Angiotensin II-mediated biphasic regulation of proximal tubular Na+/H+ exchanger 3 is impaired during oxidative stress. American Journal of Physiology. Renal Physiology. 2011;301:F364-F370
  210. 210. Han HJ, Lee YJ, Park SH, Lee JH, Taub M. High glucose-induced oxidative stress inhibits Na+/glucose cotransporter activity in renal proximal tubule cells. American Journal of Physiology - Renal Physiology. 2005;288:F988-F996
  211. 211. Moe OW, Tejedor A, Levi M, Seldin DW, Preisig PA, Alpern RJ. Dietary NaCl modulates Na(+)-H+ antiporter activity in renal cortical apical membrane vesicles. American Journal of Physiology - Renal Physiology. 1991;260:F130-F137
  212. 212. Fisher KA, Lee SH, Walker J, Dileto-Fang C, Ginsberg L, Stapleton SR. Regulation of proximal tubule sodium/hydrogen antiporter with chronic volume contraction. American Journal of Physiology - Renal Physiology. 2001;280:F922-F926
  213. 213. Silva E, Soares-da-Silva P. Reactive oxygen species and the regulation of renal Na+-K+-ATPase in opossum kidney cells. American Journal of Physiology - Regulatory, Integrative and Comparative Physiology. 2007;293:R1764-R1770
  214. 214. Yang LE, Leong PKK, McDonough AA. Reducing blood pressure in SHR with enalapril provokes redistribution of NHE3, NaPi2, and NCC and decreases NaPi2 and ACE abundance. American Journal of Physiology - Renal Physiology. 2007;293:F1197-F1208
  215. 215. Crajoinas RO, Lessa LMA, Carraro-Lacroix LR, Davel APC, Pacheco BPM, Rossoni LV, et al. Posttranslational mechanisms associated with reduced NHE3 activity in adult vs. young prehypertensive SHR. American Journal of Physiology - Renal Physiology. 2010;299:F872-F881
  216. 216. Johns EJ, O’Shaughnessy B, O’Neill S, Lane B, Healy V. Impact of elevated dietary sodium intake on NAD(P)H oxidase and SOD in the cortex and medulla of the rat kidney. American Journal of Physiology - Regulatory, Integrative and Comparative Physiology. 2010;299:R234-R240
  217. 217. Petrushanko IY, Yakushev S, Mitkevich VA, Kamanina YV, Ziganshin RH, Meng X, et al. S-glutathionylation of the Na,K-ATPase catalytic alpha subunit is a determinant of the enzyme redox sensitivity. The Journal of Biological Chemistry. 2012;287:32195-32205
  218. 218. Liu C-C, Garcia A, Mahmmoud YA, Hamilton EJ, Galougahi KK, Fry NAS, et al. Susceptibility of β1 Na+-K+ pump subunit to glutathionylation and oxidative inhibition depends on conformational state of pump. Journal of Biological Chemistry. 2012;287:12353-12364
  219. 219. Figtree GA, Liu C-C, Bibert S, Hamilton EJ, Garcia A, White CN, et al. Reversible oxidative modification: A key mechanism of Na+-K+ pump regulation. Circulation Research. 2009;105:185-193
  220. 220. Huang WH, Wang Y, Askari A. (Na+ + K+)-ATPase: Inactivation and degradation induced by oxygen radicals. The International Journal of Biochemistry. 1992;24:621-626
  221. 221. Thevenod F, Friedmann JM. Cadmium-mediated oxidative stress in kidney proximal tubule cells induces degradation of Na+/K(+)-ATPase through proteasomal and endo−/lysosomal proteolytic pathways. The FASEB Journal. 1999;13:1751-1761
  222. 222. Figtree GA, Keyvan Karimi G, Liu CC, Rasmussen HH. Oxidative regulation of the Na(+)-K(+) pump in the cardiovascular system. Free Radical Biology & Medicine. 2012;53:2263-2268
  223. 223. Bogdanova A, Boldyrev A, Gassmann M. Oxygen- and redox-induced regulation of the Na/K ATPase. Current Enzyme Inhibition. 2006;2:37-59
  224. 224. Soares-da-Silva ES. Renal Redox Balance and Na+,K+-ATPase Regulation: Role in Physiology and Pathophysiology. Rijeka: InTech; 2012
  225. 225. Ellis DZ, Rabe J, Sweadner KJ. Global loss of Na,K-ATPase and its nitric oxide-mediated regulation in a transgenic mouse model of amyotrophic lateral sclerosis. The Journal of Neuroscience. 2003;23:43-51
  226. 226. Xie ZJ, Wang YH, Askari A, Huang WH, Klaunig JE. Studies on the specificity of the effects of oxygen metabolites on cardiac sodium pump. Journal of Molecular and Cellular Cardiology. Rijeka, Croatia: InTech. 2012;22:911-920
  227. 227. Mense M, Stark G, Apell HJ. Effects of free radicals on partial reactions of the Na,K-ATPase. The Journal of Membrane Biology. 1997;156:63-71
  228. 228. White CN, Figtree GA, Liu C-C, Garcia A, Hamilton EJ, Chia KKM, et al. Angiotensin II inhibits the Na+-K+ pump via PKC-dependent activation of NADPH oxidase. American Journal of Physiology - Cell Physiology. 2009;296:C693-C700
  229. 229. Reifenberger MS, Arnett KL, Gatto C, Milanick MA. The reactive nitrogen species peroxynitrite is a potent inhibitor of renal Na-K-ATPase activity. American Journal of Physiology - Renal Physiology. 2008;295:F1191-F1198
  230. 230. Dobrota D, Matejovicova M, Kurella EG, Boldyrev AA. Na/K-ATPase under oxidative stress: Molecular mechanisms of injury. Cellular and Molecular Neurobiology. 1999;19:141-149
  231. 231. Xie Z, Kometiani P, Liu J, Li J, Shapiro JI, Askari A. Intracellular reactive oxygen species mediate the linkage of Na+/K+-ATPase to hypertrophy and its marker genes in cardiac myocytes. The Journal of Biological Chemistry. 1999;274:19323-19328
  232. 232. Tian J, Liu J, Garlid KD, Shapiro JI, Xie Z. Involvement of mitogen-activated protein kinases and reactive oxygen species in the inotropic action of ouabain on cardiac myocytes. A potential role for mitochondrial K(ATP) channels. Molecular and Cellular Biochemistry. 2003;242:181-187
  233. 233. Elkareh J, Kennedy DJ, Yashaswi B, Vetteth S, Shidyak A, Kim EG, et al. Marinobufagenin stimulates fibroblast collagen production and causes fibrosis in experimental uremic cardiomyopathy. Hypertension. 2007;49:215-224
  234. 234. Yatime L, Laursen M, Morth JP, Esmann M, Nissen P, Fedosova NU. Structural insights into the high affinity binding of cardiotonic steroids to the Na+,K+-ATPase. Journal of Structural Biology. 2011;174:296-306
  235. 235. Stadtman ER, Berlett BS. Fenton chemistry. Amino acid oxidation. Journal of Biological Chemistry. 1991;266:17201-17211
  236. 236. Stadtman ER, Levine RL. Protein oxidation. Annals of the New York Academy of Sciences. 2000;899:191-208
  237. 237. Nystrom T. Role of oxidative carbonylation in protein quality control and senescence. The EMBO Journal. 2005;24:1311-1317
  238. 238. Dalle-Donne I, Giustarini D, Colombo R, Rossi R, Milzani A. Protein carbonylation in human diseases. Trends in Molecular Medicine. 2003;9:169-176
  239. 239. Dalle-Donne I, Rossi R, Colombo R, Giustarini D, Milzani A. Biomarkers of oxidative damage in human disease. Clinical Chemistry. 2006;52:601-623
  240. 240. Dalle-Donne I, Aldini G, Carini M, Colombo R, Rossi R, Milzani A. Protein carbonylation, cellular dysfunction, and disease progression. Journal of Cellular and Molecular Medicine. 2006;10:389-406
  241. 241. Wong CM, Marcocci L, Liu L, Suzuki YJ. Cell signaling by protein carbonylation and decarbonylation. Antioxidants & Redox Signaling. 2010;12:393-404
  242. 242. Wong CM, Cheema AK, Zhang L, Suzuki YJ. Protein carbonylation as a novel mechanism in redox signaling. Circulation Research. 2008;102:310-318
  243. 243. Wong CM, Bansal G, Marcocci L, Suzuki YJ. Proposed role of primary protein carbonylation in cell signaling. Redox Report. 2012;17:90-94
  244. 244. Wong CM, Marcocci L, Das D, Wang X, Luo H, Zungu-Edmondson M, et al. Mechanism of protein decarbonylation. Free Radical Biology & Medicine. 2013;65:1126-1133
  245. 245. Munzel T, Gori T, Bruno RM, Taddei S. Is oxidative stress a therapeutic target in cardiovascular disease? European Heart Journal. 2010;31:2741-2748
  246. 246. Huang H-Y, Caballero B, Chang S, Alberg AJ, Semba RD, Schneyer CR, et al. The efficacy and safety of multivitamin and mineral supplement use to prevent cancer and chronic disease in adults: A systematic review for a National Institutes of Health State-of-the-Science Conference. Annals of Internal Medicine. 2006;145:372-385
  247. 247. Dahl LK, Heine M, Tassinari L. Role of genetic factors in susceptibility to experimental hypertension due to chronic excess salt ingestion. Nature. 1962;194:480-482
  248. 248. Dahl LK, Heine M, Thompson K. Genetic influence of the kidneys on blood pressure. Evidence from chronic renal homografts in rats with opposite predispositions to hypertension. Circulation Research. 1974;40:94-101
  249. 249. Rapp JP. Dahl salt-susceptible and salt-resistant rats. A review. Hypertension. 1982;4:753-763
  250. 250. Rapp JP, Dene H. Development and characteristics of inbred strains of Dahl salt-sensitive and salt-resistant rats. Hypertension. 1985;7:340-349
  251. 251. Mokry M, Cuppen E. The Atp1a1 gene from inbred Dahl salt sensitive rats does not contain the A1079T missense transversion. Hypertension. 2008;51:922-927
  252. 252. Joe B, G. M. Genetic Analysis of Inherited Hypertension in the Rat. Amsterdam: Elsevier Science; 2006
  253. 253. Nishi A, Bertorello AM, Aperia A. Renal Na+,K(+)-ATPase in Dahl salt-sensitive rats: K+ dependence, effect of cell environment and protein kinases. Acta Physiologica Scandinavica. 1993;149:377-384
  254. 254. Guyton AC. Blood pressure control--special role of the kidneys and body fluids. Science. 1991;252:1813-1816
  255. 255. Calhoun DA, Jones D, Textor S, Goff DC, Murphy TP, Toto RD, et al. Resistant hypertension: Diagnosis, evaluation, and treatment: A scientific statement from the American Heart Association Professional Education Committee of the Council for high blood pressure research. Hypertension. 2008;51:1403-1419
  256. 256. Meneton P, Jeunemaitre X, de Wardener HE, MacGregor GA. Links between dietary salt intake, renal salt handling, blood pressure, and cardiovascular diseases. Physiological Reviews. 2005;85:679-715
  257. 257. Haddy FJ. Role of dietary salt in hypertension. Life Sciences. 2006;79:1585-1592
  258. 258. He FJ, MacGregor GA. Effect of longer-term modest salt reduction on blood pressure. Cochrane Database of Systematic Reviews. 2004:CD004937
  259. 259. Yatime L, Buch-Pedersen MJ, Musgaard M, Morth JP, Lund Winther AM, Pedersen BP, et al. P-type ATPases as drug targets: Tools for medicine and science. Biochimica et Biophysica Acta. 2009;1787:207-220
  260. 260. Aperia A. New roles for an old enzyme: Na,K-ATPase emerges as an interesting drug target. Journal of Internal Medicine. 2007;261:44-52

Written By

Jiang Liu, Yanling Yan and Joseph I. Shapiro

Submitted: 13 January 2020 Reviewed: 21 May 2020 Published: 30 September 2020