12
1674 CLINICAL CHEMISTRY, Vol. 40, No. 9, 1994 CUN. CHEM. 40/9, 1674-1685 (1994) Understanding the Sodium Pump and Its Relevance to Disease Andrea M. Rose’ and Roland Valdes, Jr.” I 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 indi- vidual 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 glyco- sides. However, endogenous ligands structurally similar to digoxin or ouabain may control the activity of this important molecular complex. Here we review the struc- ture 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 sus- pected endogenous regulators. Also reviewed are vari- ous disorders, including cardiovascular, neurological, renal, and metabolic diseases, purported to involve dys- function of Na,K-ATPase activity. The escalation in knowledge at the molecular level concerning sodium pump function foreshadows application of this knowl- edge 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 triphos- phatase (Na,K-ATPase; sodium pump; EC 3.6.1.37) is a plasma membrane-associated protein complex that is ex- pressed 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. Regula- Departments of Pathology and 2Biochemistry, University of Louisville School of Medicine, Louisville, KY 40292. ‘Mdress correspondence to this author at: Department of Pa- thology, University of Louisville, Louisville, KY 40292. Fax 502- 852-1771; E-mail [email protected]. 4Nonstandard abbreviations: Na,K-ATPase, Na,K-activated adenosine triphosphatase; DLIF, digoxin-like immunoreactive fac- tors; EDLF, endogenous digoxin-like factors; OLF, ouabain-like factors; and AD, Alzheimer disease. 5Although technically an enzyme, Na,K-ATPase functions pri- marily as an ion transporter. Therefore, most investigators refer to the alpha and beta gene products as isoforms instead of isoen- zymes. Received December 27, 1993; accepted June 16, 1994. tion of this enzyme (transporter) and its individual iso- forms 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 endoge- nous ligands structurally similar to cardiac glycosides may act as natural regulators of the sodium pump in heart and other tissues. Identification of naturally cc- curring 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 re- view recent information related to structure and func- tion, 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-includ- ing 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 in- tracellular hydrolysis of ATP to the export of three in- tracellular Na ions and the import of two extracellular K ions. The continuous operation of this macromolec- War machine ensures the generation and maintenance of concentration gradients of Na and K across the cell membrane. This electrochemical gradient provides en- ergy for the membrane transport of metabolites and nutrients, e.g., glucose and amino acids, and such ions as protons, calcium, chloride, and phosphate. The elec- trochemical gradient is essential also for regulation of cell volume and for the action potential of muscle and nerve. The relative intra- and extracellular concentra- tions 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-span- ning 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

UnderstandingtheSodiumPumpandItsRelevancetoDisease · Thesodium-potassium-activated adenosinetriphos-phatase (Na,K-ATPase; sodium pump; EC3.6.1.37)isa plasmamembrane-associated proteincomplex

  • Upload
    others

  • View
    12

  • Download
    0

Embed Size (px)

Citation preview

Page 1: UnderstandingtheSodiumPumpandItsRelevancetoDisease · Thesodium-potassium-activated adenosinetriphos-phatase (Na,K-ATPase; sodium pump; EC3.6.1.37)isa plasmamembrane-associated proteincomplex

1674 CLINICALCHEMISTRY, Vol. 40, No. 9, 1994

CUN. CHEM. 40/9, 1674-1685 (1994)

Understanding the Sodium Pump and Its Relevance to DiseaseAndrea M. Rose’ and Roland Valdes, Jr.”

I

Na,K-ATPase (sodium pump; EC 3.6.1.37) is present inthe membrane of most eukaryotic cells and controlsdirectly or indirectly many essential cellular functions.Regulation of this enzyme (ion transporter) and its indi-vidual isoforms is believed to play a key role in theetiology of some pathological processes. The sodiumpump is the only known receptor for the cardiac glyco-sides. However, endogenous ligands structurally similarto digoxin or ouabain may control the activity of thisimportant molecular complex. Here we review the struc-ture and function of Na,K-ATPase, its expression anddistribution in tissues, and its interaction with knownligands such as the cardiac glycosides and other sus-pected endogenous regulators. Also reviewed are vari-ous disorders, including cardiovascular, neurological,renal, and metabolic diseases, purported to involve dys-function of Na,K-ATPase activity. The escalation inknowledge at the molecular level concerning sodiumpump function foreshadows application of this knowl-edge in the clinical laboratory to identify individuals atrisk for Na,K-ATPase-associated diseases.

IndexIng Terms: Na,K-ATPase/isoforms/cardiac glycosides/ age-related effects/digoxin/ouabain/hypertenslon/diabetes/Alzheimerdisease/neurological disorders

The sodium-potassium-activated adenosine triphos-phatase (Na,K-ATPase; sodium pump; EC 3.6.1.37) is aplasma membrane-associated protein complex that is ex-pressed in most eukaryotic cells.4’5 The “pump” couplesthe energy released in the intracellular hydrolysis ofadenosine tnphosphate (ATP) to the transport of cellularions, a major pathway for the controlled translocation ofsodium and potassium ions across the cell membrane.Na,K-ATPase therefore controls directly or indirectlymany essential cellular functions, e.g., cell volume, freecalcium concentration, and membrane potential. Regula-

Departments of ‘ Pathology and 2Biochemistry, University ofLouisville School of Medicine, Louisville, KY 40292.

‘Mdress correspondence to this author at: Department of Pa-thology, University of Louisville, Louisville, KY 40292. Fax 502-852-1771; E-mail [email protected].

4Nonstandard abbreviations: Na,K-ATPase, Na,K-activatedadenosine triphosphatase; DLIF, digoxin-like immunoreactive fac-tors; EDLF, endogenous digoxin-like factors; OLF, ouabain-likefactors; and AD, Alzheimer disease.

5Although technically an enzyme, Na,K-ATPase functions pri-marily as an ion transporter. Therefore, most investigators refer tothe alpha and beta gene products as isoforms instead of isoen-zymes.

Received December 27, 1993; accepted June 16, 1994.

tion of this enzyme (transporter) and its individual iso-forms is thought to play a key role in the etiology of somepathological processes.

The sodium pump is the only known receptor for thecardiac glycosidesused to treat congestive heart failureand cardiac arrhythmias. This suggests that endoge-nous ligands structurally similar to cardiac glycosidesmay act as natural regulators of the sodium pump inheart and other tissues. Identification of naturally cc-

curring regulators of Na,K-ATPase could initiate thediscovery of new hormone-like control systems involvedin the etiology of selected disease processes, hence theimportance of understanding the relation of the sodiumpump and its ligands to disease. In this article, we re-view recent information related to structure and func-tion, genetic expression and distribution in tissues, andinteraction of the sodium pump with ligands such as thecardiac glycosides and other suspected endogenouscounterparts. Several diverse disease processes-includ-ing cardiovascular, renal, neurological, and metabolic

disorders, having in common a dysfunction in salt and

water homeostasis-are emphasized, as is the clinicalneed for understanding the function and control of thisubiquitous ion transporter system.

Structure and FunctIon of Na,K-ATPaseNa,K-ATPase couples the energy released in the in-

tracellular hydrolysis of ATP to the export of three in-tracellular Na ions and the import of two extracellularK ions. The continuous operation of this macromolec-War machine ensures the generation and maintenanceof concentration gradients of Na and K across the cellmembrane. This electrochemical gradient provides en-ergy for the membrane transport of metabolites andnutrients, e.g., glucose and amino acids, and such ionsas protons, calcium, chloride, and phosphate. The elec-trochemical gradient is essential also for regulation ofcell volume and for the action potential of muscle andnerve. The relative intra- and extracellular concentra-tions of Na and K ions maintained primarily by thesodium pump and the cofactors required for activity areshown in Fig. 1 (1, 2).

The functional macromolecule is a membrane-span-

ning 270-kDa tetramer consisting of two dimers, each

composedof noncovalently interacting alpha (112 kDa)and beta (55 kDa) subunits. A model of the transportercomplex relative to its membrane location is shown inFig. 2. The presence of a smaller gamma subunit (10

Page 2: UnderstandingtheSodiumPumpandItsRelevancetoDisease · Thesodium-potassium-activated adenosinetriphos-phatase (Na,K-ATPase; sodium pump; EC3.6.1.37)isa plasmamembrane-associated proteincomplex

(150 mmol/L)

Digitalis

(5 mmol/L)

OUT

Fig. 1. SchematIc diagram of the Na,K-ATPase-associated cofactorsand Ions.Modifiedfrom Akera (2).

Extracellular

IN

Intracellular

a a

IN

Fig.2. Schematic diagram of the plasma membrane-spanning Na,K-ATPase transporter complex, indicating the positions of the alpha,beta, and putative gamma subunits.

I AlP

E1 . E2-K2

2K

A

CLINICALCHEMISTRY, Vol. 40, No. 9, 1994 1675

kDa) has been suggested; however, its role, if any, hasnot been defined (3).

The alpha (catalytic) subunit, is proposed to have 7(4)or 8(5) transmembrane domains; however, the numbercan vary from 6 to 10, based on interpretation of hydrop-athy profile data (6) and identification of specific li-gand-receptor interactions that predict alpha chain to-pology (7-9). The alpha subunit contains all the bindingsites for ligands known to stimulate or inhibit the en-zyme (10-13). Tentative models representing mem-brane-spanning segments of the alpha subunit are de-tailed in Fig. 3.

The beta subunit has a single hydrophobic transmem-brane domain and is highly glycosylated on its noncy-tosolic surface (14). Hiatt et al. postulate that the betasubunit may serve to orient and stabilize the alphasubunit in the membrane (15). Cellular expression ofthe beta subunits and assembly with the alpha subunits

are necessary for correct conformation and activity ofthe Na,K-ATPase holoenzyme (16). Although the role ofthe beta subunit in ion translocation is uncertain, itspresence appears essential for function of the sodiumpump (16).

Phosphorylation is an important step in the function

of Na,K-ATPase. The molecule undergoes an alpha-he-lix to beta-sheet transition between two principal reac-tive states, E1 and E2, in the multistep reaction bywhich Na ions and K ions traverse the membrane(17, 18). The conformational transition results via a

‘)63K327 Y1016

1263 0737 Y1018R166

Fig. 3. Foldingmodels for the Na,K-ATPase alpha transmembranesegments: (A) 10 membrane-spanning segments; (B) 8 membrane-spanning segments.Letters refer to the one-letter amino acid code. Numbers represent the topo-logical location of particular amino adds. From Kailish et at. (7); used withpermission.

cyclic reaction in which the enzyme is phosphorylatedby ATP in the presence of Mg2 ions and Na ions andthen dephosphorylated in the presence of K ions. Amodel depicting this thermodynamic cycle is shown inFig. 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 independentgenes (20-22). The sequence conservation among differ-

3Na2l(

E2-P.Na3 .p r E2P.K2

Nat. Mi2 ______________________Ei-P.Naa1,...

ATP

Fig.4. Principal reactive states (E1 and E) involved in the transportof sodium (Na) and potassium (K) ions across the cytoplasmicmembrane.The enzyme (E) Isphosphorylated(P), with ATP as thephosphatedonor andMg2 as a cofactor In the reaction. From MacGregor and Walker (19), asmodified from Sen et at. (18); used with permission.

Page 3: UnderstandingtheSodiumPumpandItsRelevancetoDisease · Thesodium-potassium-activated adenosinetriphos-phatase (Na,K-ATPase; sodium pump; EC3.6.1.37)isa plasmamembrane-associated proteincomplex

EyeHeart (ventricle)

KidneyLungSkeletal muscle

ThyroidUterus

HumanRatRatRatHumanHumanHuman

al, a2, a3a

al, a3 neonate#{176}

al, a2 adult#{176}al, a3 aged#{176}alaalaal,

Rat a1,a2,l,i32aHuman al, a2Human ala

Superscripts referto all thepreceding isoforms onthe same line: a isoform-specific expression of mRNA identified byNorthernanalysis;b isoform-specificexpressionof proteinidentifiedbyWesternanalysis.

1676 CLINICAL CHEMISTRY, Vol. 40, No. 9, 1994

ent species suggests that individual roles for the iso-forms, though not yet determined, arose early and weremaintained throughout evolution.

The three alpha isoforms (alpha 1, 2, and 3) are ex-pressed in a developmental and tissue-specific manner.Utilizing monoclonal antibodies that recognize the inch-vidual alpha isoforms, Lucchesi and Sweadner haveshown (23) that rat ventricular muscle-membrane prep-arations express alpha 1 in all stages of development;alpha 3 is present at birth through days 14 to 21, and isthen replaced by alpha 2 in the adult rat; alpha 3 againpredominates in aged rats. The physiological signifi-cance of this shift in subunit isoform expression is un-known. Tissue specificity for the different isoforms hasbeen identified both at the mRNA and protein level forvarious species (22, 24), and is summarized for isoformsfrom rat and human tissues in Table 1. Table 1, al-though not comprehensive, serves to illustrate the di-versity of isoform distribution in tissues. The vast ma-jority of studies defining isoform distribution have beendone with animals, less data being available for humantissues. Nevertheless, studies in both humans and ratssuggest that alpha 1 is the only isoform expressed ap-preciably in the kidney (33), whereas alpha 3 is associ-ated primarily with the nervous system (22, 28, 29).Isoform specificity even extends to cell type within aparticular tissue, as evidenced by studies with tissuefrom brain (34), heart (35), and eye (30), supporting thehypothesis that the isoforms have different physiologi-cal functions (36, 37).

Three Na,K-ATPase beta isoforms and one relatedH,K-ATPase (another member of the cation-transport-ing ATPases) beta isoform have been identified. Beta 1has been isolated from several vertebrate species in awide range of tissues. Beta 2 is expressed largely inbrain (26,27) and ocular ciliary epithelia (38); however,recent studies suggest that beta 2 is also expressed inglycolytic fast-twitch muscles of the rat hindlimb (32).

Table 1. Tissue dIstribution of Na,K-ATPase Isoforms.Tissu. source Species Subunit

Adipose Rat al, a2’

Brain Rat f32#{176}Human al a2, a

Human a3b

Human al, a2, a3ab

Human al, a2, a3, a

Interestingly, the beta 2 isoform appears to serve a dualfunction, both as a subunit of the Na,K-ATPase and asa mediator of neuron-astrocyte adhesion (39). This sug-gests the possibility that these proteins, as a family,may play other roles besides their traditionally definedtransport function. Finally, a protein referred to as beta3 has been isolated from Xenopus (40), and the beta-isoform of the H,K-ATPase has been characterized fromseveral vertebrate species (41-43). The beta isoformsare less similar to each other in amino acid sequencethan are their alpha isoform counterparts to each other;the beta isoforms also vary in their number of aspar-agine-linked glycosylation sites (27).

The expression of all the alpha isoforms and beta 1 aredifferentially regulated by hormones (44). Horowitz etal. (45) determined that thyroid status affected the al-pha 1, alpha 2, and beta isoform-specific expression ofmRNA and protein in rat heart, skeletal muscle, andkidney; and Gick and Ismail-Beigi (46) found that incu-bation of a rat liver cell line with thyroid hormoneresulted in an increase in alpha 1 and beta mRNAexpression and Na,K-ATPase activity. Increased so-dium concentration in response to corticosteroids is re-ported to recruit an intracellular latent pool of Na,K-ATPase 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 sequencesof the human alpha isoform genes, found that each con-tains a number of potential transacting and hormone-binding sites that do not appear to be conserved amongthe three alpha isoform genes, thus allowing for differ-ential regulation.

The ability to detect nucleotide changes that result inrestriction fragment length polymorphisms has led tothe discovery of sequence variation in some families inthe human alpha (50) and beta (51) subunit genes.Whether these genotypic differences, or others not yetidentified, correlate with a particular pathology is atpresent undetermined.

Regulators of Sodium Pump ActivityReferences Cardiac Glycosides

25 The Na,K-ATPase alpha subunit is the only known26, 27 receptor for the cardiac glycosides, which underscores

29 the clinical significance of the pump in treatment of30 congestive heart failure and arrhythmias. MacGregor

31 and Walker have written a short review of the cardiac28 glycosides (19). These inhibitors of the sodium pump are23 derived from extracts of the plant genera Digitalis, Stro-

23 phanthus, and Acocanthera. Digoxin and digitoxin are23 products of species of the foxglove plant, Digitalis, and28 ouabain is obtained from the East African Ouabaio tree28 or seeds of the plant Strophanthus gratus (52,53). These28 compounds are the most potent inotropic agents known,32 and their cardiac effects are believed to be mediated28 through their ability to inhibit the sodium pump. His-28 torically, preparations of these substances have been

used therapeutically for perhaps 3000 years, includinguse of plant extracts containing cardiac glycosides bythe ancient Egyptians (54).

Page 4: UnderstandingtheSodiumPumpandItsRelevancetoDisease · Thesodium-potassium-activated adenosinetriphos-phatase (Na,K-ATPase; sodium pump; EC3.6.1.37)isa plasmamembrane-associated proteincomplex

Digoxin, which can be administered orally and isreadily absorbed by the gastrointestinal tract, is themost widely used cardiac glycoside clinically; ouabain isthe most widely used experimentally. These compounds,all cardenolides, have a sterol skeleton. Cardenolidesare C steroids having one or more sugar residues atC-3 and a flve-membered lactone ring at C-17 (see Fig.5). As previously described, cardiac glycoside binding toNa,K-ATPase occurs on the extracellular face of theintegral membrane protein. However, recent data sug-gest that a hydrophobic binding pocket that containsmembrane-spanning amino acids may be involved aswell (55). Alpha subunit amino acids important for car-diac glycoside binding have been determined (6, 56),and recent studies with site-directed mutagenesis tomake amino acid substitutions at proposed binding siteshave helped determine which regions recognize cardiacglycoside sugars (57). A first-order-approximation bind-ing model of the interaction of digoxin with Na,K-

ATPase has been proposed by Thomas (58) and involvesa folding of the receptor-binding epitopes around theligand. Fig. 6 depicts the intermolecular forces thatmight play a role in the interaction between the lactonering, sterol section, and sugar residues of digoxin. Re-gardless 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 ac-tivity of Na,K-ATPase.

Seminal work demonstrating the inhibitory effect ofcardiac glycosides on Na.,K-ATPase has been performedby Akera and Brody (60). Akera et al. (61) were the firstto compare the in vivo sensitivity of the sodium pump toouabain in dog, sheep, guinea pig, and rat with the invitro sensitivity of Na,K-ATPase in cardiac microsomalfractions from these same species. Inhibition of the so-dium pump by cardiac glycosides increases the strength

OH

OH RHAMNOSE:

RHAMNOSE

OH OH

H

DIGrTOXOSE

DIGITOXOSE: HDIGITOXOSE

DIGITOXOSE NO OH

OH

Fig. 5. Structures of the cardiacglycosides,digoxinand ouabain.Three digitoxosesugars (Indigoxin)and one rhamnose sugar (inouabain) areattached at the C-3 position of the steroid backbone.

AECP7(:

SITED

H-BINDING SITES

11

Fig. 6. A model for drug (ouabain, digoxin) interactionwith thereceptor (Na,K-ATPase).FromThomas (58); used with permission.

of contraction (inotropic effect) and slows the beating(chronotropic effect) of the heart. Membrane excitationof cardiac myocytes is characterized by opening of theNa channel and depolarization of the sarcolemmalmembrane in response to increased intracellular so-dium. Consequently, Ca2 channels open and the Ca2ion influx triggers the release of Ca2 stores from thesarcoplasmic reticulum into the cytosol. The increase inintracellular free Ca2 activates contractile proteins,resulting in myocardial contraction. Cardiac glycosidesinhibit the exchange of Na and K via Na,K-ATPase;the result is a relative transient increase in intracellu-lar sodium. The Na/Ca-ion exchanger, present in thesarcolemmal membrane, mediates the exchange of Naions for Ca2 ions. This exchange mechanism probablyresults in a relative increase in the concentration ofintracellular Ca2. The increased intracellular Ca2 istaken up into the sarcoplasmic reticulum via a Ca2pump. After depolarization, the extra Ca2 released re-sults in enhanced contractile force (1). Somberg et al.(62) determined that a 25% reduction in pump activitywas associated with a 20% increase in contractilestrength.

The cardenolides that specifically interact with thesodium pump also have well-documented effects onother cardiovascular organs such as the peripheral vas-cular tissue (see below). Even though digoxin is widelyused in the treatment of heart disease, the therapeuticindex is low, and Smith et al. (63) cite digoxin intoxica-tion as the most widely encountered adverse drug reac-tion in clinical practice. Symptoms of digoxin toxicitycommonly involve the gastrointestinal tract and thecentral nervous system and include: anorexia, nausea,vomiting, diarrhea, headache, delirium, cardiac rhythmdisturbances, manic syndrome, and depressive syn-drome (64, 65). One explanation is due to what Langerterms the “sodium pump lag” effect (66) explained above

CUNICAL CHEMISTRY, Vol. 40, No. 9, 1994 1677

Page 5: UnderstandingtheSodiumPumpandItsRelevancetoDisease · Thesodium-potassium-activated adenosinetriphos-phatase (Na,K-ATPase; sodium pump; EC3.6.1.37)isa plasmamembrane-associated proteincomplex

1678 CLINICALCHEMISTRY, Vol. 40, No. 9, 1994

and described in the flow diagram in Fig. 7. The desir-able outcome to the proposed sequence of events de-scribed in Fig. 7 is positive inotropy, but some individ-uals may experience symptoms of cardiac glycosidetoxicity as a result of abnormally high intracellularconcentrations of calcium.

Low tolerance to cardiac glycosides has been associ-ated with old age, acute myocardial infarction/ischemia,hypoxemia, magnesium depletion, renal insufficiency,hypercalcemia, carotid sinus massage, electrical cardi-oconversion, hypothyroidism, and hypokalemia (67). In-teraction with coadministered drugs such as quinidine,verapamil, and cyclosporine is a frequent cause of toxicaccumulation of diguxin. Recent evidence suggests thatthese drugs inhibit renal excretion of digoxin by inhib-iting the MDR1 gene product, P-glycoprotein, shown tobe present on the apical membrane of mammalian kid-ney (68). P-glycoprotein is overexpressed in multidrug-resistant cells and functions as a drug-efflux pump, rec-ognizing a variety of therapeutic agents, includingvinblastine, a known P-glycoprotein substrate. Signifi-cant accumulation of digoxin or vinblastine has beenreported in both a multidrug-resistant chinese hamsterovary cell line and the drug-sensitive parent cell linewhen cyclosporine, verapamil, or quinidine was addedto the culture medium (69). This suggests digoxin ex-cretion is also mediated by P-glycoprotein.

Understanding the pharmacokinetics of the cardiacglycosides in relation to renal excretion, age-related tol-erance, and interaction with coadministered drugs hasled to better patient management and a decrease intoxicity and mortality. However, therapeutic drug mon-itoring practices for this drug (e.g., reference ranges,toxic concentrations, dosing regimens) stifi vary consid-erably throughout clinical laboratories. Examining 666institutions participating in Q-Probes (a subscriptionquality-improvement program of the College of ClinicalPathologists), Howanitz and Steindel (70) found thatparticipants 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 ION

CONCENTRATION

1INCREASE IN INTRACELLULAR Ca2 ION CONCENTRATION

POSITIVE INOTROPY Ca2 OVERLOAD LEADINGTO CARDIAC GLYCOSIDE TOXICITY

Fig. 7. Schematic representation of the extra- and intracellularevents that lead to increasedcontractileforce (positiveinotropy)orpossible cardiac glycoside toxicity.From MacGregor and Walker (19); used with permission.

g/L) and 16 different upper limits (1.1 to >2.7 pg/L) fortheir therapeutic digoxin ranges. Some blood sampleswere drawn <6 h after dosing. Blood sampling at inap-propriate times may result in digoxin serum concentra-tions in excess of the therapeutic range, and therebyincrease the likelihood of an erroneous decision to with-hold digoxin or even to administer immunoglobulinfragments of digoxin-specific antibodies (used in thetreatment of digoxin toxicity) (70).

Additionally, in some individuals, digoxin is biotrans-formed into metabolites with variable cross-reactivityin digoxin immunoassays (71, 72). Even if these metab-olites are not biologically active, their cross-reactivity indigoxin immunoassays could result in digoxin underdos-ing (72, 73). However, we (72) and others (74) havefound that some but not all of these metabolites havesignificant Na,K-ATPase inhibitory activity, the clini-cal importance of which has not been fully addressed.

Recent important findings suggest that the alpha sub-units of the Na,K-ATPase exhibit species and isoformvariation in their affinity for binding of cardenolides(75). Differences in binding affinities, previously as-cribed to variables such as assay conditions and ionconcentrations, now include differences attributable tothe presence of high- and low-affinity alpha subunitmolecular forms. Originally termed “a” and “a+ ,“ themolecular cloning of the Na,K-ATPase from variousspecies demonstrated that a+ is actually represented bythe two distinct isoforms now referred to as alpha 2 andalpha 3. The alpha 2 and alpha 3 subunits are theisoforms with greatest sensitivity to ouabain in the rat(76-78). Charlemagne et al. (79) describe high- andlow-affinity ouabain-binding sites in rat heart, with re-spective apparent dissociation constants in the 10-8 to10_6 mol/L range. Age, ion concentration, hormone con-centrations, and pathological conditions have all beendemonstrated to correlate with changes in isoform ex-pression (37). Isoform variation in affinity for digoxinand other cardiac glycosides should be considered incases where the response to cardiac glycoside therapy isinappropriate. However, work in this area is still pre-liminary. For example, Schmidt et al. (80) quantifiedthe digitalis receptor concentration in the left ventricleat autopsy, comparing patients without heart diseasewith those with end-stage heart failure who had re-ceived digitalis therapy. Previous work, based on theuse of in vitro systems and tissue culture, had shown anincrease in expression of Na,K-ATPase in response toincubation with digitalis, leading to speculation thatpatients might develop tolerance to digitalis therapy(81). However, rather than increased expression,Schmidt et al. (80) showed a lower concentration ofdigitalis receptors in failing hearts than in the controlsubjects. These investigators did not address the pres-ence or absence of specific high- or low-affinity alphaisoforms, because their quantification of digitalis recep-tors was based on [3Hlouabain binding. Affinity for oua-ham is affected by the presence or absence of certainamino acid residues at the amino terminus of the alphasubunit that correspond to isoform type (82); therefore,

Page 6: UnderstandingtheSodiumPumpandItsRelevancetoDisease · Thesodium-potassium-activated adenosinetriphos-phatase (Na,K-ATPase; sodium pump; EC3.6.1.37)isa plasmamembrane-associated proteincomplex

CLINICALCHEMISTRY, Vol. 40, No. 9, 1994 1679

the down-regulation of high-affinity alpha isoformswithout an overall loss in holoenzyme concentrationcould give the same experimental results as a decreasein overall Na,K-ATPase expression. Phenomena such aschanges in subunit expression, distribution in tissues,ligand binding affinity, and endogenous ligand concen-trations hold promise for establishing a new under-standing of the basic mechanisms underlying patho-physiology. These findings maybe of central importancein establishing hypotheses regarding the clinical role ofendogenous ouabain- or digitalis-like factors.

EndogenousLigands:Implicationsin PathologyThe ubiquitous nature of the sodium pump and its

involvement in diverse physiological processes suggeststhat alteration of pump activity by endogenous or xeno-biotic factors may play a key role in many fundamental

physiological processes (e.g., modulation of cardiac con-tractility, control of sodium in the kidney, vascular con-tractility, neurotransmitter release and processing)(19). The presence of a highly conserved Na,K-ATPasebinding domain for cardiac glycoside drugs implies theexistence of natural ligands that act as endogenousmodulator(s) of this transporter.

Substantial evidence suggests that endogenous digi-talis-like and ouabain-like factors exist. In the process ofmonitoring therapeutic digoxin concentrations by vari-ous immunoassay procedures, several investigatorsnoted increased digoxin values in subjects not treatedwith cardiac glycosides (83, 84). Also noted were in-creases in serum digoxin measurements in subjectswhose digitalis therapy had been discontinued (85).Factors giving rise to these apparent digoxin valueswere termed digoxin-like immunoreactive factors(DLIF) (84) or endogenous digitalis-like factors (EDLF)(86). Detectable concentrations of these factors havebeen observed in serum and plasma from healthy adults(87), plasma from volume-expanded dogs (86), new-bOrns (88, 89), pregnant women (90), patients with re-nal impairment (91), and patients with liver dysfunc-tion (92, 93). Aside from DLIF interference with theaccurate measurement of digoxin in human serum,these molecules, because of their structural similarity to

digoxin itself (94), may interact with the Na,K-ATPaseat the digitalis-binding site on the alpha subunit. Pre-sent evidence suggests that the likely tissue source ofthis factor is the adrenal cortex (94, 95).

In addition to the discovery of DLIF, there is compel-ling evidence that ouabain-like factors (OLF) are pre-sent in mammals (96). Hamlyn and Manunta (97) andother investigators (98) have isolated a factor from bothserum and adrenals with ouabain-like properties thatinclude structural similarity to ouabain, Na,K-ATPase

inhibitory activity, and increased concentration inpathophysiological conditions. A review of this workand the potential role of this ouabain-like factor in dis-ease are detailed by Blaustein (53), who summarizes theproposedphysiological effects of endogenous ouabain incontrol of intracellular calcium stores and cell respon-

siveness. Controversy remains, however, about thesource of this endogenous ouabain-like molecule (99).

Substantial arguments still prevail as to what ismeant by digitalis-like activity (100). It is important tounderstand that immunoreactivity does not imply func-tional activity or vice versa. Thus, DLIF should not bemistaken or confused with EDLF or OLF (84), even if,as is suspected, the identity of these factors converges asmore is learned about them. For example, these mole-cules may be related precursors, metabolic products ofeach other, or the same molecule.

Substantial evidence links endogenous digitalis-likefactors with vasoreactivity. Data supporting the hypoth-esis that endogenous digitalis-like factors interact withNa,K-ATPase to induce peripheral vasoconstriction in-clude the following

1) Subjects with some forms of essential hypertensionhave increased sodium-potassium pump inhibitory ac-tivity (101, 102), natriuretic activity (103), and digoxinimmunoactivity in their plasma (104).

2) Spontaneously hypertensive rhesus monkeys havehigh serum concentrations of digoxin-like activity(105).

3) Crude preparations of the natriuretic activity con-strict third-order arterioles, making them more respon-sive to other vasoconstrictive agents such as norepi-nephrine (106).

4) Crude preparations of the natriuretic activity fromurine cause dose-dependent contractions of isolated ano-coccygeus muscle of the rat (which resembles thesmooth muscle of blood vessels) (107).

5) Infusion of ouabain (108, 109) or digoxin (110) intohumans specifically induces peripheral vasoconstric-tion.

6) Injection of antibodies to digoxin lowers the bloodpressure of deoxycorticosterone-salt-retamning hyper-tensive rats (111).

7) Preparations containing digoxin-like immunore-activity from human urine raise blood pressure andprotect rats from acute digitalis toxicity (112).

8) Bolus infusion of digoxin induces vasoconstrictionof epicardial coronary arteries in humans (113).

9) Ouabain-like compounds isolated from human se-rum demonstrate vasoreactivity comparable with thatof ouabain (114).

Cumulatively, the digitalis-like activity of these fac-tors strongly implicate endogenous regulators of theNa,K-ATPase as vasoconstrictive agents involved in theetiology of some hypertensive states.

Clinical Conditions Linked to Dysfunction orModification of Na,K-ATPase ActivityCardiovascularDisease and Hypertension

Pathological conditions in animals and humans in-volving salt and water homeostasis have been associ-ated with alterations in Na,K-ATPase activity and (or)the presence of circulating endogenous digoxin- or oua-bain-like factors (germane articles and reviews are pro-vided in Table 2). Two of the most notable disordersinvolving salt and water homeostasis-heart disease

Page 7: UnderstandingtheSodiumPumpandItsRelevancetoDisease · Thesodium-potassium-activated adenosinetriphos-phatase (Na,K-ATPase; sodium pump; EC3.6.1.37)isa plasmamembrane-associated proteincomplex

Table 2. Clinical conditions correlating with alteredNa,K-ATPase activity or presence of modifiers.

Condition Reference.

Cardiovascular disease and hypertensionHeart disease 80 115 116

Hypertension (HTN) 52. 97, 101, 117, 118, 119

HTN, pregnancy-induced 120, 121, 122

HTN, 123hypothyroidism-related

Early experiments by Pamnani et al. aimed at under-standing the effects of hypertension on the myocardiumand vasculature at the molecular level by using rat

.

one-kidney, one-clip, and reduced renal mass-saline.

models of hypertension (102, 139). They demonstrateddecreased myocardial and vascular Na,K-ATPase activ-ity, suggesting that reduced pump activity might becommon in low-renin and other models of essential hy-pertension. In a more recent study, uninephrectomized

Impaired renal function 91 animals treated with deoxycorticosterone or angioten-Renal disease 84, 85, 124 sin II provided insight into the molecular mechanisms

Diabetes and other metabolic disorders that may be involved in some forms of hypertensionAcromegaly (140). Separation of mRNA by Northern analysis of ratAldosteronism cardiac left ventricle, aorta, and skeletal muscle RNA,Diabetes mellltus 128 by use of Na,K-ATPase alpha isoform-specific cDNAObesity

Digoxin toxicityAge-relatedDisease-related .

MultipledruginteractionFetal abnormalities

Growth retardation 130Renal abnormality 130H droce halus 130

aneuploidy ‘

Nonimmune hydrops 130

probes, showed tissue-specific changes in isoform ex-pression of mRNA transcripts in response to increasedintravascular pressure.

Greater concentrations of DLIF and OLF have beennoted in women with pregnancy-induced hypertension

.

(preeclampsia) than in normal pregnancy (84, 141).Pregnancy is a volume-expanded condition, and in bothnormal and hypertensive pregnancy the anomalousDLIF values resolve rapidly upon delivery (90, 120-122). Increased concentrations of DLIF are associated

Low birth weight 131 with acute and chronic renal disease (91, 124) and withPreterm infants 132 fluid retention due to hepatic failure (92, 93). Endoge-

Neurological disorders nous OLF is reported markedly increased in hyperten-Alzheimer disease 133, 134, 135, 136 sion that is due to hypothyroidism (123) and congestiveBipolar disorder 64 heart failure (116). DLIF is also significantly increased

Pulmonary conditions in the plasma of human subjects with electrocardio-Pulmonary disease 137 graphic evidence of reversible cardiac dysfunction in-Chronic obstructive 138

pulmonary diseaseduced by physical exhaustion (142). Weinberg et al.(143) used peritoneal dialysis fluid from patients withchronic renal failure to chromatographically isolate

three molecular species having DLIF activity. Of theand hypertension-are causally related. Hypertension three, one had a retention time identical to ouabain, andincreases the risk of myocardial infarction, congestive one had a retention time identical to digoxin. And Yuanheart failure, renal failure, and cerebral stroke (52). et al. (144) showed that administration of chronic lowThe initial reduction in myocardial contractility that doses of ouabain was associated with the development ofoccurs in some forms of heart failure results in vasocon- hypertension in normotensive rats as well as in ratsstriction and peripheral resistance. The constriction of having various degrees of reduced renal mass. Mean

the vascular beds in the kidneys causes salt and water blood pressure increased with the degree of mass reduc-retention. Alterations in Na,K-ATPase activity or ex- tion but was significantly greater than in controls evenpression can alter vascular or cardiac contractility by for rats with no renal mass reduction.affecting sodium homeostasis (37). Thus, the sequence In an effort to determine whether ouabain itself actsof events previously described strongly correlates with as a hypertensive agent or simply exacerbates the hy-the involvement of a circulating inhibitor of sodium pertensive action of mineralocorticoids, Sekihara et al.pump activity in the pathogenesis of both cardiovascu-lar disease and hypertension.

Hypertension, as evidenced by persistently high arte-

(145) treated mononephrectomized rats with ouabain (1mg), deoxycorticosterone acetate (5 mg), or a combina-tion of both, weekly for 6 weeks. Both ouabain and

rial blood pressure, can be idiopathic or secondary to deoxycorticosterone acetate lacked hypertensive actionunderlying conditions. Essential (idiopathic hyperten- individually at the dosage given but, in combination,sion) is a poorly understood though relatively common they produced a significant increase in blood pressure asdisease. An inherited predisposition has been suggested, well as cardionephromegaly and histopathologicaland such individuals may be especially sensitive to di- changes consistent with the effects of an elevation inetary salt (117). Hypertension secondary to primary blood pressure. The authors concluded that, in thosealdosteronism (126), other endocrine disorders, and hypertensive individuals secreting greater concentra-pregnancy often tends to resolve once the underlying tions of mineralocorticoids, ouabain might amplify theproblem is alleviated, hypertensive effect.

1680 CLINICAL CHEMISTRY, Vol. 40, No. 9, 1994

119, 125126118, 119, 127,119

64,65,6769

Page 8: UnderstandingtheSodiumPumpandItsRelevancetoDisease · Thesodium-potassium-activated adenosinetriphos-phatase (Na,K-ATPase; sodium pump; EC3.6.1.37)isa plasmamembrane-associated proteincomplex

CLINICAL CHEMISTRY, Vol. 40, No. 9, 1994 1681

DLIF, isolated from urine by use of cross-reactivity todigoxin antibody as evidence of activity, was admiriis-tered to normotensive rats to determine its cardiovas-cular effects (112). After an initial 60-mm stabilizationperiod, infusion of DLIF in 10 animals caused the meanarterial pressure to rise from 124.3 (±1.9) to 140.4(±5.2) mmHg at 7.5 mm, induced diuresis, and slowedthe heart rate. These results, coupled with data docu-menting the presence of endogenous digitalis-like com-pounds 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 hyper-tension (127), found that not only do type I (insulin-depen-dent) diabetic individuals have a familial predispositionfor essential hypertension, but also normotensive offspringof parents who are nondiabetic but have essential hyper-tension show increased concentrations of plasma insulinand reduced insulin sensitivity; moreover, Na retentionis characteristic of both type I and type II (non-insulin-dependent) diabetics. These authors also report that intra-cellular calcium is increased in adipocytes, in part viainsulin’s inhibition of Ca2,Mg-ATPase, and that insu-lin may increase renal sodium retention and influence theactivity of transmembrane electrolyte pumps. Insulin reg-ulation of vascular Na,K-ATPase gene expression is citedby Tirupattur et al. (148) as an important factor in thedevelopment of hypertension in diabetes. mRNA encoding

both the alpha 1 and alpha 2 isoforms was identified invascular smooth muscle cells derived from embryonic ratthoracic aorta. Although the predominant isoform wasalpha 1, only the concentrations of the alpha 2 isoformincreased in response to insulin treatment. The overallincrease in ouabain-inhibitable Na,K-ATPase activity invascular smooth muscle cells in response to insulin treat-ment suggests that, in the absence of insulin or in insulin-resistant states, Na,K-ATPase activity could decrease, re-sulting in increased vascular contractility and bloodpressure.

In their review, Clerico and Giampietro (119) citereports of decreased sodium pump activity in the nerves,heart, and aorta of diabetic humans, and in rats withstreptozotocin- or alloxan-induced diabetes; however,there was a paradoxical increase in Na,K-ATPase activ-ity in the kidneys of streptozotocin-induced diabeticrats. The authors speculate that these tissue differencescould be due to tissue-specific regulation of Na,K-ATPase activity or metabolism. They further suggestthat in metabolic diseases such as diabetes meffitus,obesity, and acromegaly (which have in common in-creased sodium retention and volume expansion) in-creased sodium intake, hyperinsulinemia, or increasedconcentrations of growth hormone could trigger the re-lease of an endogenous digitalis-like factor that couldmodulate the pump and increase blood pressure. A morerecent study by Chen et al. (128), using a similar dia-betic rat model system, confirmed the previous findings.In addition, Chen et al. found greater amounts of adigitalis-like factor in the plasma and urine of their

hypertensive diabetic rats than in controls. A review bySewers and Khoury (118) highlights the additional riskof cardiovascular disease when hypertension accompa-nies diabetes mellitus and discusses additional risk fac-tors such as obesity, genetics, and ion transport control.

Digoxin Toxicity

Although advanced age is not a pathological condi-tion, age-related changes in renal clearance and volumeof distribution can increase the likelihood of digoxintoxicity (65). In addition, evidence based on basal met-abolic rate measurements indicates that whole-bodyNa,K-ATPase activity decreases with age (64, 129).This suggests that, even though their serum cligoxinconcentrations may be within the therapeutic range,digitalis toxicity in the elderly might result, in part,from inhibition of an already less-active sodium/potas-sium transport system. Low tolerance to digoxin is alsoassociated with certain disease states, particularly thoseinvolving hypokalemia, and with multiple drug interac-tion, as discussed previously.

Fetal Abnormalities

The neonatal period is associated with volume expan-sion and sodium imbalance. As stated previously, age-related changes in Na,K-ATPase alpha isoform expres-sion in the rat have been observed (23). DLIF issignificantly increased in the plasma of newborn infantsbut usually returns to normal within several days (88,132). Increased concentrations of digoxin-like immuno-reactivity have been observed in fetuses having growthretardation, renal abnormality, hydrocephalus, aneu-ploidy, and nonimmune hydrops (reviewed in ref. 130),again suggesting a role for endogenous pump modifiersin the regulation and genetic expression of Na,K-ATPase and in the etiology of some pathological pro-cesses.

Neurological Disorders

Bipolar illness (manic depression) is characterized bysevere mood swings that alternate between episodes ofirritability, excessive energy, and distractibility (ma-nia), and mental and motor slowdown to the point ofstupor (depression). Patients with bipolar illness exhibitaltered 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-ATPaseactivity could lead to mania by increasing membraneexcitability and neurotransmitter release. A greater de-gree of pump inhibition, and consequently depolariza-tion block, could result in depression by decreasing neu-rotransmitter release (149). Patients in manic statesshow increased sodium retention and intracellular cal-cium concentrations, and therapeutic modalities such aslithium or calcium-channel blockers are theorized toaffect sodium-calcium exchange (64). Interestingly,symptoms mimicking those observed in bipolar patientsoccur with digitalis toxicity. These can include confu-sion, disorientation, drowsiness, lethargy, agitation,and hallucinations (137), further implicating Na,K-

Page 9: UnderstandingtheSodiumPumpandItsRelevancetoDisease · Thesodium-potassium-activated adenosinetriphos-phatase (Na,K-ATPase; sodium pump; EC3.6.1.37)isa plasmamembrane-associated proteincomplex

1682 CLINICAL CHEMISTRY, Vol. 40, No. 9, 1994

ATPase as an etiological agent in bipolar illness. Recentevidence in our laboratory suggests that DLIF (mea-sured with a digoxin-specific immunoassay) is increasedin acutely psychiatrically ill bipolar patients relative torecovered bipolar patients (unpublished). Increased en-dogenous ouabain-like compounds that excessively sup-press Na,K-ATPase have been proposed as a mechanismin this disorder (133).

Alzheimer disease (Al)) is an age-related neurodegen-erative disorder of unknown etiology. In addition towell-documented neuropathological changes such asneurofibrillary tangles and neuritic plaque formationwith accumulation of beta-amyloid (136), AD is associ-ated with lower cerebral blood flow and decreased use ofoxygen and glucose, especially in areas exhibiting neu-ropathological changes (134). About 50% of the energyexpenditure of resting brain is believed to supportNa,K-ATPase activity. Hank et al. (134) found a de-crease in ouabain binding of brain tissue from AD pa-tients in comparison with age-matched controls. Thereduction in ouabain binding was especially evident inthe neocortex, an area predominantly affected in Al)patients. A decrease in ouabain-inhibitable Na,K-ATPase activity in the brain subcortical but not corticalareas of patients with Al) was also noted by Liguni et al.(135). Although sodium pump dysfunction may not becausal in this disorder, the progressive dementia asso-ciated with AD may in part be due to alterations inNa,K-ATPase activity. To date, the presence of possibleendogenous pump modifiers in relation to AD has notbeen described.

Pulmonary Conditions

Little has been published in this area. Varsano et al.(138) used a digoxin radioimmunoassay to show thatpatients with advanced chronic respiratory failure hadgreater concentrations of DLIF than did controls.Chronic obstructive pulmonary disease and other formsof advanced respiratory disease are frequently associ-ated with water and sodium retention, and the authorssuggested that increased DLIF might be an attempt tocontrol water and sodium metabolism.

In summary, conditions associated with volume ex-pansion or alterations in sodium homeostasis (see Table2), many of which have been described here, also showchanges in Na,K-ATPase activity, increases in endoge-nous digitalis-like substances, or both. Modification ofpump activity can occur in response to changes in iso-form expression or modulation by endogenous pumpinhibitors or activators. As to whether altered regula-tion of Na,K-ATPase activity is directly linked to any ofthese conditions remains to be elucidated. Preliminarydata identifying polymorphisms in the human Na,K-ATPase alpha and beta subunit genes offer the potentialof linking these and other genetic differences to specificclinical disorders. Locating genetic mutations of Na,K-ATPase or of the endogenous modifiers may soon beadded to the repertoire of test capabifities in the clinicalchemistry laboratory, providing early identification of

individuals at risk for a variety of Na,K-ATPase-asso-ciated diseases.

This work was supported in part by Grant RO1-HL36172 fromthe National Institutes of Health.

References1. Fozzard HA, Sheets MF. Cellular mechanism of action ofcardiac glycosides. J Am Coil Cardiol 1985;5:10-5A.2. Akera T. Membrane adenosinetriphosphat.ase: a digitalis recep-tor? Science 1977;198:569-74.3. Forbush B, Kaplan JH, Hoffman JF. Characterization of a newphotoaffinity derivative of ouabain: labeling of the large polypep-tide and of a proteolipid component of the Na,K-ATPase. Biochem-istry 1978;17:3667-76.4. Ovchinnikov YA, Arzainazova NM, Arysturkhova EA,Gevondyan NM, Aldanova NA, Modyanov NN. Detailed structuralanalysis 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 thecatalytic subunit of the (Na’,K) ATPase deduced from a comple-mentary DNA. Nature 1985;316:691-5.6. Lingrel JB, Orlowski J, Shull MM, Price EM. Molecular genet-iceof Na,K-ATPase. Prog Nucleic Acid Res Mol Biol 1990;38:37-89.7. Karlish SJD, Goldshleger R, Jorgensen PL. Location of Asn’of the a chain of Na/K-ATPase at the cytoplasmic surface. Impli-cation for topological models, J Biol Chem 1993;268:3471-8.8. Karlish SJD, Goldshleger R, Tal DM, Stein WD. Structure ofthe cation binding sites of Na/K-ATPase. In: Kaplan JH, DeWeerP, eds. The sodium pump: structure, mechanism, and regulation.New York: Rockefeller Univ. Press, 1991:129-41.9. Mohraz M, Arystarkhova E, Sweadner KJ. Immunoelectronmicroscopy of epitopes on Na,K-ATPase catalytic subunit. J BiolChem 1994;269:2929-36.10. Farley RA, Trant CM, Carilhi CT, Hawke D, Shively JE. Theamino acid sequence of a fluorescein-labeled peptide from theactive site of (Na,K)-ATPase. J Biol Chem 1984;259:9532-5.11. Karlish SJD, Goldshleger R, Stein WD. A l9kDa C-terminaltryptic fragment of the a chain of Na/K-ATPase is essential forocclusion and transport of cations. Proc Natl Aced Sci USA1990;87:4566-70.12. Rogers TB, LazdunskiM. Photoaflinity labeling of the digitalisreceptor in the (sodium + potassium)-activated adenosinetriphos-phatase. Biochemistry 1979;18:135-40.13. Walderhaug MO, Post RL, Saccomani G, Leonard RT, BriskinDP. Structural relatedness of three ion-transport adenosinetriphosphatases around their active sites of phosphorylation. JBiol Chem 1985;260:3852-9.14. Farley RA, Miller RP, Kudrow A. Orientation of the 13subunitpolypeptide of (Na +K)ATPase in the cell membrane. BiochimBiophys Acta 1986;873:136-42.15. Hiatt A, McDonough AA, Edelman IS. Assembly of the(Na,K)-adenosine triphosphatase. Post translational membraneintegration of the alpha-subunit. J Biol Chem 1984;259:2629-35.16. McDonough AA, Geering K, Farley RA. The sodium pumpneeds its 13subunit. FASEB J 1990;4:1598-605.17. Jorgensen PL. Structure, function and regulation of Na,K-ATPase in the kidney. Kidney Int 1986;29:1O-20.18. Sen AK, Tobin T, Post RC. A cycle for ouabain inhibition ofsodium and potassium-dependent adenosine triphosphatase. J BiolChem 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, AlhikmetaRL, Ushkaryov YA, Melkov AM, et al. The family of humanNa,K-ATPase genes. A partial nucleotide sequence related tothe a-subunit. FEBS Left 1987;213:73-80.21. Shull GE, Greeb J, Lingrel JB. Molecular cloning of threedistinct 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,K-ATPase. In: Kaplan JH, DeWeer P, eds. The sodium pump:structure, mechanism, and regulation. New York: RockefellerUniv. Press, 1991:63-76.

Page 10: UnderstandingtheSodiumPumpandItsRelevancetoDisease · Thesodium-potassium-activated adenosinetriphos-phatase (Na,K-ATPase; sodium pump; EC3.6.1.37)isa plasmamembrane-associated proteincomplex

CLINICAL CHEMISTRY, Vol. 40, No. 9, 1994 1683

23. Lucchesi PA, Sweadner KJ. Postnatal changes in Na,K-ATPase isoform expression in rat cardiac ventricle. J Biol Chem1991;266:9327-31.24. Jewell EA, Shamraj Ol, Lingrel JB. Isoforms of the alphasubunit of Na,K-ATPase and their significance. Acta PhysiolScand 1992;146:161-9.25. Lytton JJ, Lin C, Guidotti G. Identification of two molecularforms 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 132subunit is expressed in rat brain and copurifles with Na,K-ATPaseactivity. 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 Chem1989;264:4613-8.28. Akopyanz NS, Broude NE, Vinogradova NG, Balabanov YA,Monastyrskaya GS, Sverdlov ED. Differential expression of threeNa,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 demon-stration of a3 isozyme of Na,K-ATPase in human brain. NeurosciLeft 1991;130:37-40.30. Martin-Vasallo P, Ghosh 5, Coca-Prados M. Expression ofNa,K-ATPase alpha subunit isoforms in the human ciliary bodyand cultured ciliary epithelial cells. J Cell Physiol 1989;141:243-52.31. Shamraj 01, Melvin D, Lingrel JB. Expression of Na,K-ATPase isoforms in human heart. Biochem Biophy Res Commun1991;179:1434-40.32. Hundal HS, Marette A, Ramlal T, Lie Z, Klip A. Expression of13subunit 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-ATPasein several tissues of the rat: tissue-specific expression of subunitmRNAs and enzyme activity. J Membr Biol 1993;131:229-36.34. Sweadner KJ. Overlapping and diverse distribution of Na-KATPase isozymes in neurons and glia. Can J Physiol Pharmacol1992;70:S255-9.35. Zahler R, Brines M, Kashgarian M, Benz EJ Jr. The cardiacconduction system in the rat expresses the a2 and a3 isoforms ofthe Na’,K-ATPase. Proc Natl Acad Sci USA 1992;89:99-103.36. Jewell EA, Lingrel JB. Chimeric rat Na,K-ATPase alIa3isoforms. Analysis of the structural basis for differences in Narequirements in the al and a3 isoforms. Ann NY Acad Sci1992;671:120-.33.37. McDonough AA, Hensley CB, Azuma KK Differential regu-lation 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,K-ATPase alpha isoform (al, a2, a3), 131,and 132/AMOG genes in theocular ciliary epithelium. In: Kaplan JH, DeWeer P, eds. Thesodium 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) isa homologue of the 13subunit of the Na,K-ATPase. J Cell Biol1990;110:165-74.40. Good PJ, Richter K, Dawid lB. A nervous system-specificisotype of the p subunit of Na,K-ATPase expressed during earlydevelopment of Xenopus laevis. Proc Natl Acad Sci USA 1990;87:9088-92.41. Canfleld VA, Okamoto CT, Chow D, Dorfman J, Gros P, ForteJG, Levenson R. Cloning of the H,K-ATPase 13subunit. Tissue-specific expression, chromosomal assignment, and relationship toNa,K-ATPase p subunits. J Biol Chem 1990;265:19878-84.42. Reuben MA, Laaater LS, Sachs G. Characterization of a 13subunit of the gastric H/K-transporting ATPase. Proc NatlAcad Sd USA 1990;87:6767-71.43. Shull GE. eDNA cloning of the 13-subunit of the rat gastricH,K-ATPase. J Biol Chem 1990;265:12123-6.44. Orlowski J, Lingrel JB. Thyroid and glucocorticoid hormonesregulate the expression of multiple Na,K-ATPase genes in cal-

tured neonatal rat cardiac myocytes. J Biol Chem 1990;265:3462-70.45. 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 BiolChem 1990;265:14305-14.46. Gick GG, Ismail-Beigi F. Thyroid hormone induction ofNa,K-ATPase and its mRNAs in a rat liver cell line. Am JPhysiol 1990;258:C544-51.47. Barlet-Bas C, Khadouri C, Marsy 5, Doucet A. Enhancedintracellular sodium concentration in kidney cells recruits a latentpool of Na-K-ATPase whose size is modulated by corticosteroids. JBiol Chem 1990;265:7799-803.48. Blot-Chabaud M, Wanstok F, Bonvalet J, Farman N. Cellsodium-induced recruitment of Na-K-ATPase pumps in rabbitcortical collecting tubules is aldosterone-dependent. J Biol Chem1990;265:11676-81.49. Lingrel JB, Orlowski J, Price EM, Pathak BG. Regulation ofthe a-subunit genes of the Na,K-ATPase and determinants ofcardiac glycoside sensitivity. In: Kaplan JH, DeWeer P, eds. Thesodium pump: structure, mechanism, and regulation. New York:Rockefeller Univ. Press, 1991:1-16.50. Shull MM, Pugh DG, Lingrel JB. MspI and PvuII polymor-phisms 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 PvuIIpolymorphisms 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 bloodpressure? [Review]. Experentia 1992;48:1102-6.53. Blaustein MP. Physiological effects of endogenous ouabain:control of intracellular Ca2 stores and cell responsiveness [Re-view]. Am J Physiol 1993;264(Cell Physiol 33):C1367-87.54. Hoffman BF, Bigger JT Jr. Cardiovascular drugs. Digitalisand allied cardiac glycosides. In: Gilman AG, Goodman LS, Gil-man 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 monoclonalantibody that enhances ouabain binding. J Biol Chem 1992;267:13694-701.56. Schultheis PJ, Waffick ET, Lingrel JB. Kinetic analysis ofouabain binding to native and mutated forms of Na,K-ATPase andidentification of a new region involved in cardiac glycoside inter-actions. J Biol Chem 1993;268:22686-94.57. O’Brien WJ, Wallick ET, Lingrel JB. Amino acid residues ofNa,K-ATPase involved in ouabain sensitivity do not bind thesugar moiety of cardiac glycosides. J Biol Chem 1993;268:7707-12.58. Thomas R. Cardiac drugs. In: Wolff M, ed. Burgers medicinalchemistry. 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 pharma-cology. Berlin: Springer-Verlag, 1981:337-68.60. Akera T, Brody TM. The role of Na,K-ATPase in theinotropic action of digitalis [Review). Pharmacol Rev 1978;29: 187-220.61. Akera T, Larsen FS, Brody TM. The effect of ouabain onsodium- and potassium-activated adenosine triphosphatase fromthe hearts of several mammalian species. J Pharmacol Exp Ther1969;170:17-26.62. Somberg JC, Barry WH, Smith TW. Differing sensitivities ofPurkinje fibers and myocardium to inhibition of monovalent cationtransport by digitalis. J Clin Invest 198 1;67:116-23.63. Smith JR, Gheorghiade M, Goldstein S. The current role ofdigoxin in the treatment of heart failure. Coronary Artery Dis1993;4:16-26.64. El-Maflakh RS, Barrett JL, Wyatt RJ. The Na,K-ATPasehypothesis for bipolar disorder: implications for normal develop-ment. J Child Adolesc Psychopharmacol 1993;3:37-52.65. Krisanda TJ. Digitalis toxicity. Using immunotherapy whensupportive care isn’t enough. Postgrad Med 1992;91:273-83.66. Langer GA. The “sodium pump lag” revisited. J Mol CellCardiol 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,

Page 11: UnderstandingtheSodiumPumpandItsRelevancetoDisease · Thesodium-potassium-activated adenosinetriphos-phatase (Na,K-ATPase; sodium pump; EC3.6.1.37)isa plasmamembrane-associated proteincomplex

1684 CLINICAL CHEMISTRY, Vol. 40, No. 9, 1994

Goldberg H, Skorecki KL. Identification of P-glycoprotein in renalbrush border membranes. Biochem Biophy Res Commun 1989; 162:244-52.69. deLannoy LAM, Silverman M. The MDR1 gene product, P-gly-coprotein, mediates the transport of the cardiac glycoside, digoxin.Biochem Biophys Res Commun 1992;189:551-7.70. Howanitz PJ, Steindel SJ. Digoxin therapeutic drug monitor-ing practices. Arch Pathol Lab Med 1993;117:684-90.71. Valdes R Jr, Brown BA, Graves SW. Variable cross-reactivityof digoxin metabolites in digoxin immunoassays. Am Clin Pathol1984;82:210-3.72. Miller JJ, Straub RW, Valdes R Jr. A digoxin immunoassay inwhich cross-reactivity of digoxin metabolites is proportional totheir biological activity. Clin Chem 1994;40:in press.73. Soldin SJ. Digoxin-issues and controversies [Review]. ClinChem 1986;32:5-12.74. Hug E, Brown L, Erdmann E. The ouabain receptor in themyocardium and conduction system of the sheep heart. In: Erd-mann 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 theNa,K-ATPase a subunit: site-directed mutagenesis of glutamine-111 to arginine and asparagine-122 to aspartic acid generates aouabain-resistant enzyme. Biochemistry 1988;27:8400-8.76. Sweadner KJ, Farshi SK. Rat cardiac ventricle has twoNa,K-ATPases with different affinities for ouabain: develop-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 a3isozyme of rat Na,K-ATPase. Biochem Biophys Res Commun1988;156:796-800.78. Kolansky DM, Brines ML, Gilmore-Heber M, Benz EJ Jr. TheA2 isoform of rat Na,K-adenosine triphosphatase is active andexhibits high ouabain affinity when expressed in transfectedfibroblasts. 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 car-diac hypertrophy. J Biol Chem 1986;261:185-9.80. Schmidt TA, Allen PD, Colucci WS, Marsh JD, Kjeldsen K. Noadaptation to digitalization as evaluated by digitalis receptor(Na,K-ATPase) quantification in explanted hearts from donorswithout heart disease and from digitalized recipients with end-stage 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 withdigitoxin or potassium-deficient diet. Eur J Pharmacol 1976;37:189-91.82. Canessa CM, Horisberger JD, Louvard D, Rossier BC. Muta-tion of a cysteine in the first transmembrane segment of Na,K-ATPase 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 prematureinfants [Abstract]. Chin Chem 1976;22:1168.84. Valdes R Jr. Endogenous digoxin-like immunoreactive factors:impact on digoxin measurements and potential physiological im-plications [Review]. Clin Chem 1985;31:1525-32.85. Craver JL, Valdes R Jr. Anomalous serum digoxin concentra-tions in uremia. Ann Intern Med 1983;98:483-4.86. Gruber KA, Whitaker JM, Buckalew VM Jr. Endogenousdigitalis-like substance in plasma of volume-expanded dogs. Na-ture 1980;287:743-5.87. Valdes R Jr, Graves SW. Protein binding of endogenousdigoxin-immunoactive factors in human serum and its variationwith clinical condition. J Clin Endocrinol Metal, 1985;60:1135-43.88. Valdes R Jr, Graves SW, Brown BA, Landt ML. Endogenoussubstance in newborn infants causing false-positive digoxin mea-surements. 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 natureand organ source of digoxin-like immunoreactive substance(s) inthe 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 digoxin-

like substance in patients with renal impairment. Ann Intern Med1983;99:604-8.92. Nanji AA, Greenway DC. Correlation between serum albuminand digoxin-like immunoreactive substance in liver disease. J ClinPharmacol 1986;26: 152-3.93. Sewell RB, Poston L, Wilkinson SP, Williams R. A circulatinginhibitor of leucocyte sodium transport in patients with advancedliver cirrhosis. Clin Sci 1984;66:741-4.94. Shaikh IM, Lau BWC, Siegfried BA, Valdes R Jr. Isolation ofdigoxin-like immunoreactive factors from mammalian adrenalcortex. J Biol Chem 1991;266:13672-8.95. Lichtein D, Samuelov S, Gati I, Wechter WJ. Digitalis-likecompounds in animal tissues. J Basic Chin Physiol Pharmacol1992;3:269-92.96. Haddy F, Pamnani M, Clough D. The sodium-potassiumpump in volume-expanded hypertension. Clin Exp Hypertens1978-79;1:295-336.97. Hamlyn JM, Manunta P. Ouabain, digitalis-like factors andhypertension [Review]. J Hyperten 1992;10:S99-111.98. Mathews WR, DuCharme DW, Hamlyn JM, Harris DW,Mandel F, Clarke MA, Ludens JH. Mass spectral characterizationof an endogenous digitalislike factor from human plasma. Hyper-tension 1991;17:930-5.99. Doris PA. Regulation of Na,K-ATPase by endogenous oua-bain-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 hormoneand essential hypertension. Lancet 1982;i:1450-4.102. Pamnani M, Huot 5, Buggy J, Clough D, Haddy F. Demon-stration of a humoral inhibitor of the Na-K pump in somemodels of experimental hypertension. Hypertension 1981;3(SupplII):II-96-101.103. Haddy FJ. Endogenous digitalis-like factor or factors [Edito-rial]. N Engl J Med 1987;316:621-3.104. Ghione S, Baizan S, Montali U, Raggi A, Clerico A, GazzettiP, Donato L. Plasma digoxin-like immunoreactivity in essentialhypertension: relation to plasma renin activity. J Hypertens 1983;1(Suppl 2):142-4.105. Gruber KA, Rudel LL, Bullock BC, Increased circulatinglevels of an endogenous digoxin-like factor in hypertensive mon-keys. Hypertension 1982;4:348-54.106. Plunkett WC, Gruber KA, Hutchins PM, Buckalew VM.Vascular reactivity is increased by factors in plasma of volume-expanded dogs [Abstract]. Clin Res 1980;28:827A.107. Chakravarty B, Mills IH, Callingham BA. Effects of natri-uretic fractions of human urine on isolated anococcygeus muscle ofthe rat. Renal Physiol 1984;7:205-17.108. Hulthen UL, Boffi P, Kiowski W, Buhler FR. Forearmvasoconstrictor response to ouabain: studies in patients with mildand moderate essential hypertension. J Cardiovasc Pharmacol1984;6:S75-81.109. DeMots H, Rahimtoola SH, McAnulty J}l, Porter GA. Effectsof ouabain on coronary and systemic vascular resistance andmyocardial oxygen consumption in patients without heart failure.Am J Cardiol 1978;41:88-93.110. Williams MH Jr, Zohman LR, Ratner AC. Hemodynamiceffects of cardiac glycosides on normal human subject during restand exercise. J Appl Physiol 1958;13:417-21.111. Kojima I, Yoshiha 5, Ogata F. Involvement of endogenousdigitalis-like substance in genesis of deoxycorticosterone-salt hy-pertension. Life Sci 1982;30:1775-81.112. Shilo L, Pomeranz AP, Rathaus M, Bernheim J, ShenkmanL. Endogenous digoxin-hike factor raises blood pressure and pro-tects 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 normaland atherosclerotic epicardial coronary arteries. Am J Cardiol1991;68:1274-8.114. Bova 5, Blaustein MP, Ludens JH, Harris DW, DuCharmeDW, Hamlyn JM. Effects of an endogenous ouabain-like compoundon 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-ATPaseinhibitory activity and digoxin-like immunoreactivity after acutemyocardial infarction. Cardiovase Res 1991;25:371-7.

Page 12: UnderstandingtheSodiumPumpandItsRelevancetoDisease · Thesodium-potassium-activated adenosinetriphos-phatase (Na,K-ATPase; sodium pump; EC3.6.1.37)isa plasmamembrane-associated proteincomplex

CLINICAL CHEMISTRY, Vol. 40, No. 9, 1994 1685

116. Gottlieb SS, Rogowski AC, Weinberg M, Krichten CM,Hamilton BP, Hamlyn JM. Elevated concentrations of endogenousouabain in patients with congestive heart failure. Circulation1992;86:420-5.117 Haddy FJ, Overbeck 11W. The role of humoral agents involume 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-likefactor the link between hypertension and metabolic disorders asdiabetes mellitus, obesity and acromegaly? [Review]. Clin PhysiolBiochem 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 andin pre-eclampsia. Eur J Clin Invest 1989;19:95-100.121. Gusdon JP Jr, Buckalew VM Jr, Hennessey JF. A digoxin-like immunoreactive substance in preeclampsia. Am J ObstetGynecol 1984;150:83-5.122. Valdes R Jr, Graves SW, Knight AB, Craig HR. Endogenousdigoxin immunoactivity is elevated in hypertensive pregnancy.Prog Clin Biol Res 1988;192:229-32.123. Argento NB, Hamilton BP, Valente WA, Hamlyn JM. In-creased circulating levels of a ouabain-like compound in hypothy-roid hypertension [Abstract]. Hypertension 199 1;18:425.124. Izumo H, Izumo S, Deluise M, Flier JS. Erythrocyte Na,Kpump in uremia. Acute correction of a transport defect by hemo-dialysis. 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 factorin 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 immu-noreactivity in patients with primary aldosteronism. BiochemBiophys Res Cominun 1986;135:41-5.127. Weidmann P, Ferrari P. Central role of sodium in hyperten-sion in diabetic subjects. Diabetes Care 1991;14:220-32.128. Chen 5, Yuan C, Clough D, Schooley J, Haddy F, PamnaniMB. Role of digitalis-like substance in the hypertension of strep-tozotocin-induced diabetes in reduced renal mass rats. Am JHypertens 1993;6:397-406.129. Poehlman ET. Regulation of energy expenditure in aginghumans. 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 pathophysiologi-cal 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, MishraOP, Delivoria-Papadopoulos M. Decreased erythrocyte Na,K-ATPase activity associated with cellular potassium loss in ex-tremely low birth weight infants with nonoliguric hyperkalemia. JPediatr 1993;122:276-84.132 Seccombe DW, Pudek MR, Whitfield MF, Jacobson BE,Wittmann BK, King JF. Perinatal changes in a digoxin-likeimmunoreactive substance. Pediatr Res 1984;18:1097-9.

133. Christo PJ, El-Mallakh RS. Possible role of endogenousouabain-like compounds in the pathophysiology of bipolar illness.Med Hypotheses 1993;41:378-83.134. Hank SI, Mitchell MJ, Kalaria RN. Ouabain binding in thehuman brain. Arch Neurol 1989;46:951-4.135. Liguri G, Taddei N, Latorruca S, Nediani C, Sorbi S. Changesin Na,K-ATPase, Ca2-ATPase and some soluble enzymes re-lated to energy metabolism in brains of patients with Alzheimer’sdisease. Neurosci Lett 1990;112:338-42.136. Markesbery WR. Alzheimer’s disease-a mini review. J KyMed Assoc 1989;87:333-5.137. Smith H, Janz TG, Erker M. Digoxin toxicity presenting asaltered mental status in a patient with severe chronic obstructivelung 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 inadvanced chronic respiratory failure. Chest 1992;1O1:146-9.139. Clough DL, Paxnnani MB, Haddy FJ. Decreased myocardialNa’-K-ATPase activity in one-kidney, one-clip hypertensiverats. Am J Physiol 1983;245:H244-51.140. Herrera VLM, Chobanian AV, Ruiz-Opazo N. Isoform-spe-cific modulation of Na,K-ATPase a-subunit gene expression inhypertension. Science 1988;241:221-3.141. Graves SW, Williams GH. Endogenous digitalis-like natri-uretic 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 isincreased 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 oftwo human digitalis-like substances that are structurally relatedto digoxin and ouabain. Biochem Biophys Res Commun 1992;188:1024-9.144. Yuan CM, Manuals P, Hamlyn JM, Chen S, Bohen E, YeunJ, et al. Long-term ouabain administration produces hypertensionin rats. Hypertension 1993;22:178-87.145. Sekihara H, Yazaki Y, Kojima T. Ouabain as an amplifier ofmineralocorticoid-induced hypertension. Endocrinology 1992;131:3077-82.146. Shimoni Y, Gotsman M, Deutsch J, Kachalsky S, LichtsteinD. Endogenous ouabain-like compound increases heart musclecontractility. Nature 1984;307:369-71.147. Devynck MA, Pernollet MG, De The H, Rosenfeld JB, MeyerP. Endogenous digitalis-like compound in essential hypertension.In: Davison AM, Guillou PJ, eds. Proc European Dialysis andTransplant Association-European Renal Association. London: Pit-man Publ., 1983:489-92.148. Tirupattur PR, Ram JL, Standley PR, Sowers JR. Regulationof Na,K-ATPase gene expression by insulin in vascular smoothmuscle 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: Cole-man P, Higgins G, Phelps C, eds. Progress in brain research. NewYork: Elsevier Science Publ., 1990:221-5.