I CUN. CHEM. 40/9, 1674-1685 (1994) Understanding the Sodium Pump and Its Relevance to Disease Andrea M. Rose’ and Roland Valdes, Jr.” Na,K-ATPase (sodium pump; EC 3.6.1.37) is present in the membrane of most eukaryotic cells and controls directly or indirectly many essential cellular functions. Regulation of this enzyme (ion transporter) and its individual isoforms is believed to play a key role in the etiology of some pathological processes. The sodium pump is the only known receptor for the cardiac glycosides. However, endogenous ligands structurally similar to digoxin or ouabain may control the activity of this important molecular complex. Here we review the structure and function of Na,K-ATPase, its expression and distribution in tissues, and its interaction with known ligands such as the cardiac glycosides and other suspected endogenous regulators. Also reviewed are various disorders, including cardiovascular, neurological, renal, and metabolic diseases, purported to involve dysfunction of Na,K-ATPase activity. The escalation in knowledge at the molecular level concerning sodium pump function foreshadows application of this knowledge in the clinical laboratory to identify individuals at risk for Na,K-ATPase-associated diseases. IndexIng Terms: Na,K-ATPase/isoforms/cardiac glycosides/ age- related effects/digoxin/ouabain/hypertenslon/diabetes/Alzheimer disease/neurological disorders The sodium-potassium-activated adenosine triphosphatase (Na,K-ATPase; sodium pump; EC 3.6.1.37) is a plasma membrane-associated protein complex that is expressed in most eukaryotic cells.4’5 The “pump” couples the energy released in the intracellular hydrolysis of adenosine tnphosphate (ATP) to the transport of cellular ions, a major pathway for the controlled translocation of sodium and potassium ions across the cell membrane. Na,K-ATPase therefore controls directly or indirectly many essential cellular functions, e.g., cell volume, free calcium concentration, and membrane potential. RegulaDepartments of ‘ Pathology and 2Biochemistry, University of Louisville School of Medicine, Louisville, KY 40292. ‘Mdress correspondence to this author at: Department of Pathology, University of Louisville, Louisville, KY 40292. Fax 502852-1771; E-mail [email protected]. 4Nonstandard abbreviations: Na,K-ATPase, Na,K-activated adenosine triphosphatase; DLIF, digoxin-like immunoreactive factors; EDLF, endogenous digoxin-like factors; OLF, ouabain-like factors; and AD, Alzheimer disease. 5Although technically an enzyme, Na,K-ATPase functions primarily as an ion transporter. Therefore, most investigators refer to the alpha and beta gene products as isoforms instead of isoenzymes. Received December 27, 1993; accepted June 16, 1994. 1674 CLINICAL CHEMISTRY, Vol. 40, No. 9, 1994 tion of this enzyme (transporter) and its individual isoforms is thought to play a key role in the etiology of some pathological processes. The sodium pump is the only known receptor for the cardiac glycosides used to treat congestive heart failure and cardiac arrhythmias. This suggests that endogenous ligands structurally similar to cardiac glycosides may act as natural regulators of the sodium pump in heart and other tissues. Identification of naturally cccurring regulators of Na,K-ATPase could initiate the discovery of new hormone-like control systems involved in the etiology of selected disease processes, hence the importance of understanding the relation of the sodium pump and its ligands to disease. In this article, we review recent information related to structure and function, genetic expression and distribution in tissues, and interaction of the sodium pump with ligands such as the cardiac glycosides and other suspected endogenous counterparts. Several diverse disease processes-including cardiovascular, renal, neurological, and metabolic disorders, having in common a dysfunction in salt and water homeostasis-are emphasized, as is the clinical need for understanding the function and control of this ubiquitous ion transporter system. Structure and FunctIon of Na,K-ATPase Na,K-ATPase couples the energy released in the intracellular hydrolysis of ATP to the export of three intracellular Na ions and the import of two extracellular K ions. The continuous operation of this macromolecWar machine ensures the generation and maintenance of concentration gradients of Na and K across the cell membrane. This electrochemical gradient provides energy for the membrane transport of metabolites and nutrients, e.g., glucose and amino acids, and such ions as protons, calcium, chloride, and phosphate. The electrochemical gradient is essential also for regulation of cell volume and for the action potential of muscle and nerve. The relative intra- and extracellular concentrations of Na and K ions maintained primarily by the sodium pump and the cofactors required for activity are shown in Fig. 1 (1, 2). The functional macromolecule is a membrane-spanning 270-kDa tetramer consisting of two dimers, each composedof noncovalently interacting alpha (112 kDa) and beta (55 kDa) subunits. A model of the transporter complex relative to its membrane location is shown in Fig. 2. The presence of a smaller gamma subunit (10 A OUT (150 mmol/L) Digitalis (5 mmol/L) Y1016 ‘)63K327 IN Fig. 1. SchematIc diagram of the Na,K-ATPase-associated cofactors and Ions. Modifiedfrom Akera (2). Extracellular 1263 R166 0737 Y1018 IN Intracellular Fig. 3. Foldingmodels for the Na,K-ATPase alpha transmembrane segments: (A) 10 membrane-spanning segments; (B) 8 membranespanning segments. a a Fig.2. Schematic diagram of the plasma membrane-spanning Na,KATPase transporter complex, indicating the positions of the alpha, beta, and putative gamma subunits. kDa) has been suggested; however, its role, if any, has not been defined (3). The alpha (catalytic) subunit, is proposed to have 7(4) or 8(5) transmembrane domains; however, the number can vary from 6 to 10, based on interpretation of hydropathy profile data (6) and identification of specific ligand-receptor interactions that predict alpha chain topology (7-9). The alpha subunit contains all the binding sites for ligands known to stimulate or inhibit the enzyme (10-13). Tentative models representing membrane-spanning segments of the alpha subunit are de- Letters refer to the one-letter amino acid code. Numbers represent the topological location of particular amino adds. From Kailish et at. (7); used with permission. cyclic reaction in which the enzyme is phosphorylated by ATP in the presence of Mg2 ions and Na ions and then dephosphorylated in the presence of K ions. A model depicting this thermodynamic cycle is shown in Fig. 4. Isoform Regulation and Genetic Expression With the advent of molecular biological techniques, three alpha and four beta isoforms of the Na,K-ATPase have been identified, which are encoded by independent genes (20-22). The sequence conservation among differ3Na 2l( tailed in Fig. 3. The beta subunit has a single hydrophobic transmem- brane domain and is highly glycosylated on its noncytosolic surface (14). Hiatt et al. postulate that the beta may serve to orient and stabilize the alpha subunit in the membrane (15). Cellular expression of the beta subunits and assembly with the alpha subunits are necessary for correct conformation and activity of the Na,K-ATPase holoenzyme (16). Although the role of the beta subunit in ion translocation is uncertain, its presence appears essential for function of the sodium pump (16). Phosphorylation is an important step in the function of Na,K-ATPase. The molecule undergoes an alpha-helix to beta-sheet transition between two principal reactive states, E1 and E2, in the multistep reaction by which Na ions and K ions traverse the membrane (17, 18). The conformational transition results via a subunit .p E2-P.Na3 I Ei-P.Naa1,... r E2P.K2 AlP Nat. Mi2 E1 ATP ______________________ E2-K2 . 2K Fig. 4. Principal reactive states (E1 and E) involved in the transport of sodium (Na) and potassium (K) ions across the cytoplasmic membrane. The enzyme (E) Is phosphorylated(P), with ATP as thephosphatedonor and Mg2 as a cofactor In the reaction. From MacGregor and Walker (19), as modified from Sen et at. (18); used with permission. CLINICAL CHEMISTRY, Vol. 40, No. 9, 1994 1675 ent species suggests that individual roles for the isoforms, though not yet determined, arose early and were maintained throughout evolution. The three alpha isoforms (alpha 1, 2, and 3) are expressed in a developmental and tissue-specific manner. Utilizing monoclonal antibodies that recognize the inchvidual alpha isoforms, Lucchesi and Sweadner have shown (23) that rat ventricular muscle-membrane preparations express alpha 1 in all stages of development; alpha 3 is present at birth through days 14 to 21, and is then replaced by alpha 2 in the adult rat; alpha 3 again predominates in aged rats. The physiological significance of this shift in subunit isoform expression is unknown. Tissue specificity for the different isoforms has been identified both at the mRNA and protein level for various species (22, 24), and is summarized for isoforms from rat and human tissues in Table 1. Table 1, although not comprehensive, serves to illustrate the diversity of isoform distribution in tissues. The vast majority of studies defining isoform distribution have been done with animals, less data being available for human tissues. Nevertheless, studies in both humans and rats suggest that alpha 1 is the only isoform expressed appreciably in the kidney (33), whereas alpha 3 is associated primarily with the nervous system (22, 28, 29). Isoform specificity even extends to cell type within a particular tissue, as evidenced by studies with tissue from brain (34), heart (35), and eye (30), supporting the hypothesis that the isoforms have different physiological functions (36, 37). Three Na,K-ATPase beta isoforms and one related H,K-ATPase (another member of the cation-transporting ATPases) beta isoform have been identified. Beta 1 has been isolated from several vertebrate species in a wide range of tissues. Beta 2 is expressed largely in brain (26,27) and ocular ciliary epithelia (38); however, recent studies suggest that beta 2 is also expressed in glycolytic fast-twitch muscles of the rat hindlimb (32). Table 1. Tissue dIstribution of Na,K-ATPase Isoforms. Tissu. source Species Adipose Rat Brain Rat Human Human Human Human Human Eye Heart (ventricle) Rat Rat Rat Kidney Lung Skeletal muscle Human Human Human Rat Thyroid Uterus Human Human References Subunit Regulators of Sodium Pump Activity Cardiac Glycosides al, a2’ f32#{176} al a2, a 25 26, 27 The Na,K-ATPase alpha subunit receptor for the cardiac glycosides, a3b 29 30 the clinical significance congestive heart failure 31 and Walker have written a short review of the cardiac glycosides (19). These inhibitors of the sodium pump are derived from extracts of the plant genera Digitalis, Strophanthus, and Acocanthera. Digoxin and digitoxin are products of species of the foxglove plant, Digitalis, and ouabain is obtained from the East African Ouabaio tree or seeds of the plant Strophanthus gratus (52,53). These compounds are the most potent inotropic agents known, and their cardiac effects are believed to be mediated through their ability to inhibit the sodium pump. Historically, preparations of these substances have been used therapeutically for perhaps 3000 years, including use of plant extracts containing cardiac glycosides by the ancient Egyptians (54). al, a2, a3ab al, a2, a3, a al, a2, a3a al, a3 neonate#{176} al, a2 adult#{176} al, a3 aged#{176} ala ala al, a1,a2,l,i32a al, a2 ala 28 23 23 23 28 28 28 32 28 28 Superscripts referto all the preceding isoforms onthe same line: a isoformspecific expression of mRNA identified byNorthernanalysis;b isoform-specific expressionof proteinidentifiedby Westernanalysis. 1676 Interestingly, the beta 2 isoform appears to serve a dual function, both as a subunit of the Na,K-ATPase and as a mediator of neuron-astrocyte adhesion (39). This suggests the possibility that these proteins, as a family, may play other roles besides their traditionally defined transport function. Finally, a protein referred to as beta 3 has been isolated from Xenopus (40), and the betaisoform of the H,K-ATPase has been characterized from several vertebrate species (41-43). The beta isoforms are less similar to each other in amino acid sequence than are their alpha isoform counterparts to each other; the beta isoforms also vary in their number of asparagine-linked glycosylation sites (27). The expression of all the alpha isoforms and beta 1 are differentially regulated by hormones (44). Horowitz et al. (45) determined that thyroid status affected the alpha 1, alpha 2, and beta isoform-specific expression of mRNA and protein in rat heart, skeletal muscle, and kidney; and Gick and Ismail-Beigi (46) found that incubation of a rat liver cell line with thyroid hormone resulted in an increase in alpha 1 and beta mRNA expression and Na,K-ATPase activity. Increased sodium concentration in response to corticosteroids is reported to recruit an intracellular latent pool of Na,KATPase complexes to the cell membrane in the cortical collecting tubules of rat (47) and rabbit (48) kidney. Lingrel et al. (49), examining the 5’-flanking sequences of the human alpha isoform genes, found that each contains a number of potential transacting and hormonebinding sites that do not appear to be conserved among the three alpha isoform genes, thus allowing for differential regulation. The ability to detect nucleotide changes that result in restriction fragment length polymorphisms has led to the discovery of sequence variation in some families in the human alpha (50) and beta (51) subunit genes. Whether these genotypic differences, or others not yet identified, correlate with a particular pathology is at present undetermined. CLINICAL CHEMISTRY, Vol. 40, No. 9, 1994 is the only known which underscores of the pump in treatment of MacGregor and arrhythmias. Digoxin, which can be administered orally and is readily absorbed by the gastrointestinal tract, is the most widely used cardiac glycoside clinically; ouabain is the most widely used experimentally. These compounds, all cardenolides, have a sterol skeleton. Cardenolides are C steroids having one or more sugar residues at C-3 and a flve-membered lactone ring at C-17 (see Fig. 5). As previously described, cardiac glycoside binding to Na,K-ATPase occurs on the extracellular face of the integral membrane protein. However, recent data suggest that a hydrophobic binding pocket that contains membrane-spanning amino acids may be involved as well (55). Alpha subunit amino acids important for cardiac glycoside binding have been determined (6, 56), and recent studies with site-directed mutagenesis to make amino acid substitutions at proposed binding sites have helped determine which regions recognize cardiac glycoside sugars (57). A first-order-approximation binding model of the interaction of digoxin with Na,KATPase has been proposed by Thomas (58) and involves a folding of the receptor-binding epitopes around the ligand. Fig. 6 depicts the intermolecular forces that might play a role in the interaction between the lactone ring, sterol section, and sugar residues of digoxin. Regardless of the details, the interaction is very specific, with dissociation constants in the iO mol/L range (59), and clearly leads to selective inhibition of the activity of Na,K-ATPase. Seminal work demonstrating the inhibitory effect of cardiac glycosides on Na.,K-ATPase has been performed by Akera and Brody (60). Akera et al. (61) were the first to compare the in vivo sensitivity of the sodium pump to ouabain in dog, sheep, guinea pig, and rat with the in vitro sensitivity of Na,K-ATPase in cardiac microsomal fractions from these same species. Inhibition of the sodium pump by cardiac glycosides increases the strength OH OH RHAMNOSE: RHAMNOSE OH OH H DIGrTOXOSE DIGITOXOSE DIGITOXOSE DIGITOXOSE: H NO OH OH Fig. 5. Structures of the cardiacglycosides,digoxinand ouabain. Three digitoxose sugars (Indigoxin)and one rhamnose sugar (in ouabain) are attached at the C-3 position of the steroid backbone. AECP7(: SITED H-BINDING SITES 11 Fig. 6. A model for drug (ouabain, digoxin) interactionwith the receptor (Na,K-ATPase). FromThomas (58); used with permission. of contraction (inotropic effect) and slows the beating (chronotropic effect) of the heart. Membrane excitation of cardiac myocytes is characterized by opening of the Na channel and depolarization of the sarcolemmal membrane in response to increased intracellular sodium. Consequently, Ca2 channels open and the Ca2 ion influx triggers the release of Ca2 stores from the sarcoplasmic reticulum into the cytosol. The increase in intracellular free Ca2 activates contractile proteins, resulting in myocardial contraction. Cardiac glycosides inhibit the exchange of Na and K via Na,K-ATPase; the result is a relative transient increase in intracellular sodium. The Na/Ca-ion exchanger, present in the sarcolemmal membrane, mediates the exchange of Na ions for Ca2 ions. This exchange mechanism probably results in a relative increase in the concentration of intracellular Ca2. The increased intracellular Ca2 is taken up into the sarcoplasmic reticulum via a Ca2 pump. After depolarization, the extra Ca2 released results in enhanced contractile force (1). Somberg et al. (62) determined that a 25% reduction in pump activity was associated with a 20% increase in contractile strength. The cardenolides that specifically interact with the sodium pump also have well-documented effects on other cardiovascular organs such as the peripheral vascular tissue (see below). Even though digoxin is widely used in the treatment of heart disease, the therapeutic index is low, and Smith et al. (63) cite digoxin intoxication as the most widely encountered adverse drug reaction in clinical practice. Symptoms of digoxin toxicity commonly involve the gastrointestinal tract and the central nervous system and include: anorexia, nausea, vomiting, diarrhea, headache, delirium, cardiac rhythm disturbances, manic syndrome, and depressive syndrome (64, 65). One explanation is due to what Langer terms the “sodium pump lag” effect (66) explained above CUNICAL CHEMISTRY, Vol. 40, No. 9, 1994 1677 and described in the flow diagram in Fig. 7. The desirable outcome to the proposed sequence of events described in Fig. 7 is positive inotropy, but some individuals may experience symptoms of cardiac glycoside toxicity as a result of abnormally high intracellular concentrations of calcium. Low tolerance to cardiac glycosides has been associated with old age, acute myocardial infarction/ischemia, hypoxemia, magnesium depletion, renal insufficiency, hypercalcemia, carotid sinus massage, electrical cardioconversion, hypothyroidism, and hypokalemia (67). Interaction with coadministered drugs such as quinidine, verapamil, and cyclosporine is a frequent cause of toxic accumulation of diguxin. Recent evidence suggests that these drugs inhibit renal excretion of digoxin by inhibiting the MDR1 gene product, P-glycoprotein, shown to be present on the apical membrane of mammalian kidney (68). P-glycoprotein is overexpressed in multidrugresistant cells and functions as a drug-efflux pump, recognizing a variety of therapeutic agents, including vinblastine, a known P-glycoprotein substrate. Significant accumulation of digoxin or vinblastine has been reported in both a multidrug-resistant chinese hamster ovary cell line and the drug-sensitive parent cell line when cyclosporine, verapamil, or quinidine was added to the culture medium (69). This suggests digoxin excretion is also mediated by P-glycoprotein. Understanding the pharmacokinetics of the cardiac glycosides in relation to renal excretion, age-related tolerance, and interaction with coadministered drugs has led to better patient management and a decrease in toxicity and mortality. However, therapeutic drug monitoring practices for this drug (e.g., reference ranges, toxic concentrations, dosing regimens) stifi vary considerably throughout clinical laboratories. Examining 666 institutions participating in Q-Probes (a subscription quality-improvement program of the College of Clinical Pathologists), Howanitz and Steindel (70) found that participants used 13 different lower limits (0 to >1.0 CARDIAC GLYCOSIDES 4, INHIBITION OF Na , - ATPase BY BINDING AT EXTRACELLULAR ENZYME SURFACE 4, INCREASE IN INTRACELLULARNa CONCENTRATION ION 1 INCREASE IN INTRACELLULAR Ca2 POSITIVE INOTROPY Fig. 7. Schematic representation ION CONCENTRATION Ca2 OVERLOAD LEADING TO CARDIAC GLYCOSIDE TOXICITY of the extra- and intracellular events that lead to increasedcontractileforce (positiveinotropy)or possible cardiac glycoside toxicity. From MacGregor and Walker (19); used with permission. 1678 CLINICAL CHEMISTRY, Vol. 40, No. 9, 1994 g/L) and 16 different upper limits (1.1 to >2.7 pg/L) for their therapeutic digoxin ranges. Some blood samples were drawn <6 h after dosing. Blood sampling at inappropriate times may result in digoxin serum concentrations in excess of the therapeutic range, and thereby increase the likelihood of an erroneous decision to withhold digoxin or even to administer immunoglobulin fragments of digoxin-specific antibodies (used in the treatment of digoxin toxicity) (70). Additionally, in some individuals, digoxin is biotransformed into metabolites with variable cross-reactivity in digoxin immunoassays (71, 72). Even if these metabolites are not biologically active, their cross-reactivity in digoxin immunoassays could result in digoxin underdosing (72, 73). However, we (72) and others (74) have found that some but not all of these metabolites have significant Na,K-ATPase inhibitory activity, the clinical importance of which has not been fully addressed. Recent important findings suggest that the alpha subunits of the Na,K-ATPase exhibit species and isoform variation in their affinity for binding of cardenolides (75). Differences in binding affinities, previously ascribed to variables such as assay conditions and ion concentrations, now include differences attributable to the presence of high- and low-affinity alpha subunit molecular forms. Originally termed “a” and “a+ ,“ the molecular cloning of the Na,K-ATPase from various species demonstrated that a+ is actually represented by the two distinct isoforms now referred to as alpha 2 and alpha 3. The alpha 2 and alpha 3 subunits are the isoforms with greatest sensitivity to ouabain in the rat (76-78). Charlemagne et al. (79) describe high- and low-affinity ouabain-binding sites in rat heart, with respective apparent dissociation constants in the 10-8 to 10_6 mol/L range. Age, ion concentration, hormone concentrations, and pathological conditions have all been demonstrated to correlate with changes in isoform expression (37). Isoform variation in affinity for digoxin and other cardiac glycosides should be considered in cases where the response to cardiac glycoside therapy is inappropriate. However, work in this area is still preliminary. For example, Schmidt et al. (80) quantified the digitalis receptor concentration in the left ventricle at autopsy, comparing patients without heart disease with those with end-stage heart failure who had received digitalis therapy. Previous work, based on the use of in vitro systems and tissue culture, had shown an increase in expression of Na,K-ATPase in response to incubation with digitalis, leading to speculation that patients might develop tolerance to digitalis therapy (81). However, rather than increased expression, Schmidt et al. (80) showed a lower concentration of digitalis receptors in failing hearts than in the control subjects. These investigators did not address the presence or absence of specific high- or low-affinity alpha isoforms, because their quantification of digitalis receptors was based on [3Hlouabain binding. Affinity for ouaham is affected by the presence or absence of certain amino acid residues at the amino terminus of the alpha subunit that correspond to isoform type (82); therefore, the down-regulation of high-affinity alpha isoforms without an overall loss in holoenzyme concentration could give the same experimental results as a decrease in overall Na,K-ATPase expression. Phenomena such as changes in subunit expression, distribution in tissues, ligand binding affinity, and endogenous ligand concentrations hold promise for establishing a new understanding of the basic mechanisms underlying pathophysiology. These findings maybe of central importance in establishing hypotheses regarding the clinical role of endogenous ouabain- or digitalis-like factors. EndogenousLigands:Implicationsin Pathology The ubiquitous nature of the sodium pump and its involvement in diverse physiological processes suggests that alteration of pump activity by endogenous or xenobiotic factors may play a key role in many fundamental physiological processes (e.g., modulation of cardiac contractility, control of sodium in the kidney, vascular contractility, neurotransmitter release and processing) (19). The presence of a highly conserved Na,K-ATPase binding domain for cardiac glycoside drugs implies the existence of natural ligands that act as endogenous modulator(s) of this transporter. Substantial evidence suggests that endogenous digitalis-like and ouabain-like factors exist. In the process of monitoring therapeutic digoxin concentrations by various immunoassay procedures, several investigators noted increased digoxin values in subjects not treated with cardiac glycosides (83, 84). Also noted were increases in serum digoxin measurements in subjects whose digitalis therapy had been discontinued (85). Factors giving rise to these apparent digoxin values were termed digoxin-like immunoreactive factors (DLIF) (84) or endogenous digitalis-like factors (EDLF) (86). Detectable concentrations of these factors have been observed in serum and plasma from healthy adults (87), plasma from volume-expanded dogs (86), newbOrns (88, 89), pregnant women (90), patients with renal impairment (91), and patients with liver dysfunction (92, 93). Aside from DLIF interference with the accurate measurement of digoxin in human serum, these molecules, because of their structural similarity to digoxin itself (94), may interact with the Na,K-ATPase at the digitalis-binding site on the alpha subunit. Present evidence suggests that the likely tissue source of this factor is the adrenal cortex (94, 95). In addition to the discovery of DLIF, there is compelling evidence that ouabain-like factors (OLF) are present in mammals (96). Hamlyn and Manunta (97) and other investigators (98) have isolated a factor from both serum and adrenals with ouabain-like properties that include structural similarity to ouabain, Na,K-ATPase inhibitory activity, and increased concentration in pathophysiological conditions. A review of this work and the potential role of this ouabain-like factor in disease are detailed by Blaustein (53), who summarizes the proposed physiological effects of endogenous ouabain in control of intracellular calcium stores and cell respon- siveness. Controversy remains, however, about the source of this endogenous ouabain-like molecule (99). Substantial arguments still prevail as to what is meant by digitalis-like activity (100). It is important to understand that immunoreactivity does not imply functional activity or vice versa. Thus, DLIF should not be mistaken or confused with EDLF or OLF (84), even if, as is suspected, the identity of these factors converges as more is learned about them. For example, these molecules may be related precursors, metabolic products of each other, or the same molecule. Substantial evidence links endogenous digitalis-like factors with vasoreactivity. Data supporting the hypothesis that endogenous digitalis-like factors interact with Na,K-ATPase to induce peripheral vasoconstriction include the following 1) Subjects with some forms of essential hypertension have increased sodium-potassium pump inhibitory activity (101, 102), natriuretic activity (103), and digoxin immunoactivity in their plasma (104). 2) Spontaneously hypertensive rhesus monkeys have high serum concentrations of digoxin-like activity (105). 3) Crude preparations of the natriuretic activity constrict third-order arterioles, making them more responsive to other vasoconstrictive agents such as norepinephrine (106). 4) Crude preparations of the natriuretic activity from urine cause dose-dependent contractions of isolated anococcygeus muscle of the rat (which resembles the smooth muscle of blood vessels) (107). 5) Infusion of ouabain (108, 109) or digoxin (110) into humans specifically induces peripheral vasoconstriction. 6) Injection of antibodies to digoxin lowers the blood pressure of deoxycorticosterone-salt-retamning hypertensive rats (111). 7) Preparations containing digoxin-like immunoreactivity from human urine raise blood pressure and protect rats from acute digitalis toxicity (112). 8) Bolus infusion of digoxin induces vasoconstriction of epicardial coronary arteries in humans (113). 9) Ouabain-like compounds isolated from human serum demonstrate vasoreactivity comparable with that of ouabain (114). Cumulatively, the digitalis-like activity of these factors strongly implicate endogenous regulators of the Na,K-ATPase as vasoconstrictive agents involved in the etiology of some hypertensive states. Clinical Conditions Linked to Dysfunction or Modification of Na,K-ATPase Activity CardiovascularDisease and Hypertension Pathological conditions in animals and humans involving salt and water homeostasis have been associated with alterations in Na,K-ATPase activity and (or) the presence of circulating endogenous digoxin- or ouabain-like factors (germane articles and reviews are provided in Table 2). Two of the most notable disorders involving salt and water homeostasis-heart disease CLINICAL CHEMISTRY, Vol. 40, No. 9, 1994 1679 Table 2. Clinical conditions correlating with altered Na,K-ATPase activity or presence of modifiers. Condition . Cardiovascular disease and hypertension Heart disease Hypertension Reference 52. 97, 101, 117, 118, 119 HTN, pregnancy-induced 120, 121, 122 HTN, 123 hypothyroidism-related Impaired renal function 91 Renal disease 84, 85, 124 Diabetes and other metabolic disorders Acromegaly Aldosteronism Diabetes 119, 125 126 118, 119, 127, 128 119 mellltus Obesity Digoxin toxicity Age-related Disease-related Multipledrug interaction . Fetal abnormalities Growth retardation Renal abnormality H droce halus 64,65, 67 69 . 130 130 130 aneuploidy Nonimmune hydrops Low birth weight 131 Preterm infants 132 ‘ Neurological disorders Alzheimer disease Bipolar disorder Pulmonary conditions Pulmonary disease Chronic obstructive pulmonary disease . one-kidney, one-clip, and reduced renal mass-saline models of hypertension (102, 139). They demonstrated decreased myocardial and vascular Na,K-ATPase activity, suggesting that reduced pump activity might be common in low-renin and other models of essential hypertension. In a more recent study, uninephrectomized animals treated with deoxycorticosterone or angiotensin II provided insight into the molecular mechanisms that may be involved in some forms of hypertension (140). Separation of mRNA by Northern analysis of rat cardiac left ventricle, aorta, and skeletal muscle RNA, by use of Na,K-ATPase alpha isoform-specific cDNA probes, showed tissue-specific changes in isoform expression of mRNA transcripts in response to increased intravascular pressure. Greater concentrations of DLIF and OLF have been noted in women with pregnancy-induced hypertension (preeclampsia) than in normal pregnancy (84, 141). Pregnancy is a volume-expanded condition, and in both normal and hypertensive pregnancy the anomalous DLIF values resolve rapidly upon delivery (90, 120122). Increased concentrations of DLIF are associated with acute and chronic renal disease (91, 124) and with fluid retention due to hepatic failure (92, 93). Endogenous OLF is reported markedly increased in hypertension that is due to hypothyroidism (123) and congestive heart failure (116). DLIF is also significantly increased in the plasma of human subjects with electrocardiographic evidence of reversible cardiac dysfunction induced by physical exhaustion (142). Weinberg et al. (143) used peritoneal dialysis fluid from patients with . 80 115 116 (HTN) Early experiments by Pamnani et al. aimed at underthe effects of hypertension on the myocardium and vasculature at the molecular level by using rat standing 130 133, 134, 135, 136 64 137 138 chronic and hypertension-are causally related. Hypertension increases the risk of myocardial infarction, congestive heart failure, renal failure, and cerebral stroke (52). The initial reduction in myocardial contractility that occurs in some forms of heart failure results in vasoconstriction and peripheral resistance. The constriction of the vascular beds in the kidneys causes salt and water retention. Alterations in Na,K-ATPase activity or expression can alter vascular or cardiac contractility by affecting sodium homeostasis (37). Thus, the sequence of events previously described strongly correlates with the involvement of a circulating inhibitor of sodium pump activity in the pathogenesis of both cardiovascular disease and hypertension. Hypertension, as evidenced by persistently high arterial blood pressure, can be idiopathic or secondary to underlying conditions. Essential (idiopathic hypertension) is a poorly understood though relatively common disease. An inherited predisposition has been suggested, and such individuals may be especially sensitive to dietary salt (117). Hypertension secondary to primary aldosteronism (126), other endocrine disorders, and pregnancy often tends to resolve once the underlying problem is alleviated, 1680 CLINICAL CHEMISTRY, Vol. 40, No. 9, 1994 renal failure to chromatographically isolate three molecular species having DLIF activity. Of the three, one had a retention time identical to ouabain, and one had a retention time identical to digoxin. And Yuan et al. (144) showed that administration of chronic low doses of ouabain was associated with the development of hypertension in normotensive rats as well as in rats having various degrees of reduced renal mass. Mean blood pressure increased with the degree of mass reduction but was significantly greater than in controls even for rats with no renal mass reduction. In an effort to determine whether ouabain itself acts as a hypertensive agent or simply exacerbates the hypertensive action of mineralocorticoids, Sekihara et al. (145) treated mononephrectomized rats with ouabain (1 mg), deoxycorticosterone acetate (5 mg), or a combination of both, weekly for 6 weeks. Both ouabain and deoxycorticosterone acetate lacked hypertensive action individually at the dosage given but, in combination, they produced a significant increase in blood pressure as well as cardionephromegaly and histopathological changes consistent with the effects of an elevation in blood pressure. The authors concluded that, in those hypertensive individuals secreting greater concentrations of mineralocorticoids, ouabain might amplify the hypertensive effect. DLIF, isolated from urine by use of cross-reactivity to digoxin antibody as evidence of activity, was admiriistered to normotensive rats to determine its cardiovascular effects (112). After an initial 60-mm stabilization period, infusion of DLIF in 10 animals caused the mean arterial pressure to rise from 124.3 (±1.9) to 140.4 (±5.2) mmHg at 7.5 mm, induced diuresis, and slowed the heart rate. These results, coupled with data documenting the presence of endogenous digitalis-like compounds in the serum of hypertensive animals (102, 146, 147), suggest a role for DLIF in hypertension. Diabetes and Other Metabolic Diseases Weidmann and Ferrari, studying diabetes and hypertension (127), found that not only do type I (insulin-dependent) diabetic individuals have a familial predisposition for essential hypertension, but also normotensive offspring of parents who are nondiabetic but have essential hypertension show increased concentrations of plasma insulin and reduced insulin sensitivity; moreover, Na retention is characteristic of both type I and type II (non-insulindependent) diabetics. These authors also report that intracellular calcium is increased in adipocytes, in part via insulin’s inhibition of Ca2,Mg-ATPase, and that insulin may increase renal sodium retention and influence the activity of transmembrane electrolyte pumps. Insulin regulation of vascular Na,K-ATPase gene expression is cited by Tirupattur et al. (148) as an important factor in the development of hypertension in diabetes. mRNA encoding both the alpha 1 and alpha 2 isoforms was identified in vascular smooth muscle cells derived from embryonic rat thoracic aorta. Although the predominant isoform was alpha 1, only the concentrations of the alpha 2 isoform increased in response to insulin treatment. The overall increase in ouabain-inhibitable Na,K-ATPase activity in vascular smooth muscle cells in response to insulin treatment suggests that, in the absence of insulin or in insulinresistant states, Na,K-ATPase activity could decrease, resulting in increased vascular contractility and blood pressure. In their review, Clerico and Giampietro (119) cite reports of decreased sodium pump activity in the nerves, heart, and aorta of diabetic humans, and in rats with streptozotocinor alloxan-induced diabetes; however, there was a paradoxical increase in Na,K-ATPase activity in the kidneys of streptozotocin-induced diabetic rats. The authors speculate that these tissue differences could be due to tissue-specific regulation of Na,KATPase activity or metabolism. They further suggest that in metabolic diseases such as diabetes meffitus, obesity, and acromegaly (which have in common increased sodium retention and volume expansion) increased sodium intake, hyperinsulinemia, or increased concentrations of growth hormone could trigger the release of an endogenous digitalis-like factor that could modulate the pump and increase blood pressure. A more recent study by Chen et al. (128), using a similar diabetic rat model system, confirmed the previous findings. In addition, Chen et al. found greater amounts of a digitalis-like factor in the plasma and urine of their hypertensive diabetic rats than in controls. A review by Sewers and Khoury (118) highlights the additional risk of cardiovascular disease when hypertension accompanies diabetes mellitus and discusses additional risk factors such as obesity, genetics, and ion transport control. Digoxin Toxicity Although advanced age is not a pathological condition, age-related changes in renal clearance and volume of distribution can increase the likelihood of digoxin toxicity (65). In addition, evidence based on basal metabolic rate measurements indicates that whole-body Na,K-ATPase activity decreases with age (64, 129). This suggests that, even though their serum cligoxin concentrations may be within the therapeutic range, digitalis toxicity in the elderly might result, in part, from inhibition of an already less-active sodium/potassium transport system. Low tolerance to digoxin is also associated with certain disease states, particularly those involving hypokalemia, and with multiple drug interaction, as discussed previously. Fetal Abnormalities The neonatal period is associated with volume expansion and sodium imbalance. As stated previously, agerelated changes in Na,K-ATPase alpha isoform expression in the rat have been observed (23). DLIF is significantly increased in the plasma of newborn infants but usually returns to normal within several days (88, 132). Increased concentrations of digoxin-like immunoreactivity have been observed in fetuses having growth retardation, renal abnormality, hydrocephalus, aneuploidy, and nonimmune hydrops (reviewed in ref. 130), again suggesting a role for endogenous pump modifiers in the regulation and genetic expression of Na,KATPase and in the etiology of some pathological processes. Neurological Disorders Bipolar illness (manic depression) is characterized by severe mood swings that alternate between episodes of irritability, excessive energy, and distractibility (mania), and mental and motor slowdown to the point of stupor (depression). Patients with bipolar illness exhibit altered ion distribution and transport. In one model, El-Mallakh et al. (64) propose a biphasic phenomenon, in which mild or moderate reduction in Na,K-ATPase activity could lead to mania by increasing membrane excitability and neurotransmitter release. A greater degree of pump inhibition, and consequently depolarization block, could result in depression by decreasing neurotransmitter release (149). Patients in manic states show increased sodium retention and intracellular calcium concentrations, and therapeutic modalities such as lithium or calcium-channel blockers are theorized to affect sodium-calcium exchange (64). Interestingly, symptoms mimicking those observed in bipolar patients occur with digitalis toxicity. These can include confusion, disorientation, drowsiness, lethargy, agitation, and hallucinations (137), further implicating Na,KCLINICAL CHEMISTRY, Vol. 40, No. 9, 1994 1681 ATPase as an etiological agent in bipolar illness. Recent evidence in our laboratory suggests that DLIF (measured with a digoxin-specific immunoassay) is increased in acutely psychiatrically ill bipolar patients relative to recovered bipolar patients (unpublished). Increased endogenous ouabain-like compounds that excessively suppress Na,K-ATPase have been proposed as a mechanism in this disorder (133). Alzheimer disease (Al)) is an age-related neurodegenerative disorder of unknown etiology. In addition to well-documented neuropathological changes such as neurofibrillary tangles and neuritic plaque formation with accumulation of beta-amyloid (136), AD is associated with lower cerebral blood flow and decreased use of oxygen and glucose, especially in areas exhibiting neuropathological changes (134). About 50% of the energy expenditure of resting brain is believed to support Na,K-ATPase activity. Hank et al. (134) found a decrease in ouabain binding of brain tissue from AD patients in comparison with age-matched controls. The reduction in ouabain binding was especially evident in the neocortex, an area predominantly affected in Al) patients. A decrease in ouabain-inhibitable Na,KATPase activity in the brain subcortical but not cortical areas of patients with Al) was also noted by Liguni et al. (135). Although sodium pump dysfunction may not be causal in this disorder, the progressive dementia associated with AD may in part be due to alterations in Na,K-ATPase activity. To date, the presence of possible endogenous pump modifiers in relation to AD has not been described. Pulmonary Conditions Little has been published in this area. Varsano et al. (138) used a digoxin radioimmunoassay to show that patients with advanced chronic respiratory failure had greater concentrations of DLIF than did controls. Chronic obstructive pulmonary disease and other forms of advanced respiratory disease are frequently associated with water and sodium retention, and the authors suggested that increased DLIF might be an attempt to control water and sodium metabolism. In summary, conditions associated with volume expansion or alterations in sodium homeostasis (see Table 2), many of which have been described here, also show changes in Na,K-ATPase activity, increases in endogenous digitalis-like substances, or both. Modification of pump activity can occur in response to changes in isoform expression or modulation by endogenous pump inhibitors or activators. As to whether altered regulation of Na,K-ATPase activity is directly linked to any of these conditions remains to be elucidated. Preliminary data identifying polymorphisms in the human Na,KATPase alpha and beta subunit genes offer the potential of linking these and other genetic differences to specific clinical disorders. Locating genetic mutations of Na,KATPase or of the endogenous modifiers may soon be added to the repertoire of test capabifities in the clinical chemistry laboratory, providing early identification of 1682 CLINICAL CHEMISTRY, Vol. 40, No. 9, 1994 individuals at risk for a variety ciated diseases. This work was supported of Na,K-ATPase-asso- in part by Grant RO1-HL36172 from the National Institutes of Health. References 1. Fozzard HA, Sheets MF. Cellular mechanism of action of cardiac glycosides. J Am Coil Cardiol 1985;5:10-5A. 2. Akera T. Membrane adenosinetriphosphat.ase: a digitalis receptor? Science 1977;198:569-74. 3. Forbush B, Kaplan JH, Hoffman JF. Characterization of a new photoaffinity derivative of ouabain: labeling of the large polypep- tide and of a proteolipid component of the Na,K-ATPase. Biochemistry 1978;17:3667-76. 4. Ovchinnikov YA, Arzainazova NM, Arysturkhova EA, Gevondyan NM, Aldanova NA, Modyanov NN. Detailed structural analysis of exposed domains of membrane-bound Na,K-ATPase. FEBS Left 1987;217:269-74. 5. Shull GE, Schwartz A, Lingrel JB. Amino acid sequence of the catalytic subunit of the (Na’,K) ATPase deduced from a complementary DNA. Nature 1985;316:691-5. 6. Lingrel JB, Orlowski J, Shull MM, Price EM. Molecular genetice of Na,K-ATPase. Prog Nucleic Acid Res Mol Biol 1990;38:3789. 7. Karlish SJD, Goldshleger R, Jorgensen PL. Location of Asn’ of the a chain of Na/K-ATPase at the cytoplasmic surface. Implication for topological models, J Biol Chem 1993;268:3471-8. 8. Karlish SJD, Goldshleger R, Tal DM, Stein WD. Structure of the cation binding sites of Na/K-ATPase. In: Kaplan JH, DeWeer P, eds. The sodium pump: structure, mechanism, and regulation. New York: Rockefeller Univ. Press, 1991:129-41. 9. Mohraz M, Arystarkhova E, Sweadner KJ. Immunoelectron microscopy of epitopes on Na,K-ATPase catalytic subunit. J Biol Chem 1994;269:2929-36. 10. Farley RA, Trant CM, Carilhi CT, Hawke D, Shively JE. The amino acid sequence of a fluorescein-labeled peptide from the active site of (Na,K)-ATPase. J Biol Chem 1984;259:9532-5. 11. Karlish SJD, Goldshleger R, Stein WD. A l9kDa C-terminal tryptic fragment of the a chain of Na/K-ATPase is essential for occlusion and transport of cations. Proc Natl Aced Sci USA 1990;87:4566-70. 12. Rogers TB, LazdunskiM. Photoaflinity labeling of the digitalis receptor in the (sodium + potassium)-activated adenosinetriphosphatase. Biochemistry 1979;18:135-40. 13. Walderhaug MO, Post RL, Saccomani G, Leonard RT, Briskin DP. Structural relatedness of three ion-transport adenosine triphosphatases around their active sites of phosphorylation. J Biol Chem 1985;260:3852-9. 14. Farley RA, Miller RP, Kudrow A. Orientation of the 13subunit in the cell membrane. Biochim Biophys Acta 1986;873:136-42. 15. Hiatt A, McDonough AA, Edelman IS. Assembly of the (Na,K)-adenosine triphosphatase. Post translational membrane polypeptide of (Na +K)ATPase integration of the alpha-subunit. J Biol Chem 1984;259:2629-35. 16. McDonough AA, Geering K, Farley RA. The sodium pump needs its 13subunit. FASEB J 1990;4:1598-605. 17. Jorgensen PL. Structure, function and regulation ATPase in the kidney. Kidney Int 1986;29:1O-20. of Na,K- 18. Sen AK, Tobin T, Post RC. A cycle for ouabain inhibition of sodium and potassium-dependent adenosine triphosphatase. J Biol Chem 1969;244:6596-604. 19. MacGregor SE, WalkerJM. Inhibitors of the Na,K-ATPase [Reviewl. Comp Biochem Physiol 1993;105C:1-9. 20. Ovchinnikov YA, Monastyrskaya GS, Broude NE, Alhikmeta RL, Ushkaryov YA, Melkov AM, et al. The family of human Na,K-ATPase genes. A partial nucleotide sequence related to the a-subunit. FEBS Left 1987;213:73-80. 21. Shull GE, Greeb J, Lingrel JB. Molecular cloning of three distinct forms of the Na,K-ATPase a-subunit from rat brain. Biochemistry 1986;25:8125-32. 22. Sweadner KJ. 1991. Overview: subunit diversity in the Na,KATPase. In: Kaplan JH, DeWeer P, eds. The sodium pump: mechanism, and regulation. New York: Rockefeller Univ. Press, 1991:63-76. structure, 23. Lucchesi PA, Sweadner KJ. Postnatal changes in Na,KATPase isoform expression in rat cardiac ventricle. J Biol Chem 1991;266:9327-31. 24. Jewell EA, Shamraj Ol, Lingrel JB. Isoforms of the alpha subunit of Na,K-ATPase and their significance. Acta Physiol Scand 1992;146:161-9. 25. Lytton JJ, Lin C, Guidotti G. Identification of two molecular forms of Na,K-ATPase in rat adipocytes. J Biol Chem 1985;260: 1177-84. 26. Shyjan AW, Gottardi C, Levenson R. The Na,K-ATPase 132 subunit is expressed in rat brain and copurifles with Na,K-ATPase activity. J Biol Chem 1990;265:5166-9. 27. Martin-Vasallo P, Dackowski W, Emanuel JR, Levenson R. Identification of a putative isoform of the Na,K-ATPase 13subunit. Primary structure and tissue-specific expression. J Biol Chem 1989;264:4613-8. 28. Akopyanz NS, Broude NE, Vinogradova NG, Balabanov YA, Monastyrskaya GS, Sverdlov ED. Differential expression of three Na,K-ATPase catalytic subunit isoforms in human tissues, organs, and cell lines. In: Kaplan JH, DeWeer P, eds. The sodium pump: recent developments. New York: Rockefeller Univ. Press, 1991: 189-93. 29. Peng JHF, Parker JC Jr, Tsai FY. Immunochemical demonstration of a3 isozyme of Na,K-ATPase in human brain. Neurosci Left 1991;130:37-40. 30. Martin-Vasallo P, Ghosh 5, Coca-Prados M. Expression of Na,K-ATPase alpha subunit isoforms in the human ciliary body and cultured ciliary epithelial cells. J Cell Physiol 1989;141:243- 52. 31. Shamraj 01, Melvin D, Lingrel JB. Expression of Na,KATPase isoforms in human heart. Biochem Biophy Res Commun 1991;179:1434-40. 32. Hundal HS, Marette A, Ramlal T, Lie Z, Klip A. Expression of 13subunit isoforms of the Na,K-ATPase is muscle type-specific. FEBS Lett 1993;328:253-8. 33. Gick GG, Hatala MA, Chon D, Ismail-Beigi F. Na,K-ATPase in several tissues of the rat: tissue-specific expression of subunit mRNAs and enzyme activity. J Membr Biol 1993;131:229-36. 34. Sweadner KJ. Overlapping and diverse distribution of Na-K ATPase isozymes in neurons and glia. Can J Physiol Pharmacol 1992;70:S255-9. 35. Zahler R, Brines M, Kashgarian M, Benz EJ Jr. The cardiac conduction system in the rat expresses the a2 and a3 isoforms of the Na’,K-ATPase. Proc Natl Acad Sci USA 1992;89:99-103. 36. Jewell EA, Lingrel JB. Chimeric rat Na,K-ATPase alIa3 isoforms. Analysis of the structural basis for differences in Na requirements in the al and a3 isoforms. Ann NY Acad Sci 1992;671:120-.33. 37. McDonough AA, Hensley CB, Azuma KK Differential regulation of sodium pump isoforms in heart. Sem Nephrol 1992;12: 49-55. 38. Coca-Prados M, Martin-Vasallo P, Hernando N, Ghosh S. Cellular distribution and differential expression of the Na,KATPase alpha isoform (al, a2, a3), 131,and 132/AMOG genes in the ocular ciliary epithelium. In: Kaplan JH, DeWeer P, eds. The sodium pump: recent developments. New York: Rockefeller Univ. Press, 1991:157-63. 39. Gloor S, Antonicek H, Sweadner KJ, Pagliusi 5, Rainer F, Moos M, Schachner M. The adhesion molecule on Glia (AMOG) is a homologue of the 13subunit of the Na,K-ATPase. J Cell Biol 1990;110:165-74. 40. Good PJ, Richter K, Dawid lB. A nervous system-specific isotype of the p subunit of Na,K-ATPase expressed during early development of Xenopus laevis. Proc Natl Acad Sci USA 1990;87: 9088-92. 41. Canfleld VA, Okamoto CT, Chow D, Dorfman J, Gros P, Forte JG, Levenson R. Cloning of the H,K-ATPase 13subunit. Tissuespecific expression, chromosomal assignment, and relationship to Na,K-ATPase p subunits. J Biol Chem 1990;265:19878-84. 42. Reuben MA, Laaater LS, Sachs G. Characterization of a 13 subunit of the gastric H/K-transporting ATPase. Proc Natl Acad Sd USA 1990;87:6767-71. 43. Shull GE. eDNA cloning of the 13-subunit of the rat gastric H,K-ATPase. J Biol Chem 1990;265:12123-6. 44. Orlowski J, Lingrel JB. Thyroid and glucocorticoid hormones regulate the expression of multiple Na,K-ATPase genes in cal- tured neonatal rat cardiac myocytes. J Biol Chem 1990;265:346270. 45. Horowitz B, Hensley CB, Quintero M, Azuma KK, Putnam D, McDonough AA. Differential regulation of Na,K-ATPase al, a2, and 13subunit mRNA and protein levels by thyroid hormone. J Biol Chem 1990;265:14305-14. 46. Gick GG, Ismail-Beigi F. Thyroid hormone induction of Na,K-ATPase and its mRNAs in a rat liver cell line. Am J Physiol 1990;258:C544-51. 47. Barlet-Bas C, Khadouri C, Marsy 5, Doucet A. Enhanced intracellular sodium concentration in kidney cells recruits a latent pool of Na-K-ATPase whose size is modulated by corticosteroids. J Biol Chem 1990;265:7799-803. 48. Blot-Chabaud M, Wanstok F, Bonvalet J, Farman N. Cell sodium-induced recruitment of Na-K-ATPase pumps in rabbit cortical collecting tubules is aldosterone-dependent. J Biol Chem 1990;265:11676-81. 49. Lingrel JB, Orlowski J, Price EM, Pathak BG. Regulation of the a-subunit genes of the Na,K-ATPase and determinants of cardiac glycoside sensitivity. In: Kaplan JH, DeWeer P, eds. The sodium pump: structure, mechanism, and regulation. New York: Rockefeller Univ. Press, 1991:1-16. 50. Shull MM, Pugh DG, Lingrel JB. MspI and PvuII polymorphisms in the Na,K-ATPase a subunit related to gene ATP1AL1. Nucleic Acids Res 1990;18:205. 51. Shull MM, Pugh DG, Lane LK, Lingrel JB. MspI and PvuII polymorphisms in the Na,K-ATPase p subunit gene ATP1B1. Nucleic Acids Res 1990;18:1087. 52. Ruegg UT. Ouabain-a link in the genesis of high blood pressure? [Review]. Experentia 1992;48:1102-6. 53. Blaustein MP. Physiological effects of endogenous ouabain: control of intracellular Ca2 stores and cell responsiveness [Review]. Am J Physiol 1993;264(Cell Physiol 33):C1367-87. 54. Hoffman BF, Bigger JT Jr. Cardiovascular drugs. Digitalis and allied cardiac glycosides. In: Gilman AG, Goodman LS, Gilman A, eds. The pharmacological basis of therapeutics. New York: Macmillan, 1980:729-60. 55. Aiystarkhova E, Gasparian M, Modyanov NN, Sweadner KJ. Na,K-ATPase extracellular surface probed with a monoclonal antibody that enhances ouabain binding. J Biol Chem 1992;267: 13694-701. 56. Schultheis PJ, Waffick ET, Lingrel JB. Kinetic analysis of ouabain binding to native and mutated forms of Na,K-ATPase and identification of a new region involved in cardiac glycoside interactions. J Biol Chem 1993;268:22686-94. 57. O’Brien WJ, Wallick ET, Lingrel JB. Amino acid residues of Na,K-ATPase involved in ouabain sensitivity do not bind the sugar moiety of cardiac glycosides. J Biol Chem 1993;268:7707-12. 58. Thomas R. Cardiac drugs. In: Wolff M, ed. Burgers medicinal chemistry. New York: Wiley, 1980:47-96. 59. Erdmann EE. Influence of cardiac glycosides on their receptor. In: Greeff K, ad. Cardiac glycosides. Part I: experimental pharmacology. Berlin: Springer-Verlag, 1981:337-68. 60. Akera T, Brody TM. The role of Na,K-ATPase in the inotropic action of digitalis [Review). Pharmacol Rev 1978;29: 187220. 61. Akera T, Larsen FS, Brody TM. The effect of ouabain on sodium- and potassium-activated adenosine triphosphatase from the hearts of several mammalian species. J Pharmacol Exp Ther 1969;170:17-26. 62. Somberg JC, Barry WH, Smith TW. Differing sensitivities of Purkinje fibers and myocardium to inhibition of monovalent cation transport by digitalis. J Clin Invest 198 1;67:116-23. 63. Smith JR, Gheorghiade M, Goldstein S. The current role of digoxin in the treatment of heart failure. Coronary Artery Dis 1993;4:16-26. 64. El-Maflakh RS, Barrett JL, Wyatt RJ. The Na,K-ATPase hypothesis for bipolar disorder: implications for normal development. J Child Adolesc Psychopharmacol 1993;3:37-52. 65. Krisanda TJ. Digitalis toxicity. Using immunotherapy when supportive care isn’t enough. Postgrad Med 1992;91:273-83. 66. Langer GA. The “sodium pump lag” revisited. J Mol Cell Cardiol 1983;15:647-51. 67. Akera T, Ng Y. Digitalis sensitivity of Na,K-ATPase, myocytes and the heart. Life Sci 1991;48:97-106. 68. Lieberman DM, Reithmeier RAF, Ling V, Charuk JHM, CLINICAL CHEMISTRY, Vol. 40, No. 9, 1994 1683 Goldberg H, Skorecki KL. Identification of P-glycoprotein in renal brush border membranes. Biochem Biophy Res Commun 1989; 162: 244-52. 69. deLannoy LAM, Silverman M. The MDR1 gene product, P-glycoprotein, mediates the transport of the cardiac glycoside, digoxin. Biochem Biophys Res Commun 1992;189:551-7. 70. Howanitz PJ, Steindel SJ. Digoxin therapeutic drug monitoring practices. Arch Pathol Lab Med 1993;117:684-90. 71. Valdes R Jr, Brown BA, Graves SW. Variable cross-reactivity of digoxin metabolites in digoxin immunoassays. Am Clin Pathol 1984;82:210-3. 72. Miller JJ, Straub RW, Valdes R Jr. A digoxin immunoassay in which cross-reactivity of digoxin metabolites is proportional to their biological activity. Clin Chem 1994;40:in press. 73. Soldin SJ. Digoxin-issues and controversies [Review]. Clin Chem 1986;32:5-12. 74. Hug E, Brown L, Erdmann E. The ouabain receptor in the myocardium and conduction system of the sheep heart. In: Erdmann E, Greeff K, Skou JC, ads. Cardiac glycosides 1785-1985. New York: Steinkopff Verlag Darmstadt, 1986:61-8. 75. Price EM, Lingrel JB. Structure-function relationships in the Na,K-ATPase a subunit: site-directed mutagenesis of glutamine111 to arginine and asparagine-122 to aspartic acid generates a ouabain-resistant 76. Sweadner Na,K-ATPases enzyme. Biochemistry 1988;27:8400-8. SK. Rat cardiac ventricle has two with different affinities for ouabain: develop- KJ, Farshi mental changes in immunologically different catalytic subunits. Proc Nati Acad Sci USA 1987;84:8404-7. 77. Urayama 0, Sweadner LI. Ouabain sensitivity of the a3 isozyme of rat Na,K-ATPase. Biochem Biophys Res Commun 1988;156:796-800. 78. Kolansky DM, Brines ML, Gilmore-Heber M, Benz EJ Jr. The A2 isoform of rat Na,K-adenosine triphosphatase is active and exhibits high ouabain affinity when expressed in transfected fibroblasts. FEBS Lett 1992;303:147-53. 79. Charlemagne D, Maccent JM, Preteseille M, Lelievre LG. Ouabain binding sites and (Na,K)-ATPase activity in rat cardiac hypertrophy. J Biol Chem 1986;261:185-9. 80. Schmidt TA, Allen PD, Colucci WS, Marsh JD, Kjeldsen K. No adaptation to digitalization as evaluated by digitalis receptor (Na,K-ATPase) quantification in explanted hearts from donors without heart disease and from digitalized recipients with endstage heart failure. Cardiovasc Pharmacol 1993;71:110-4. 81. Bluschke V, Bonn R, Greeff K. Increase in the (Na + K)-ATPase activity in heart muscle after chronic treatment with digitoxin or potassium-deficient diet. Eur J Pharmacol 1976;37: 189-91. 82. Canessa CM, Horisberger JD, Louvard D, Rossier BC. Mutation of a cysteine in the first transmembrane segment of Na,KATPase a subunit confers ouabain resistance. EMBO J 1992;11: 1681-7. 83. Beach HR Jr, Hufferd S, Lake M, Hurwitz R, Watanabe AM. False elevation of apparent digoxin levels in plasma of premature infants [Abstract]. Chin Chem 1976;22:1168. 84. Valdes R Jr. Endogenous digoxin-like immunoreactive factors: impact on digoxin measurements and potential physiological implications [Review]. Clin Chem 1985;31:1525-32. 85. Craver JL, Valdes R Jr. Anomalous serum digoxin concentrations in uremia. Ann Intern Med 1983;98:483-4. 86. Gruber KA, Whitaker JM, Buckalew VM Jr. Endogenous digitalis-like substance in plasma of volume-expanded dogs. Nature 1980;287:743-5. 87. Valdes R Jr, Graves SW. Protein binding of endogenous digoxin-immunoactive factors in human serum and its variation with clinical condition. J Clin Endocrinol Metal, 1985;60:1135-43. 88. Valdes R Jr, Graves SW, Brown BA, Landt ML. Endogenous substance in newborn infants causing false-positive digoxin measurements. J Pediatr 1983;102:947-50. 89. Seccombe DW, Pudek MR, Humphries KH, Matthewson B, Taylor GP, Jacobson BE, Whitfield MP. A study into the nature and organ source of digoxin-like immunoreactive substance(s) in the perinatal period. Biol Neonate 1989;56:136-46. 90. Graves SW, Valdes R Jr, Brown B, Knight AB, Craig HR. Endogenous digoxin-immunoreactive substance in human preg- nancies. J Clin Endocrinol Metab 1984;58:748-51. 91. Graves SW, Brown B, Valdes R Jr. An endogenous 1684 CLINICAL CHEMISTRY, Vol. 40, No. 9, 1994 digoxin- like substance in patients with renal impairment. Ann Intern Med 1983;99:604-8. 92. Nanji AA, Greenway DC. Correlation between serum albumin and digoxin-like immunoreactive substance in liver disease. J Clin Pharmacol 1986;26: 152-3. 93. Sewell RB, Poston L, Wilkinson SP, Williams R. A circulating inhibitor of leucocyte sodium transport in patients with advanced liver cirrhosis. Clin Sci 1984;66:741-4. 94. Shaikh IM, Lau BWC, Siegfried BA, Valdes R Jr. Isolation of digoxin-like immunoreactive factors from mammalian adrenal cortex. J Biol Chem 1991;266:13672-8. 95. Lichtein D, Samuelov S, Gati I, Wechter WJ. Digitalis-like compounds in animal tissues. J Basic Chin Physiol Pharmacol 1992;3:269-92. 96. Haddy F, Pamnani M, Clough D. The sodium-potassium pump in volume-expanded hypertension. Clin Exp Hypertens 1978-79;1:295-336. 97. Hamlyn JM, Manunta P. Ouabain, digitalis-like factors and hypertension [Review]. J Hyperten 1992;10:S99-111. 98. Mathews WR, DuCharme DW, Hamlyn JM, Harris DW, Mandel F, Clarke MA, Ludens JH. Mass spectral characterization of an endogenous digitalislike factor from human plasma. Hypertension 1991;17:930-5. 99. Doris PA. Regulation of Na,K-ATPase by endogenous ouabain-like material. Proc Soc Exp Biol Med 1994;205:202-12. 100. Kelly RA, Smith TW. Is ouabain the endogenous digitalis? [Editorial]. Circulation 1992;86:694-7. 101. deWardener HE, MacGregor GA. The natriuretic hormone and essential hypertension. Lancet 1982;i:1450-4. 102. Pamnani M, Huot 5, Buggy J, Clough D, Haddy F. Demonstration of a humoral inhibitor of the Na-K pump in some models of experimental hypertension. Hypertension 1981;3(Suppl II):II-96-101. 103. Haddy FJ. Endogenous digitalis-like factor or factors [Editorial]. N Engl J Med 1987;316:621-3. 104. Ghione S, Baizan S, Montali U, Raggi A, Clerico A, Gazzetti P, Donato L. Plasma digoxin-like immunoreactivity in essential hypertension: relation to plasma renin activity. J Hypertens 1983; 1(Suppl 2):142-4. 105. Gruber KA, Rudel LL, Bullock BC, Increased circulating levels of an endogenous digoxin-like factor in hypertensive monkeys. Hypertension 1982;4:348-54. 106. Plunkett WC, Gruber KA, Hutchins PM, Buckalew VM. Vascular reactivity is increased by factors in plasma of volumeexpanded dogs [Abstract]. Clin Res 1980;28:827A. 107. Chakravarty B, Mills IH, Callingham BA. Effects of natriuretic fractions of human urine on isolated anococcygeus muscle of the rat. Renal Physiol 1984;7:205-17. 108. Hulthen UL, Boffi P, Kiowski W, Buhler FR. Forearm vasoconstrictor response to ouabain: studies in patients with mild and moderate essential hypertension. J Cardiovasc Pharmacol 1984;6:S75-81. 109. DeMots H, Rahimtoola SH, McAnulty J}l, Porter GA. Effects of ouabain on coronary and systemic vascular resistance and myocardial oxygen consumption in patients without heart failure. Am J Cardiol 1978;41:88-93. 110. Williams MH Jr, Zohman LR, Ratner AC. Hemodynamic effects of cardiac glycosides on normal human subject during rest and exercise. J Appl Physiol 1958;13:417-21. 111. Kojima I, Yoshiha 5, Ogata F. Involvement of endogenous digitalis-like substance in genesis of deoxycorticosterone-salt hypertension. Life Sci 1982;30:1775-81. 112. Shilo L, Pomeranz AP, Rathaus M, Bernheim J, Shenkman L. Endogenous digoxin-hike factor raises blood pressure and protects against digitalis toxicity. Life Sci 1989;44:1867-70. 113. Indolfi C, Piscione F, Russolillo E, Villari B, Golino P, Ambrosini V, et al. Digoxin-induced vasoconstriction of normal and atherosclerotic epicardial coronary arteries. Am J Cardiol 1991;68:1274-8. 114. Bova 5, Blaustein MP, Ludens JH, Harris DW, DuCharme DW, Hamlyn JM. Effects of an endogenous ouabain-like compound on heart and aorta. Hypertension 199 1;17:944-50. 115. Bagrov AY, Fedorova OV, Maslova MN, Roukoyatkina NI, Ukhanova MV, Zhabko EP. Endogenous plasma Na,K-ATPase inhibitory activity and digoxin-like immunoreactivity after acute myocardial infarction. Cardiovase Res 1991;25:371-7. 116. 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;86:420-5. 117 Haddy FJ, Overbeck 11W. The role of humoral agents in volume expanded hypertension [Review]. Life Sci 1976;19:935-48. 118. Sowers JR, Khoury S. Diabetes and hypertension [Review]. Primary Care 1991;18:509-24. 119. Clerico A, Giampietro 0. Is the endogenous digitalis-like factor the link between hypertension and metabolic disorders as diabetes mellitus, obesity and acromegaly? [Review]. Clin Physiol Biochem 1990;8:153-68. 120 Delva P, Capra C, Degan M, Minuz P, Coy G, Milan L, et al. High plasma levels of ouabain-like factor in normal pregnancy and in pre-eclampsia. Eur J Clin Invest 1989;19:95-100. 121. Gusdon JP Jr, Buckalew VM Jr, Hennessey JF. A digoxinlike immunoreactive substance in preeclampsia. Am J Obstet Gynecol 1984;150:83-5. 122. Valdes R Jr, Graves SW, Knight AB, Craig HR. Endogenous digoxin immunoactivity is elevated in hypertensive pregnancy. Prog Clin Biol Res 1988;192:229-32. 123. Argento NB, Hamilton BP, Valente WA, Hamlyn JM. Increased circulating levels of a ouabain-like compound in hypothyroid hypertension [Abstract]. Hypertension 199 1;18:425. 124. Izumo H, Izumo S, Deluise M, Flier JS. Erythrocyte Na,K pump in uremia. Acute correction of a transport defect by hemodialysis. J Chin Invest 1984;74:581-8. 125. Deray G, Rieu M, Devynck MA, Pernollet MG, Chanson P, Luton JP, Meyer P. Evidence of an endogenous digitalis-like factor in the plasma of patients with acromegaly. N Engl J Med 1987; 316:575-80. 126. Masugi F, Ogthara T, Hasegawa A, Tomii M, Nagano K, Higashimori K, et al. Circulating factor with ouabain-like immunoreactivity in patients with primary aldosteronism. Biochem Biophys Res Cominun 1986;135:41-5. 127. Weidmann P, Ferrari P. Central role of sodium in hypertension in diabetic subjects. Diabetes Care 1991;14:220-32. 128. Chen 5, Yuan C, Clough D, Schooley J, Haddy F, Pamnani MB. Role of digitalis-like substance in the hypertension of streptozotocin-induced diabetes in reduced renal mass rats. Am J Hypertens 1993;6:397-406. 129. Poehlman ET. Regulation of energy expenditure in aging humans. Geriatric Biosci 1993;41:552-9. 130. Biver P, Clerico A, Path A, Balzan 5, Boldrini A, Cipollom C. Endogenous digitalis-like factors: their possible pathophysiological implications with particular regard to the perinatal period [Review]. Child Nephrol Urol 1990;10:164-.80. 131. Stefano JL, Norman ME, Morales MC, Gopherud JM, Mishra OP, Delivoria-Papadopoulos M. Decreased erythrocyte Na,KATPase activity associated with cellular potassium loss in extremely low birth weight infants with nonoliguric hyperkalemia. J Pediatr 1993;122:276-84. 132 Seccombe DW, Pudek MR, Whitfield MF, Jacobson BE, Wittmann BK, King JF. Perinatal changes in a digoxin-like immunoreactive substance. Pediatr Res 1984;18:1097-9. 133. Christo PJ, El-Mallakh RS. Possible role of endogenous ouabain-like compounds in the pathophysiology of bipolar illness. Med Hypotheses 1993;41:378-83. 134. Hank SI, Mitchell MJ, Kalaria RN. Ouabain binding in the human brain. Arch Neurol 1989;46:951-4. 135. Liguri G, Taddei N, Latorruca S, Nediani C, Sorbi S. Changes in Na,K-ATPase, Ca2-ATPase and some soluble enzymes related to energy metabolism in brains of patients with Alzheimer’s disease. Neurosci Lett 1990;112:338-42. 136. Markesbery WR. Alzheimer’s disease-a mini review. J Ky Med Assoc 1989;87:333-5. 137. Smith H, Janz TG, Erker M. Digoxin toxicity presenting as altered mental status in a patient with severe chronic obstructive lung disease. Heart Lung 1992;21:78-80. 138. Varsano 5, Shilo L, Bruderman I, Dolev S, Shenkman L. Endogenous digoxin-like immunoreactive factor is elevated in advanced chronic respiratory failure. Chest 1992;1O1:146-9. 139. Clough DL, Paxnnani MB, Haddy FJ. Decreased myocardial Na’-K-ATPase activity in one-kidney, one-clip hypertensive rats. Am J Physiol 1983;245:H244-51. 140. Herrera VLM, Chobanian AV, Ruiz-Opazo N. Isoform-specific modulation of Na,K-ATPase a-subunit gene expression in hypertension. Science 1988;241:221-3. 141. Graves SW, Williams GH. Endogenous digitalis-like natriuretic factors. Ann Rev Med 1987;38:433-44. 142. Valdes R Jr, Hagberg J, Vaughan TE, Lau BWC, Seals DR, Ehsani AA. Endogenous digoxin-like immunoreactivity in blood is increased during prolonged strenuous exercise. Life Sd 1988;42: 103-10. 143. Weinberg U, Dolev S, Werber MM, Shapiro MS, Shilo L, Shenkman L. Identification and preliminary characterization of two human digitalis-like substances that are structurally related to digoxin and ouabain. Biochem Biophys Res Commun 1992;188: 1024-9. 144. Yuan CM, Manuals P, Hamlyn JM, Chen S, Bohen E, Yeun J, et al. Long-term ouabain administration produces hypertension in rats. Hypertension 1993;22:178-87. 145. Sekihara H, Yazaki Y, Kojima T. Ouabain as an amplifier of mineralocorticoid-induced hypertension. Endocrinology 1992;131: 3077-82. 146. Shimoni Y, Gotsman M, Deutsch J, Kachalsky S, Lichtstein D. Endogenous ouabain-like compound increases heart muscle contractility. Nature 1984;307:369-71. 147. Devynck MA, Pernollet MG, De The H, Rosenfeld JB, Meyer P. Endogenous digitalis-like compound in essential hypertension. In: Davison AM, Guillou PJ, eds. Proc European Dialysis and Transplant Association-European Renal Association. London: Pitman Publ., 1983:489-92. 148. Tirupattur PR, Ram JL, Standley PR, Sowers JR. Regulation of Na,K-ATPase gene expression by insulin in vascular smooth muscle cells. Am J Hypertens 1993;6:626-9. 149. Pontzer NJ, Chandler U, Stevens BR, Crews VF. Receptors, phosphoinositol hydrolysis and plasticity of nerve cells. In: Coleman P, Higgins G, Phelps C, eds. Progress in brain research. New York: Elsevier Science Publ., 1990:221-5. CLINICAL CHEMISTRY, Vol. 40, No. 9, 1994 1685
© Copyright 2026 Paperzz