Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2008 Oct 28.
Published in final edited form as: Nat Clin Pract Nephrol. 2008 Jun 10;4(7):378–392. doi: 10.1038/ncpneph0848

Endogenous digitalis

pathophysiologic roles and therapeutic applications

Alexei Y Bagrov 1,*, Joseph I Shapiro 1
PMCID: PMC2574729  NIHMSID: NIHMS54192  PMID: 18542120

SUMMARY

Endogenous digitalis-like factors, also called cardiotonic steroids, have been thought for nearly half a century to have important roles in health and disease. The endogenous cardiotonic steroids ouabain and marinobufagenin have been identified in humans, and an effector mechanism has been delineated by which these hormones signal through the sodium/potassium-transporting ATPase. These findings have increased interest in this field substantially. Although cardiotonic steroids were first considered important in the regulation of renal sodium transport and arterial pressure, subsequent work has implicated these hormones in the control of cell growth, apoptosis and fibrosis, among other processes. This Review focuses on the role of endogenous cardiotonic steroids in the pathophysiology of essential hypertension, congestive heart failure, end-stage renal disease and pre-eclampsia. We also discuss potential therapeutic strategies that have emerged as a result of the increased understanding of the regulation and actions of cardiotonic steroids.

Keywords: end-stage renal disease, endogenous cardiotonic steroids, hypertension, Na+/K+-ATPase, natriuretic hormone

INTRODUCTION

For many years, high dietary salt intake was suspected to increase the risk of cardiovascular and renal diseases.1,2 During the past few decades, however, the role of salt intake in the development of hypertension has shifted from a topic of debate to a well-established phenomenon.3-6 The effect of dietary salt on cardiovascular disease is indicated by data from several large trials, such as the International Study of Salt and Blood Pressure (INTERSALT)7 and the Dietary Approaches to Stop Hypertension (DASH) study.8 Against this background, an understanding of the specific mechanisms underlying the deleterious effects of salt becomes critically important.

The topic of this Review is the role of endogenous cardiotonic steroids—also known as digitalis-like factors or inhibitors of the sodium/potassium-transporting ATPase (Na+/K+-ATPase)9—in linking dietary salt with cardiovascular and renal disease. We focus particularly on effects in humans and how therapy might be targeted and outcomes affected. Endogenous cardiotonic steroids have been the focus of research at our laboratories for the past two decades. The importance and the very existence of such factors has been a matter of controversy;10,11 however, notable breakthroughs in the field include the identification of specific endogenous cardiotonic steroids in experimental and clinical studies,12-15 the detection of altered concentrations of endogenous cardiotonic steroids in different disease states, the elucidation of the roles of endogenous cardiotonic steroids in these settings,16,17 and, in parallel, the discovery of the signaling functions of the Na+/K+-ATPase and its involvement in many elements of basic cell biology.18,19 These issues, particularly the last, have been reviewed in a series of excellent papers.16-23

The main purpose of the present Review is to emphasize the roles of endogenous cardiotonic steroids in human health and disease—in particular with regard to the renal and cardiovascular systems—and to highlight potential therapeutic targets that have arisen as a result of research in this area. In short, we hope to establish that endogenous cardiotonic steroids have begun their journey from bench to bedside.

HISTORICAL PERSPECTIVE

In the 1960s, it became obvious that the actions of the renin-angiotensin-aldosterone system, vasopressin, and the sympathetic nervous system could not adequately explain physiologic and pathophysiologic responses to acute or chronic expansion of blood volume.24,25 This point was elegantly demonstrated in 1961 in a classic paper by de Wardener and colleagues,26 who demonstrated that natriuresis induced by saline infusion is maintained even if renal perfusion pressure and glomerular filtration rate are prevented from increasing. The factor responsible for this phenomenon (the so-called ‘third factor’) was a topic of great interest in the 1960s and 1970s.9 Cort and Lichardus27 and Buckalew and colleagues28 demonstrated that volume expansion is associated with increased levels of a circulating substance that inhibits active sodium transport in vitro. Extremely important contributions to measuring and understanding the implications of this ‘third factor’ were made by Schrier and co-workers29-31 and by Kramer and Gonick.32 The latter group of investigators demonstrated in rats that volume expansion stimulated the production of a substance that could inhibit activity of the Na+/K+-ATPase in the kidney. In fact, Bricker and colleagues33,34 incorporated the concept of a circulating inhibitor of the Na+/K+-ATPase in their model of renal failure progression and the pathogenesis of the uremic syndrome, termed the ‘trade-off’ schema. In 1980, Gruber et al.35 demonstrated in dogs that plasma volume expansion was associated with elevated levels of circulating digoxin-like immunoreactive material. Shortly thereafter, Hamlyn et al.36 demonstrated that plasma Na+/K+-ATPase-inhibitory activity correlated positively with blood pressure in a group of patients with essential hypertension. At the same time, Kojima et al.37 showed that administration of an antibody to digoxin lowered blood pressure in rats with hypertension induced by administration of deoxycorticosterone and salt.

Interest in the concept of inhibitors of the Na+/K+-ATPase as the so-called third factor decreased during the 1980s and 1990s, in part because of inconsistencies within the experimental data. Most of the assays originally developed for inhibitors of the Na+/K+-ATPase were based on cross-reactivity of the putative endogenous factors with various antibodies to digoxin, but results of these tests varied dramatically.38-40 Probably the most important factor causing this inconsistency is that digitalis, the prototypical inhibitor of the Na+/K+-ATPase, is not natriuretic in normal individuals.41

Enthusiasm for the study of endogenous cardiotonic steroids has steadily increased during the past decade, mainly because several endogenous cardiotonic steroids, including ouabain,12,42,43 marinobufagenin,14,15 telocinobufagin15 and bufalin,13 have been isolated and characterized in both animals and humans (Figure 1). In addition, an effector function of the Na+/K+-ATPase signal cascade has been identified that does not seem to depend on inhibition of the ion transport function of the pump by endogenous cardiotonic steroids, but rather is activated by binding of cardiotonic steroids to the caveolar Na+/K+-ATPase in the presence of Src and the epidermal growth factor receptor (EGFR)19,44-47 (Figure 2). Finally, as a result of the identification of specific endogenous cardiotonic steroids, reliable immunoassays have been developed.16,48-54

Figure 1.

Figure 1

Chemical structures of selected cardiotonic steroids. Ouabain belongs to the cardenolides, a class of cardiotonic steroids found in the common foxglove (Digitalis purpurea) and in other plants. The bufadienolides marinobufagenin and proscillaridin A are found in the cane toad (Bufo marinus) and sea squill (Urginea maritima), respectively. The image ‘Urginea maritima flowers’ by Júlio Reis is reproduced under the Creative Commons Attribution ShareAlike 2.5 License (http://creativecommons.org/licenses/by-sa/2.5/). The image of Bufo marinus is reproduced under the Creative Commons Attribution 3.0 Unported License (http://creativecommons.org/licenses/by/3.0/).

Figure 2.

Figure 2

The two pathways via which binding of cardiotonic steroids to the Na+/K+-ATPase exerts genomic and non-genomic effects. In the classic ‘ionic’ pathway (left), inhibition of the pump function of the Na+/K+-ATPase by CTS results in an increase in cytosolic sodium concentration and a decrease in cytosolic potassium concentration. These changes induce an increase in cytosolic calcium level, which in turn activates a variety of pathways with genomic and non-genomic effects. The pump function of the caveolar Na+/K+-ATPase might be more sensitive to CTS than that of the noncaveolar Na+/K+-ATPase. The ‘signaling’ pathway (right) involves the association of Src with the Na+/K+-ATPase in a caveolar domain. Binding of CTS to the Na+/K+-ATPase activates Src, which in turn transactivates the EGFR and PLC, leading to a cascade that involves generation of ROS, activation of ERK through activation of MEK, activation of Akt (protein kinase B) via PI(3)K, stimulation of endocytosis and activation of PKC. These steps induce the genomic and non-genomic effects of CTS. Note that both the classic and signaling pathways allow intervention at the level of the binding of CTS to the Na+/K+-ATPase, by immunoneutralization or pharmacological antagonism; however, the signaling pathway presents several additional targets for interference, such as Src activation and transactivation of the EGFR, PLC activation, activation of MEK, generation of ROS and activation of PI(3)K. Modulation of the signaling pathway at the level of PKC, ERK and Akt might also be possible. Abbreviations: CTS, cardiotonic steroids; EGFR; epidermal growth factor receptor; ERK, extracellular regulated kinase; MEK, mitogen-activated protein kinase; Na+/K+-ATPase, sodium/potassium-transporting ATPase; PI(3)K, phosphoinositide-3 kinase; PKC, protein kinase C; PLC, phospholipase C; ROS, reactive oxygen species.

SUBTYPES OF ENDOGENOUS CARDIOTONIC STEROIDS

Endogenous cardenolides

In 1991, Hamlyn and co-workers12 isolated from human plasma a cardiotonic steroid that was indistinguishable in every way from the plant-derived cardenolide ouabain, and was, therefore, named endogenous ouabain. Since this report was published, endogenous ouabain has been isolated from bovine adrenal gland,42 bovine hypothalamus43 and rat adrenomedullary cells.55 Mass spectrometry and nuclear magnetic resonance studies have indicated that mammalian endogenous ouabain is identical to plant-derived ouabain.42,43,55 The adrenal cortex and hypothalamus are considered to be the sites of ouabain production in mammals.55-57 Adrenocorticotropic hormone, angiotensin II, vasopressin, and phenylephrine stimulate the release of ouabain from the adrenal cortex in vitro.58,59

Evidence suggests that digoxin is an endogenous cardiotonic steroid60,61 that might be an endogenous antagonist of endogenous ouabain;16,62 however, the extensive variability in the detection of digoxin-like immunoreactive material by highly specific commercial digoxin immunoassays in digoxin-naive populations38-41,63 argues against this idea.

Endogenous bufadienolides

Amphibians produce cardiotonic steroids belonging to the bufadienolide class, which differ from cardenolides in that they possess a doubly unsaturated six-membered lactone ring (Figure 1).64 Bufadienolide-containing preparations from frog and toad skin have been used for the treatment of congestive heart failure in traditional medicines of the Far East.65 The highest levels of bufadienolides are detected in the skin of species that migrate from dry to aquatic environments.66 The skin regulates water and electrolyte homeostasis in amphibians; bufadienolides and the Na+/K+-ATPase appear to be integral to this process.66,67 In support of this concept, brain and skin levels of bufadienolides in toads have been shown to fluctuate in response to changes in environmental salinity.67

The above observations triggered a search for mammalian bufadienolides. Initially, bufalin-like immunoreactive material was detected in human bile and plasma.68-71 Lichtstein et al.13 detected bufalin derivatives in the lenses of several mammalian species by using mass spectroscopy. Other workers14,15 demonstrated marinobufagenin in mammalian plasma and urine by the use of specific immunoassays, mass spectrometry and, subsequently, nuclear magnetic resonance spectrometry.15 Sich et al.72 reported that human plasma and bovine adrenal glands contained material that cross-reacted with antibodies against proscillaridin A, a rare example of a plant-derived bufadienolide (Figure 1). Hilton and co-workers73 identified a bufadienolide compound in human placentae and plasma by the use of mass spectroscopy.

Marinobufagenin emerged as a candidate mammalian endogenous cardiotonic steroid largely because of the findings of studies that characterized the pharmacological properties of amphibian bufadienolides.74-76 Our laboratory showed that venom from the toad Bufo marinus contained digoxin-like immuno-reactive material with vasoconstrictive, Na+/K+-ATPase-inhibiting, and positive inotropic effects.74,75 Subsequently, this substance was identified as marinobufagenin, a steroid previously described in toads.14,15 Other studies found that various antibodies to marinobufagenin cross-reacted with material from human, canine and rat plasma and/or urine.14,51,63,64,76

Komiyama et al.15 used tandem mass spectrometry and nuclear magnetic resonance spectrometry to demonstrate that uremic human plasma contained increased levels of another bufadienolide, telocinobufagin. This bufadienolide differs from marinobufagenin because it has a hydroxyl at position 14 of the lactone ring rather than an epoxy group at position 14/15; the authors hypothesized that telocinobufagin is a natural precursor of marinobufagenin.15

ROLE OF CARDIOTONIC STEROIDS IN HYPERTENSION

Endogenous ouabain

Effects on the kidney

Endogenous ouabain does not fulfill the criteria for classification as a putative natriuretic hormone (i.e. it does not increase sodium excretion), but it does have a role in the adaptation to both sodium depletion and sodium loading. Although a few studies have shown that salt loading of normotensive rats stimulates release of ouabain,77,78 other experiments performed in dogs,79,80 rats81 and humans48,49 have not reported this finding. Among 180 patients with untreated hypertension, plasma levels of endogenous ouabain did not change during 2 weeks of salt loading (administration of 170 mmol sodium per day), but increased following 2 weeks of sodium depletion (intake restricted to 70 mmol per day).48 In another study, salt depletion produced a four-fold rise in mean plasma endogenous ouabain levels in 13 healthy men,50 and salt loading (171 mmol sodium daily) was associated with a 13-fold elevation in plasma endogenous ouabain levels after 3 days; levels decreased within 2 days, but remained higher than baseline.50 This pattern of ouabain response after salt loading has also been observed in Dahl salt-sensitive rats82,83 and in humans with normal blood pressure.84

Several lines of evidence support the idea that endogenous ouabain has a prohypertensive role; these include the ‘adducin paradigm’,85 the induction of hypertension in ouabain-treated rodents, the elevation of endogenous ouabain levels in hypertensive rats, and the central prohypertensive action of this hormone. In rats, chronic peripheral administration of low doses of ouabain (10-50 μg/kg per day) increased arterial pressure and induced cardiac hypertrophy.86-91 Interestingly, administration of digoxin (200 μg/kg per day) actually reversed the ouabain-induced hypertension in these rats.62

A mechanism for the prohypertensive effect of endogenous ouabain has been suggested by experiments performed in Milan hypertensive rats. These rats carry a mutation in the gene that encodes the cytoskeletal protein adducin and also exhibit increased circulating levels of endogenous ouabain.92 Both of these characteristics are associated with heightened expression and activity of the Na+/K+-ATPase in the renotubular epithelium; in the case of adducin, the mutation leads to an increase in the residence time of the sodium pump in the cellular membrane.87,93 Unlike the ouabain-resistant α1 Na+/K+-ATPase, expressed in the renal epithelium, the α1 isoform found in the caveolae of renal tubular cells exhibits remarkable sensitivity to ouabain.87 Subnanomolar concentrations of ouabain in the plasma of Milan hypertensive rats lead to activation of this sodium pump and stimulation of the Src-EGFR-extracellular-signal-regulated protein kinase (ERK)-dependent signaling pathway, which results in renal sodium retention and hypertension.87

Endogenous ouabain can also raise blood pressure by inhibiting the transport function of the α2 Na+/K+-ATPase in vascular smooth muscle, and thus promoting entry of calcium via the sodium-calcium exchanger.88,94 Mice genetically engineered to express the ouabain-resistant α2 Na+/K+-ATPase do not manifest an increase in blood pressure following chronic administration of ouabain, unlike control mice with ouabain-sensitive α2 Na+/K+-ATPases.88 Accordingly, vascular smooth muscle from mice with ouabain-resistant α2 sodium pumps is insensitive to the pressor effect of ouabain.88 Furthermore, mice genetically engineered to have reduced expression of the α2 Na+/K+-ATPase (but not of the α1 Na+/K+-ATPase) become hypertensive and their arteries exhibit enhanced tone in vitro.94

Some studies have not found major differences in ouabain affinity between the caveolar and the non-caveolar Na+/K+-ATPase.47 We have observed, however, that in renal proximal tubular epithelial cells both ouabain-induced signal transduction through the Na+/K+-ATPase-Src-EGFR cascade and ouabain-induced clathrin-mediated endocytosis of the Na+/K+-ATPase were limited to the Na+/K+-ATPase residing in caveolae.95,96 These discrepancies might be explained by different levels of expression of the γ subunit of the Na+/K+-ATPase in the preparations employed, as suggested by Nguyen and co-workers.97

In accord with the sodium-retaining effect of endogenous ouabain on the kidney, polymorphisms of the adducin gene and raised levels of endogenous ouabain are associated with altered renal sodium reabsorption both in experimental animals and in patients with hypertension.98 Furthermore, in Milan hypertensive rats, administration of the digitoxin derivative rostafuroxin (PST 2238)99 antagonized the interacting effect of endogenous ouabain and mutated adducin on the caveolar renal Na+/K+-ATPase, lowered blood pressure and inhibited the activity of the Na+/K+-ATPase in the renal medulla.87

Effects on the brain

Substantial evidence indicates that endogenous brain ouabain contributes to the pathogenesis of salt-sensitive hypertension.100-103 In rats, centrally administered ouabain elicits pressor and natriuretic responses, which are dependent on the activation of the renin-angiotensin system.22,104,105 Likewise, central administration of salt and systemic salt loading both lead to increases in brain levels of endogenous ouabain in Dahl salt-sensitive rats; these increases are mediated by the brain renin-angiotensin system.102,103,105 Leenen and co-workers106,107 demonstrated that an increase in the level of sodium chloride in cerebrospinal fluid precedes the development of hypertension and that sodium ions in the brain enter the intracellular space via epithelial sodium channels. Entry is modulated by central mineralocorticoid receptors.108 Recent evidence indicates, however, that brain-specific sodium channels, rather than epithelial sodium channels, have a key role as sensors of cerebrospinal fluid sodium level.109 The above observations indicate that endogenous ouabain might act as a central mediator of salt-sensitive hypertension (Figure 3).

Figure 3.

Figure 3

Interactions between brain endogenous ouabain, the central renin-angiotensin system, and circulating marinobufagenin in the pathogenesis of salt-sensitive hypertension. In salt-loaded Dahl salt-sensitive rats, sodium retention occurs because renal sodium transport is impaired. Sodium retention stimulates the release of endogenous ouabain in the hippocampus, hypothalamus and pituitary. Brain endogenous ouabain stimulates the RAS in the hypothalamus and pituitary, which activates the SNS. These events stimulate the RAS in the adrenal cortex, and activate adrenocortical production of marinobufagenin. Marinobufagenin, a natriuretic and a vasoconstrictor, induces natriuresis via inhibition of the renotubular Na+/K+-ATPase. Excessive release of marinobufagenin leads to inhibition of the Na+/K+-ATPase in vascular smooth muscle cells—which potentiates vasoconstriction—and in the heart—which increases cardiac contractility—causing hypertension. Abbreviations: Na+/K+-ATPase, sodium/potassium-transporting ATPase; RAS, renin-angiotensin system; SNS, sympathetic nervous system.

Endogenous marinobufagenin

In normotensive rats, plasma levels of marinobufagenin increase in response to acute plasma volume expansion, as well as following chronic administration of a high-salt diet.81,110,111 Enhanced production of marinobufagenin has also been demonstrated in humans with volume expansion,51 pre-eclampsia,52 essential hypertension, primary aldosteronism, and end-stage renal disease.53,54 At concentrations comparable to in vivo plasma levels, marinobufagenin induces vasoconstriction in isolated human pulmonary and umbilical arteries52,76 as well as substantial inhibition of the ouabain-resistant α1 Na+/K+-ATPase in rat aorta and rat renal medulla.83,112 Additionally, immunoneutralization of marinobufagenin with a specific antibody reduces blood pressure and renal sodium excretion in salt-loaded Dahl salt-sensitive rats.83,102 These rats are, in fact, the same experimental model of hypertension in which Lewis Dahl predicted the existence of an endogenous vasoconstrictive and natriuretic substance.113

Interaction between endogenous ouabain and marinobufagenin

In Dahl salt-sensitive rats with salt-induced hypertension, an important interaction seems to occur between brain and peripheral cardiotonic steroids. After acute and chronic salt-loading in these animals, a transient increase in circulating endogenous ouabain precedes a sustained increase in circulating marinobufagenin.82,83 This observation has led us to postulate that endogenous ouabain, acting as a neurohormone, triggers release of marinobufagenin, which in turn causes increases in cardiac contractility, peripheral vasoconstriction and natriuresis by inhibiting the Na+/K+-ATPase (Figure 3).102,103 We have subsequently demonstrated that the greatest transient increases in brain endogenous ouabain level occur in the amygdala, hippocampus and supraoptic nucleus of the hypothalamus.102 Endogenous ouabain in the brain activates the central renin-angiotensin system, which—possibly via sympathoactivation—in turn activates the renin-angiotensin system in the adrenal cortex.102,103 Activation of the adrenocortical renin-angiotensin system facilitates production and secretion of marinobufagenin, which results in increased plasma and urinary levels of the bufadienolide. This sequence of events was fully mimicked by intrahippocampal administration of a very low dose (60 pg) of plant-derived ouabain.102 More recently, we demonstrated that, similar to Dahl salt-sensitive rats, salt-loaded normotensive humans exhibit a transient increase in urinary endogenous ouabain, which precedes a more sustained increase in renal marinobufagenin excretion.84

Thus, it seems that two scenarios involving different patterns of response to cardiotonic steroids are involved in the pathogenesis of hypertension. At least one of these scenarios—the adducin paradigm—has been demonstrated to be relevant to human hypertension: levels of endogenous ouabain become elevated in hypertensive individuals who possess the appropriate mutation or mutations of the adducin gene.98 The clinical relevance of the other scenario, the interplay between brain endogenous ouabain and circulating marinobufagenin, remains to be established.

ROLE OF CARDIOTONIC STEROIDS IN RENAL SALT HANDLING

Although the definition of a natriuretic substance is one that increases urinary sodium excretion, the prototypical cardiotonic steroid—i.e. digitalis or digoxin—is not natriuretic at typical clinical doses.41 Nevertheless, studies indicate that some other cardiotonic steroids do seem to function as natriuretic substances in vitro and in vivo.114 We have observed dose-dependent and time-dependent endocytosis of the plasmalemmal Na+/K+-ATPase induced by cardiotonic steroids in LLC-PK1 cells; this cell line has features of proximal tubule cells.115 In a canine cell line that resembles distal tubular cells, no such depletion was observed. Endocytosis of the plasmalemmal Na+/K+-ATPase has been shown to proceed via caveolae and via clathrin-coated pits following activation of phosphoinositide-3 kinase by the Src-EGFR pathway.95,96 In an in vivo rat model, we noted that endocytosis induced by marinobufagenin contributed to the altered sodium reabsorption seen with increases in dietary sodium.111 Specifically, we observed that increases in dietary sodium led to increased urinary excretion of marinobufagenin and sodium, decreased proximal tubule expression of the Na+/K+-ATPase, increased accumulation of the Na+/K+-ATPase in both early and late endosomes, and decreased sodium reabsorption. Administration of an antibody to marinobufagenin blocked endocytosis of the Na+/K+-ATPase and blunted the increase in urinary sodium excretion.111

Other work has demonstrated that cardiotonic steroids can induce decreases in the apical expression of the sodium/hydrogen exchanger 3 (NHE3) in proximal tubule cells.114,116 Some of the long-term decrease in NHE3 expression caused by cardiotonic steroids is related to decreases in NHE3 transcription, whereas binding of ouabain to the basolateral Na+/K+-ATPase also seems to rapidly induce endocytosis of the apical NHE3.117 Taken together, the above data suggest that salt loading is accompanied by an increase in the circulating level of marinobufagenin, which in turn induces a decrease in both basolateral and apical sodium transport in the proximal tubule, resulting in increased urinary sodium excretion.

ROLE OF CARDIOTONIC STEROIDS IN CONGESTIVE HEART FAILURE

Congestive heart failure (CHF) is associated with fluid retention and plasma volume expansion, conditions under which one would expect cardiotonic steroids to be released (Figure 3).118 As early as 1981, Schreiber, Kölbel et al.119 hypothesized that cardiotonic steroids might be involved in myocardial hypertrophy and the regulation of tissue growth. In 1988, Morise et al.120 demonstrated in rats that development of CHF was associated with increased plasma activity of a Na+/K+-ATPase inhibitory factor. In 1990, Liu et al.121 showed in a group of 50 patients that the severity of CHF was positively associated with the degree of digoxin-like immunoreactivity in plasma, as well as with erythrocyte sodium concentrations. The relationship between cardiotonic steroids, cardiac geometry and central hemodynamic parameters has been analyzed in several studies. Gottlieb et al.122 found that although plasma endogenous ouabain level did not increase as cardiac failure progresses, levels were elevated in patients with severely impaired left ventricular performance (left ventricular ejection fraction less than 30%).

Manunta et al.123 demonstrated that the plasma concentration of endogenous ouabain positively correlated with systolic and diastolic blood pressure in a patient group that comprised 110 normotensive individuals and 100 hypertensive individuals; levels also positively correlated with left ventricular mass index and left ventricular end diastolic volume in the hypertensive patients. Pierdomenico et al.124 found that circulating endogenous ouabain levels in 92 hypertensive patients positively correlated with mean blood pressure and total peripheral resistance index, and that left ventricular end diastolic volume index, stroke index and cardiac index exhibited inverse correlations with the plasma ouabain level. In the same study, the plasma endogenous ouabain level was substantially higher in patients with eccentric remodeling than in those with normal left ventricular geometry or concentric hypertrophy.124 In another study, the plasma ouabain level was higher in patients with left ventricular dysfunction than in normal individuals, but did not correlate with left ventricular ejection fraction.125

Pitzalis et al.126 found that levels of circulating endogenous ouabain predicted the progression of heart failure in 140 patients with idiopathic dilated cardiomyopathy. Levels of endogenous ouabain have also been found to predict ventricular hypertrophy in patients with end-stage renal disease.127

In 23 consecutive hypertensive male patients with CHF, plasma marinobufagenin levels exhibited a progressive increase that paralleled the progression of CHF and the increase in plasma α-atrial natriuretic peptide.54 Although plasma endogenous ouabain levels did not vary with the severity of CHF, it was noted that ouabain levels were substantially elevated in a subset of patients who had an ejection fraction less than 30% (AY Bagrov, unpublished data), a finding similar to that observed by Gottlieb and coworkers.122

Experimental data also indicate an association between elevated plasma levels of cardiotonic steroids and cardiovascular remodeling. In one study, normotensive Fisher 344 × Brown Norway rats subjected to a 4% salt diet exhibited increases in plasma marinobufagenin and proportional increases in cardiac weight, in the absence of hypertension.81 Sustained ouabain infusion, sufficient to cause a two-fold elevation in plasma ouabain immunoreactivity, also induced left ventricular hypertrophy in a study of normotensive rats.87

A study performed in Dahl salt-sensitive rats subjected to a high-salt diet revealed coordinated shifts in the physiological function of the left ventricle (compensated left ventricular hypertrophy progressing to dilated cardiomyopathy), the plasma levels of cardiotonic steroids, and the relative amounts of the Na+/K+-ATPase α1, α2 and α3 isoforms within the left ventricular myocardium.128 Specifically, the advancing stages of hypertrophy were associated with an elevated plasma marinobufagenin level, increased expression of the α1 Na+/K+-ATPase, decreased expression of the α2 Na+/K+-ATPase in the left ventricular myocardium, and heightened sensitivity of the cardiac sodium pump to marinobufagenin. The transition to CHF was associated with a decline in plasma marinobufagenin levels and decreased absolute levels of the α1 Na+/K+-ATPase in the left ventricle. Levels of endogenous ouabain rose substantially with the development of CHF, and this enhanced ouabain production was associated with increased levels of the ouabain-sensitive α3 Na+/K+-ATPase in the left ventricular myocardium, along with an increase in the sensitivity of the cardiac Na+/K+-ATPase to ouabain.128

Ouabain and marinobufagenin induce apoptosis and growth-promoting signaling, respectively, in cultured renal tubular cells.129 The existence of endogenous cardiotonic steroids that have different effects on cell survival makes teleological sense, since different in vivo scenarios might require modulation of sodium-pump-dependent functions either with concurrent cytotoxic effects or without. Thus, the transition from compensated left ventricular hypertrophy to CHF is accompanied by a decrease in plasma marinobufagenin level, but a three-fold increase in plasma endogenous ouabain level occurs at the stage of decompensated CHF,128 when induction of cell death might be considered desirable.130

ROLE OF CARDIOTONIC STEROIDS IN RENAL FAILURE

The modern concept of uremic cardiomyopathy is of ventricular hypertrophy and progressive loss of diastolic function in the context of renal disease, which can ultimately progress to eccentric hypertrophy and, rarely, to systolic dysfunction.131 Echocardiographic studies indicate that diastolic dysfunction and ventricular hypertrophy manifest extremely frequently, whereas systolic dysfunction occurs in less than 20% of patients with end-stage renal disease. Neither the diastolic dysfunction nor the ventricular hypertrophy can be explained solely by the hypertension and anemia that generally complicate end-stage renal disease.132 As mentioned earlier, plasma levels of telocinobufagin and marinobufagenin are substantially elevated in patients with end-stage renal disease.15,53

To examine the role of cardiotonic steroids in renal disease, our laboratory has established models of chronic kidney disease by the use of partial nephrectomy in the rat133 and, more recently, the mouse.134 Animals subjected to partial nephrectomy develop increases in marinobufagenin (similar to those seen in patients with renal disease) and diastolic dysfunction, ventricular hypertrophy, and evidence of signaling through the Na+/K+-ATPase-Src-EGFR-ERK cascade.133,134 Cardiac fibrosis occurs prominently in these models133-136 and also complicates human uremic cardiomyopathy, albeit to a lesser degree.137 In the rat, infusion of marinobufagenin to achieve a similar elevation in plasma level to that seen after partial nephrectomy results in activation of the Na+/K+-ATPase-Src-EGFR-ERK cascade and development of many of the phenotypical features of experimental uremic cardiomyopathy. Active immunization against marinobufagenin attenuates most of the biochemical, physiological and morphological features of uremic cardiomyopathy in animals subjected to partial nephrectomy.135,136

Exposure to very small amounts of marinobufagenin (and of other cardiotonic steroids) virtually identical to the circulating plasma concentrations seen in experimental and clinical renal failure directly stimulated the production of collagen in primary cultures of cardiac fibroblasts.134,135 Again, this effect required signaling through the Na+/K+-ATPase-Src-EGFR cascade. The stimulation of collagen production was associated with increased transcription and translation of procollagen, but no change in the stability of procollagen or collagen was identified.134

ROLE OF CARDIOTONIC STEROIDS IN PRE-ECLAMPSIA

Since pregnancy is associated with plasma volume expansion as a result of renal sodium and fluid retention,138,139 it is logical to examine the role of cardiotonic steroids in pregnancy and in pregnancy-associated diseases. Graves et al.140,141 were the first to demonstrate increased circulating levels of cardiotonic steroids in pregnancy and to hypothesize that cardiotonic steroids are involved in the pathogenesis of pre-eclampsia. After the introduction of assays for specific cardiotonic steroids, levels of endogenous ouabain and marinobufagenin were found to be increased by four-fold and eight-fold, respectively, in patients with severe pre-eclampsia.52 In a study of women with milder pre-eclampsia, elevated levels of marinobufagenin accompanied inhibition of the Na+/K+-ATPase in erythrocytes, but endogenous ouabain levels were not markedly increased; ex vivo treatment of erythrocytes with an antimarinobufagenin antibody—but not an antiouabain antibody—reversed this inhibition.142

The role of cardiotonic steroids in pregnancy is not yet understood; however, in a study of 15 patients with pre-eclampsia, the anti-digoxin antibody Digibind® (GlaxoSmithKline, Philadelphia, PA) lowered blood pressure.143-145 The mechanism underlying this effect is believed to involve cross-reactivity with endogenous cardiotonic steroids.143-145 These data are in agreement with the observation of a vasorelaxant action of Digibind® in isolated perfused placentae from pre-eclamptic women146 and of heightened sensitivity of the Na+/K+-ATPase to digitalis in such placentae.147 A characterization study showed that marinobufagenin-like immunoreactive material from the plasma of pre-eclamptic women exhibits chromatographic properties similar to that of marinobufagenin produced by cultured murine adrenocortical cells.148

The role of cardiotonic steroids in pre-eclampsia has been explored further in experimental animals. Salt supplementation—with drinking water containing 1.8% sodium chloride—during days 14-20 of gestation in pregnant Sprague-Dawley rats was associated with an increase in the plasma levels of marinobufagenin (but not of ouabain), an elevation of blood pressure, proteinuria, and a decrease in fetal weight, size, and number.149 Administration of an anti-marinobufagenin antibody resulted in a 28 mmHg decrease in blood pressure and a simultaneous increase in the activity of the Na+/K+-ATPase in thoracic aortae.149 In pregnant rats rendered hypertensive by deoxycorticosterone acetate and salt supplementation, uterine arteries exhibited enhanced sensitivity to the vasoconstrictor action of marinobufagenin, and an antimarinobufagenin antibody had an antihypertensive effect.150 Thus, it appears that increases in circulating marinobufagenin levels are responsible for the Na+/K+-ATPase inhibition induced by preeclampsia and contribute to the pathogenesis of pre-eclampsia. Accordingly, low concentrations of marinobufagenin impair the differentiation of the cytotrophoblast in vitro.151

ROLE OF CARDIOTONIC STEROIDS IN OTHER CONDITIONS

Considering the fact that cardiotonic steroids regulate the transport and signaling functions of a key membrane enzyme, it is not surprising that the list of disorders in which these factors are implicated is not limited to essential hypertension, end-stage renal disease, pre-eclampsia, and cardiac failure. Several lines of evidence indicate the involvement of cardiotonic steroids in behavioral stress,81,152 physiological response to exercise,153 myocardial ischemia-induced arrhythmias,154 manic-depressive disorder,155,156 and ethanol addiction.157

Elevated levels of cardiotonic steroids, along with perturbed function of the Na+/K+-ATPase, have been found in patients and experimental animals with diabetes mellitus.158,159 More recent evidence indicates that rats with type 1 diabetes exhibit higher urinary levels of marinobufagenin and more profound inhibition of the Na+/K+-ATPase than rats with type 2 diabetes, implying that the degree of glycemia seen in diabetes co-varies with level of marinobufagenin excretion and extent of Na+/K+-ATPase inhibition.160 Along with clinical data demonstrating that cardiotonic steroids are stimulated by oral glucose challenge,161 this evidence implicates cardiotonic steroids in tissue glucose tolerance.

Cardiotonic steroids are also likely to be involved in the pathogenesis of cancer,162 and the growing body of in vitro evidence of growth-promoting but anticancer effects of cardiotonic steroids raises the possibility of using inhibitors of the Na+/K+-ATPase to treat cancer.163

NOVEL THERAPEUTIC POSSIBILITIES

Both the ion transport function of the Na+/K+-ATPase and the Na+/K+-ATPase signaling pathway offer potential targets for therapeutic intervention (Figure 2). Although most of these targets have not yet been specifically addressed, it is interesting to note that some have been investigated, whether deliberately or inadvertently.

The development of antagonists to the Na+/K+-ATPase is an obvious approach to developing therapies that target cardiotonic steroids, but would not discriminate between the two mechanisms of Na+/K+-ATPase activity. Interesting data exist for rostafuroxin, a digitoxin derivative that is designed to serve as an endogenous ouabain antagonist.164 Rostafuroxin has been shown to have beneficial effects in a rodent ouabain infusion model, causing a reduction in blood pressure and in left ventricular and renal weight.87,99 The efficacy of rostafuroxin in hypertensive patients and the possible dependence of this efficacy on the adducin genotype is being assessed in the ongoing phase II multicenter Ouabain and Adducin for Specific Intervention on Sodium in Hypertension (OASIS-HT) study.165 Spironolactone and its major metabolite canrenone are also potential antagonists of cardiotonic steroids. In experimental studies, both spironolactone and canrenone antagonized the binding of ouabain to the Na+/K+-ATPase as well as ouabain-induced inhibition of the pump.166 Canrenone reduced blood pressure and restored activity of the Na+/K+-ATPase in a study of rats with experimental volume-dependent hypertension,167 and has been proposed as a treatment for digitalis toxicity.168

In vivo immunoneutralization of cardiotonic steroids might at first appear an eccentric approach to the treatment of hypertension, and blockade of volume-sensitive hormones in a volume-contracted state may seem counterintuitive. The clinical evidence of the efficacy of Digibind® in pre-eclampsia, however, constitutes one of the most convincing arguments for a prohypertensive role of cardiotonic steroids and for therapeutic immunoneutralization.143-145 A multicenter, double-blind, placebo-controlled efficacy study of Digibind® in pre-eclampsia (DEEP) is in progress.169 Immunoneutralization of cardiotonic steroids might also be effective in patients with cerebral salt-wasting syndrome, a condition that frequently accompanies cerebral injury and is associated with life-threatening natriuresis. For example, in a study by Menezes and co-workers,170 Digibind® reduced renal sodium excretion in a patient following brain tumor removal.

Another potential target for intervention is protein kinase C (Figure 2). Cicletanine, an anti-hypertensive agent that inhibits protein kinase C, is effective in the treatment of experimental salt-sensitive hypertension and attenuates inhibition of the Na+/K+-ATPase both in vitro and in vivo.171 Atrial natriuretic peptide potentiates marinobufagenin-induced inhibition of the renal Na+/K+-ATPase in vitro via cGMP-dependent mechanisms, but reduces marinobufagenin-induced inhibition of the sodium pump in vascular smooth muscle.172 Thus, atrial natriuretic peptide might have the capacity in some settings to antagonize the undesirable effects of cardiotonic steroids while potentiating their desired effects. As our understanding of the Na+/K+-ATPase signaling pathway expands, additional targets for clinical intervention will become obvious.

CONCLUSIONS

Understanding of the role of cardiotonic steroids in health and disease has progressed tremendously during the past few decades, with the discovery of several endogenous cardiotonic steroids in mammals and the elucidation of the roles of these molecules in a wide range of diseases; however, current knowledge is only the tip of the iceberg. We expect that many additional therapeutic avenues will open up as the capacity to measure levels of endogenous cardiotonic steroids improves, the understanding of the biosynthesis, metabolism and regulation of these molecules deepens, and the knowledge of the role of Na+/K+-ATPase signaling in health and disease becomes more detailed.

KEY POINTS.

  • Several endogenous cardiotonic steroids, or digitalis-like factors, have been isolated and characterized in humans; of these, ouabain, from the cardenolide class, and marinobufagenin, from the bufadienolide class, are the most extensively studied

  • In addition to inhibiting the ion transport function of the sodium/potassium-transporting ATPase, binding of endogenous cardiotonic steroids to this sodium pump can activate signaling via the Src-epidermal growth factor receptor pathway

  • In Dahl salt-sensitive rats with salt-induced hypertension, endogenous ouabain acts as a neurohormone and stimulates the release of marinobufagenin, a natriuretic and vasoconstrictor

  • Endogenous cardiotonic steroids are implicated in congestive heart failure, pre-eclampsia and diabetes mellitus

  • Potential therapeutic approaches to targeting endogenous cardiotonic steroids include immunoneutralization, receptor antagonism and protein kinase C inhibition

Acknowledgments

Supported in part by the Intramural Research Program of the National Institute on Aging, part of the NIH. We would like to thank Dr Olga V Fedorova for critical review of the manuscript and Ms Carol Woods for her excellent secretarial efforts.

Footnotes

REVIEW CRITERIA

The authors reviewed manuscripts from their personal libraries and searched PubMed for articles referring to “cardiotonic steroids”, “digitalis-like factors”, “natriuretic hormones”, “Na+/K+-ATPase”, “bufadienolides”, “bufanolides”, “ouabain”, “sodium metabolism”, “sodium pump signaling”, or “hypertension”. No date or language restriction was placed on the search.

Competing interests

The authors declared no competing interests.

References

  • 1.Ritz E. The history of salt—aspects of interest to the nephrologist. Nephrol Dial Transplant. 1996;11:969–975. doi: 10.1093/oxfordjournals.ndt.a027517. [DOI] [PubMed] [Google Scholar]
  • 2.Meneton P, et al. Links between dietary salt intake, renal salt handling, blood pressure, and cardiovascular diseases. Physiol Rev. 2005;85:679–715. doi: 10.1152/physrev.00056.2003. [DOI] [PubMed] [Google Scholar]
  • 3.Taubes G. The (political) science of salt. Science. 1998;281:898–901. doi: 10.1126/science.281.5379.898. [DOI] [PubMed] [Google Scholar]
  • 4.De Wardener HE, MacGregor GA. Sodium and blood pressure. Curr Opin Cardiol. 2002;17:360–367. doi: 10.1097/00001573-200207000-00007. [DOI] [PubMed] [Google Scholar]
  • 5.Weinberger MH. Pathogenesis of salt sensitivity of blood pressure. Curr Hypertens Rep. 2006;8:166–170. doi: 10.1007/s11906-006-0014-y. [DOI] [PubMed] [Google Scholar]
  • 6.Ritz E, et al. Salt—a potential ‘uremic toxin’? Blood Purif. 2006;24:63–66. doi: 10.1159/000089439. [DOI] [PubMed] [Google Scholar]
  • 7.Stamler J, et al. Findings of the international cooperative INTERSALT study. Hypertension. 1991;17(suppl 1):19–15. doi: 10.1161/01.hyp.17.1_suppl.i9. [DOI] [PubMed] [Google Scholar]
  • 8.Appel LJ, et al. for the DASH Collaborative Research Group A clinical trial of the effects of dietary patterns on blood pressure. N Engl J Med. 1997;336:1117–1124. doi: 10.1056/NEJM199704173361601. [DOI] [PubMed] [Google Scholar]
  • 9.de Wardener HE, Clarkson EM. Concept of natriuretic hormone. Physiol Rev. 1985;65:658–759. doi: 10.1152/physrev.1985.65.3.658. [DOI] [PubMed] [Google Scholar]
  • 10.Kelly RA, Smith TW. Is ouabain the endogenous digitalis? Circulation. 1992;86:694–697. doi: 10.1161/01.cir.86.2.694. [DOI] [PubMed] [Google Scholar]
  • 11.Hansen O. No evidence for a role in signal-transduction of Na+/K+-ATPase interaction with putative endogenous ouabain. Eur J Biochem. 2003;270:1916–1919. doi: 10.1046/j.1432-1033.2003.03554.x. [DOI] [PubMed] [Google Scholar]
  • 12.Hamlyn JM, et al. Identification and characterization of an ouabain-like compound from human plasma. Proc Natl Acad Sci USA. 1991;88:6259–6263. doi: 10.1073/pnas.88.14.6259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Lichtstein D, et al. Identification of digitalis-like compounds in human cataractous lenses. Eur J Biochem. 1993;216:261–268. doi: 10.1111/j.1432-1033.1993.tb18141.x. [DOI] [PubMed] [Google Scholar]
  • 14.Bagrov AY, et al. Characterization of a urinary bufodienolide Na+,K+-ATPase inhibitor in patients after acute myocardial infarction. Hypertension. 1998;31:1097–1103. doi: 10.1161/01.hyp.31.5.1097. [DOI] [PubMed] [Google Scholar]
  • 15.Komiyama Y, et al. A novel endogenous digitalis, telocinobufagin, exhibits elevated plasma levels in patients with terminal renal failure. Clin Biochem. 2005;38:36–45. doi: 10.1016/j.clinbiochem.2004.08.005. [DOI] [PubMed] [Google Scholar]
  • 16.Schoner W, Scheiner-Bobis G. Endogenous and exogenous cardiac glycosides: their roles in hypertension, salt metabolism, and cell growth. Am J Physiol Cell Physiol. 2007;293:C509–C536. doi: 10.1152/ajpcell.00098.2007. [DOI] [PubMed] [Google Scholar]
  • 17.Haddy FJ. Role of dietary salt in hypertension. Life Sci. 2006;79:1585–1592. doi: 10.1016/j.lfs.2006.05.017. [DOI] [PubMed] [Google Scholar]
  • 18.Orlov SN, Hamet P. The death of cardiotonic steroid-treated cells: evidence of Na+i,K+i-independent H+i-sensitive signalling. Acta Physiol (Oxf) 2006;187:231–240. doi: 10.1111/j.1748-1716.2006.01546.x. [DOI] [PubMed] [Google Scholar]
  • 19.Pierre SV, Xie Z. The Na,K-ATPase receptor complex: its organization and membership. Cell Biochem Biophys. 2006;46:303–316. doi: 10.1385/cbb:46:3:303. [DOI] [PubMed] [Google Scholar]
  • 20.Nesher M, et al. The digitalis-like steroid hormones: new mechanisms of action and biological significance. Life Sci. 2007;80:2093–2107. doi: 10.1016/j.lfs.2007.03.013. [DOI] [PubMed] [Google Scholar]
  • 21.Wasserstrom JA, Aistrup GL. Digitalis: new actions for an old drug. Am J Physiol Heart Circ Physiol. 2005;289:H1781–H1793. doi: 10.1152/ajpheart.00707.2004. [DOI] [PubMed] [Google Scholar]
  • 22.Huang BS, et al. The central role of the brain in salt-sensitive hypertension. Curr Opin Cardiol. 2006;21:295–304. doi: 10.1097/01.hco.0000231398.64362.94. [DOI] [PubMed] [Google Scholar]
  • 23.Blaustein MP, et al. How does salt retention raise blood pressure? Am J Physiol Regul Integr Comp Physiol. 2006;290:R514–R523. doi: 10.1152/ajpregu.00819.2005. [DOI] [PubMed] [Google Scholar]
  • 24.Schrier RW, Berl T. Nonosmolar factors affecting renal water excretion (first of two parts) N Engl J Med. 1975;292:81–88. doi: 10.1056/NEJM197501092920207. [DOI] [PubMed] [Google Scholar]
  • 25.Schrier RW. Body fluid volume regulation in health and disease: a unifying hypothesis. Ann Intern Med. 1990;113:155–159. doi: 10.7326/0003-4819-113-2-155. [DOI] [PubMed] [Google Scholar]
  • 26.de Wardener H, et al. Studies on the efferent mechanism of the sodium diuresis which follows the administration of intravenous saline in the dog. Clin Sci. 1961;21:249–258. [PubMed] [Google Scholar]
  • 27.Cort JH, Lichardus B. The natriuretic activity of jugular vein blood during carotid occlusion. Physiol Bohemoslov. 1963;12:497–501. [PubMed] [Google Scholar]
  • 28.Buckalew VM, Jr, et al. The effect of dialysates and ultrafiltrates of plasma of saline-loaded dogs on toad bladder sodium transport. J Clin Invest. 1970;49:926–935. doi: 10.1172/JCI106312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Schrier RW, et al. Absence of natriuretic response to acute hypotonic intravascular volume expansion in dogs. Clin Sci. 1968;34:57–72. [PubMed] [Google Scholar]
  • 30.Schrier RW, et al. Effect of isotonic saline infusion and acute haemorrhage on plasma oxytocin and vasopressin concentrations in dogs. Clin Sci. 1968;35:433–443. [PubMed] [Google Scholar]
  • 31.de Wardener HE, et al. Evidence for a hormone other than aldosterone which controls urinary sodium excretion. Adv Nephrol Necker Hosp. 1971;1:97–111. [PubMed] [Google Scholar]
  • 32.Kramer HJ, Gonick HC. Effect of extracellular volume expansion on renal Na-K-ATPase and cell metabolism. Nephron. 1974;12:281–296. doi: 10.1159/000180341. [DOI] [PubMed] [Google Scholar]
  • 33.Kaplan MA, et al. The effects of the natriuretic factor from uremic urine on sodium transport, water and electrolyte content, and pyruvate oxidation by the isolated toad bladder. J Clin Invest. 1974;53:1568–1577. doi: 10.1172/JCI107707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Bricker NS, et al. On the biology of sodium excretion: the search for a natriuretic hormone. Yale J Biol Med. 1975;48:293–303. [PMC free article] [PubMed] [Google Scholar]
  • 35.Gruber KA, et al. Endogenous digitalis-like substance in plasma of volume-expanded dogs. Nature. 1980;287:743–745. doi: 10.1038/287743a0. [DOI] [PubMed] [Google Scholar]
  • 36.Hamlyn JM, et al. A circulating inhibitor of (Na+ + K+)ATPase associated with essential hypertension. Nature. 1982;300:650–652. doi: 10.1038/300650a0. [DOI] [PubMed] [Google Scholar]
  • 37.Kojima I, et al. Involvement of endogenous digitalis-like substance in genesis of deoxycorticosterone-salt hypertension. Life Sci. 1982;30:1775–1781. doi: 10.1016/0024-3205(82)90313-7. [DOI] [PubMed] [Google Scholar]
  • 38.Goto A, et al. Physiology and pharmacology of endogenous digitalis-like factors. Pharmacol Rev. 1992;44:377–399. [PubMed] [Google Scholar]
  • 39.Bergdahl B, Molin L. Precision of digoxin radioimmunoassays and matrix effects: four kits compared. Clin Biochem. 1981;14:67–71. doi: 10.1016/s0009-9120(81)90704-9. [DOI] [PubMed] [Google Scholar]
  • 40.Pleasants RA, et al. Interference of digoxin-like immunoreactive substances with three digoxin immunoassays in patients with various degrees of renal function. Clin Pharm. 1986;5:810–816. [PubMed] [Google Scholar]
  • 41.Hauptman PJ, Kelly RA. Digitalis. Circulation. 1999;99:1265–1270. doi: 10.1161/01.cir.99.9.1265. [DOI] [PubMed] [Google Scholar]
  • 42.Schneider R, et al. Bovine adrenals contain, in addition to ouabain, a second inhibitor of the sodium pump. J Biol Chem. 1998;273:784–792. doi: 10.1074/jbc.273.2.784. [DOI] [PubMed] [Google Scholar]
  • 43.Kawamura A, et al. On the structure of endogenous ouabain. Proc Natl Acad Sci USA. 1999;96:6654–6659. doi: 10.1073/pnas.96.12.6654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Xie Z, Askari A. Na(+)/K(+)-ATPase as a signal transducer. Eur J Biochem. 2002;269:2434–2439. doi: 10.1046/j.1432-1033.2002.02910.x. [DOI] [PubMed] [Google Scholar]
  • 45.Liu L, et al. Role of caveolae in signal-transducing function of cardiac Na+/K+-ATPase. Am J Physiol Cell Physiol. 2003;284:C1550–C1560. doi: 10.1152/ajpcell.00555.2002. [DOI] [PubMed] [Google Scholar]
  • 46.Wang H, et al. Ouabain assembles signaling cascades through the caveolar Na+/K+-ATPase. J Biol Chem. 2004;279:17250–17259. doi: 10.1074/jbc.M313239200. [DOI] [PubMed] [Google Scholar]
  • 47.Liang M, et al. Identification of a pool of non-pumping Na/K-ATPase. J Biol Chem. 2007;282:10585–10593. doi: 10.1074/jbc.M609181200. [DOI] [PubMed] [Google Scholar]
  • 48.Manunta P, et al. Plasma ouabain-like factor during acute and chronic changes in sodium balance in essential hypertension. Hypertension. 2001;38:198–203. doi: 10.1161/01.hyp.38.2.198. [DOI] [PubMed] [Google Scholar]
  • 49.Balzan S, et al. Endogenous ouabain and acute salt loading in low-renin hypertension. Am J Hypertens. 2005;18:906–909. doi: 10.1016/j.amjhyper.2005.01.007. [DOI] [PubMed] [Google Scholar]
  • 50.Manunta P, et al. Salt intake and depletion increase circulating levels of endogenous ouabain in normal men. Am J Physiol Regul Integr Comp Physiol. 2006;290:R553–R559. doi: 10.1152/ajpregu.00648.2005. [DOI] [PubMed] [Google Scholar]
  • 51.Bagrov AY, et al. Endogenous marinobufagenin-like immunoreactive factor and Na,K-ATPase inhibition during voluntary hypoventilation. Hypertension. 1995;26:781–788. doi: 10.1161/01.hyp.26.5.781. [DOI] [PubMed] [Google Scholar]
  • 52.Lopatin DA, et al. Circulating bufodienolide and cardenolide sodium pump inhibitors in preeclampsia. J Hypertens. 1999;17:1179–1187. doi: 10.1097/00004872-199917080-00018. [DOI] [PubMed] [Google Scholar]
  • 53.Gonick HC, et al. Simultaneous measurement of marinobufagenin, ouabain and hypertension-associated protein in various disease states. Clin Exp Hypertens. 1998;20:617–627. doi: 10.3109/10641969809053240. [DOI] [PubMed] [Google Scholar]
  • 54.Fridman AI, et al. Marinobufagenin, an endogenous ligand of alpha-1 Na/K-ATPase, is a marker of congestive heart failure severity. J Hypertens. 2002;20:1189–1194. doi: 10.1097/00004872-200206000-00032. [DOI] [PubMed] [Google Scholar]
  • 55.Komiyama Y, et al. Identification of endogenous ouabain in culture supernatant of PC12 cells. J Hypertens. 2001;19:229–236. doi: 10.1097/00004872-200102000-00009. [DOI] [PubMed] [Google Scholar]
  • 56.Murrell JR, et al. Endogenous ouabain: upregulation of steroidogenic genes in hypertensive hypothalamus but not adrenal. Circulation. 2005;112:1301–1308. doi: 10.1161/CIRCULATIONAHA.105.554071. [DOI] [PubMed] [Google Scholar]
  • 57.el-Masri MA, et al. Human adrenal cells in culture produce both ouabain-like and dihydroouabain-like factors. Clin Chem. 2002;48:1720–1730. [PubMed] [Google Scholar]
  • 58.Laredo J, et al. Angiotensin II stimulates secretion of endogenous ouabain from bovine adrenocortical cells via angiotensin type 2 receptors. Hypertension. 1997;29:401–407. doi: 10.1161/01.hyp.29.1.401. [DOI] [PubMed] [Google Scholar]
  • 59.Shah JR, et al. Effects of angiotensin II on sodium potassium pumps, endogenous ouabain, and aldosterone in bovine zona glomerulosa cells. Hypertension. 1999;33:373–377. doi: 10.1161/01.hyp.33.1.373. [DOI] [PubMed] [Google Scholar]
  • 60.Goto A, et al. Isolation of a urinary digitalis-like factor indistinguishable from digoxin. Biochem Biophys Res Commun. 1990;173:1093–1101. doi: 10.1016/s0006-291x(05)80898-8. [DOI] [PubMed] [Google Scholar]
  • 61.Qazzaz HM, et al. Deglycosylated products of endogenous digoxin-like immunoreactive factor in mammalian tissue. J Biol Chem. 1996;271:8731–8737. doi: 10.1074/jbc.271.15.8731. [DOI] [PubMed] [Google Scholar]
  • 62.Huang BS, et al. Digoxin prevents ouabain and high salt intake-induced hypertension in rats with sinoaortic denervation. Hypertension. 1999;34:733–738. doi: 10.1161/01.hyp.34.4.733. [DOI] [PubMed] [Google Scholar]
  • 63.Goto A, et al. Digoxin-like immunoreactivity: is it still worth measuring? Life Sci. 1991;49:1667–1678. doi: 10.1016/0024-3205(91)90309-y. [DOI] [PubMed] [Google Scholar]
  • 64.Meyer K, Linde H. Collection of toad venoms and chemistry of the toad venom steroids. In: Bucherl W, Buckley E, editors. Venomous animals and their venoms. Academic Press; London: 1971. pp. 521–556. [Google Scholar]
  • 65.Chen KK, Kowarikowa A. Pharmacology and toxicology of toad venom. J Pharmacol Sci. 1967;56:1535–1542. doi: 10.1002/jps.2600561202. [DOI] [PubMed] [Google Scholar]
  • 66.Flier J, et al. Widespread occurrence in frogs and toads of skin compounds interacting with the ouabain site of Na+,K+-ATPase. Science. 1980;208:503–505. doi: 10.1126/science.6245447. [DOI] [PubMed] [Google Scholar]
  • 67.Lichtstein D, et al. Effect of salt acclimation on digitalis-like compounds in the toad. Biochim Biophys Acta. 1981;1073:65–68. doi: 10.1016/0304-4165(91)90183-h. [DOI] [PubMed] [Google Scholar]
  • 68.Kieval RS, et al. Cellular electrophysiologic effects of vertebrate digitalis-like substances. J Am Col Cardiol. 1988;11:637–643. doi: 10.1016/0735-1097(88)91543-4. [DOI] [PubMed] [Google Scholar]
  • 69.Goto A, et al. Immunoreactivity of endogenous digitalis-like factors. Biochem Pharmacol. 1991;41:1261–1263. doi: 10.1016/0006-2952(91)90668-u. [DOI] [PubMed] [Google Scholar]
  • 70.Numazawa S, et al. A cardiotonic steroid bufalin-like factor in human plasma induces leukemia cell differentiation. Leuk Res. 1995;19:945–953. doi: 10.1016/0145-2126(95)00081-x. [DOI] [PubMed] [Google Scholar]
  • 71.Oda M, et al. Determination of bufalin-like immunoreactivity in serum of humans and rats by time-resolved fluoroimmunoassay for using a monoclonal antibody. Life Sci. 2001;68:1107–1117. doi: 10.1016/s0024-3205(00)01013-4. [DOI] [PubMed] [Google Scholar]
  • 72.Sich B, et al. Pulse pressure correlates in humans with a proscillaridin A immunoreactive compound. Hypertension. 1996;27:1073–1078. doi: 10.1161/01.hyp.27.5.1073. [DOI] [PubMed] [Google Scholar]
  • 73.Hilton PJ, et al. An inhibitor of the sodium pump obtained from human placenta. Lancet. 1996;348:303–305. doi: 10.1016/s0140-6736(96)02257-x. [DOI] [PubMed] [Google Scholar]
  • 74.Bagrov AY, et al. Digitalis-Like and vasoconstrictor properties of endogenous digoxin-like factor from Bufo marinus toad. Eur J Pharmacol. 1993;234:165–172. doi: 10.1016/0014-2999(93)90950-m. [DOI] [PubMed] [Google Scholar]
  • 75.Bagrov AY, et al. Effects of two endogenous digitalis-like factors, ouabain and marinobufagenin in isolated rat aorta. Eur J Pharmacol. 1995;274:151–158. doi: 10.1016/0014-2999(94)00735-p. [DOI] [PubMed] [Google Scholar]
  • 76.Bagrov AY, et al. Endogenous marinobufagenin-like immunoreactive substance: a possible endogenous Na,K-ATPase inhibitor with vasoconstrictor activity. Am J Hypertens. 1996;9:982–990. doi: 10.1016/0895-7061(96)00148-3. [DOI] [PubMed] [Google Scholar]
  • 77.Ho CS, et al. Effect of carbidopa on the excretion of sodium, dopamine, and ouabain-like substance in the rat. Hypertension. 1997;30:1544–1548. doi: 10.1161/01.hyp.30.6.1544. [DOI] [PubMed] [Google Scholar]
  • 78.Butt AN, et al. Effect of high salt intake on plasma and tissue concentration of endogenous ouabain-like substance in the rat. Life Sci. 1997;61:2367–7233. doi: 10.1016/s0024-3205(97)00953-3. [DOI] [PubMed] [Google Scholar]
  • 79.Ludens JH, et al. Digitalis-like factor and ouabain-like compound in plasma of volume-expanded dogs. J Cardiovasc Pharmacol. 1993;22(suppl 2):S38–S41. doi: 10.1097/00005344-199322002-00014. [DOI] [PubMed] [Google Scholar]
  • 80.Bagrov AY, et al. Plasma marinobufagenin-like and ouabain-like immunorecativity during acute saline volume expansion in anesthetized dogs. Cardiovasc Res. 1996;206:296–305. [PubMed] [Google Scholar]
  • 81.Fedorova OV, et al. Interaction of high sodium chloride intake and psychosocial stress on endogenous ligands of the sodium pump and blood pressure in normotensive rats. Am J Physiol. 2001;281:R352–R358. doi: 10.1152/ajpregu.2001.281.1.R352. [DOI] [PubMed] [Google Scholar]
  • 82.Fedorova OV, et al. Differential effects of acute NaCl loading on endogenous ouabain-like and marinobufagenin-like ligands of the sodium pump in Dahl hypertensive rats. Circulation. 2000;102:3009–3014. doi: 10.1161/01.cir.102.24.3009. [DOI] [PubMed] [Google Scholar]
  • 83.Fedorova OV, et al. An endogenous ligand of α-1 sodium pump, marinobufagenin, is a novel mediator of sodium chloride dependent hypertension. Circulation. 2002;105:1122–1127. doi: 10.1161/hc0902.104710. [DOI] [PubMed] [Google Scholar]
  • 84.Anderson DE, et al. Endogenous sodium pump inhibitors and age-associated increases in salt sensitivity of blood pressure in normotensives. Am J Physiol Regul Integr Comp Physiol. 2008;294:R1248–R1254. doi: 10.1152/ajpregu.00782.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Ferrari P, et al. Rostafuroxin: an ouabain antagonist that corrects renal and vascular Na+-K+-ATPase alterations in ouabain and adducin-dependent hypertension. Am J Physiol Regul Integr Comp Physiol. 2006;290:R529–R535. doi: 10.1152/ajpregu.00518.2005. [DOI] [PubMed] [Google Scholar]
  • 86.Rossoni LV, et al. Ouabain-induced hypertension is accompanied by increases in endothelial vasodilator factors. Am J Physiol Heart Circ Physiol. 2002;283:H2110–H2118. doi: 10.1152/ajpheart.00454.2002. [DOI] [PubMed] [Google Scholar]
  • 87.Ferrandi M, et al. Organ hypertrophic signaling within caveolae membrane subdomains triggered by ouabain and antagonized by PST 2238. J Biol Chem. 2004;279:33306–33314. doi: 10.1074/jbc.M402187200. [DOI] [PubMed] [Google Scholar]
  • 88.Dostanic-Larson I, et al. The highly conserved cardiac glycoside binding site of Na,K-ATPase plays a role in blood pressure regulation. Proc Natl Acad Sci USA. 2005;102:15845–15850. doi: 10.1073/pnas.0507358102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Briones, et al. Alterations in structure and mechanics of resistance arteries from ouabain-induced hypertensive rats. Am J Physiol Heart Circ Physiol. 2006;291:H193–H201. doi: 10.1152/ajpheart.00802.2005. [DOI] [PubMed] [Google Scholar]
  • 90.Cheung WJ, et al. Central and peripheral renin-angiotensin systems in ouabain-induced hypertension. Am J Physiol Heart Circ Physiol. 2006;291:H624–H630. doi: 10.1152/ajpheart.01148.2005. [DOI] [PubMed] [Google Scholar]
  • 91.Rossoni LV, et al. Ouabain-induced hypertension enhances left ventricular contractility in rats. Life Sci. 2006;79:1537–1545. doi: 10.1016/j.lfs.2006.04.017. [DOI] [PubMed] [Google Scholar]
  • 92.Bianchi G, et al. Two point mutations within the adducin genes are involved in blood pressure variation. Proc Natl Acad Sci USA. 1994;91:3999–4003. doi: 10.1073/pnas.91.9.3999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Efendiev R, et al. Hypertension-linked mutation in the adducin alpha-subunit leads to higher AP2-mu2 phosphorylation and impaired Na+K+-ATPase trafficking in response to GPCR signals and intracellular sodium. Circ Res. 2004;95:1100–1108. doi: 10.1161/01.RES.0000149570.20845.89. [DOI] [PubMed] [Google Scholar]
  • 94.Zhang J, et al. Sodium pump α2 subunits control myogenic tone and blood pressure in mice. J Physiol. 2005;569:243–256. doi: 10.1113/jphysiol.2005.091801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Liu J, et al. Ouabain induces endocytosis of plasmalemmal Na/K-ATPase in LLC-PK1 cells by a clathrin-dependent mechanism. Kidney Int. 2004;66:227–241. doi: 10.1111/j.1523-1755.2004.00723.x. [DOI] [PubMed] [Google Scholar]
  • 96.Liu J, et al. Ouabain-induced endocytosis of the plasmalemmal Na/K-ATPase in LLC-PK1 cells requires caveolin-1. Kidney Int. 2005;67:1844–1854. doi: 10.1111/j.1523-1755.2005.00283.x. [DOI] [PubMed] [Google Scholar]
  • 97.Nguyen AN, et al. Ouabain binds with high affinity to the Na,K-ATPase in human polycystic kidney cells and induces extracellular signal-regulated kinase activation and cell proliferation. J Am Soc Nephrol. 2007;18:46–57. doi: 10.1681/ASN.2006010086. [DOI] [PubMed] [Google Scholar]
  • 98.Wang JG, et al. Salt, endogenous ouabain and blood pressure interactions in the general population. J Hypertens. 2003;21:1475–1481. doi: 10.1097/00004872-200308000-00010. [DOI] [PubMed] [Google Scholar]
  • 99.Ferrari P, et al. PST 2238: a new antihypertensive compound that modulates Na,K-ATPase in genetic hypertension. J Pharmacol Exp Ther. 1999;288:1074–1083. [PubMed] [Google Scholar]
  • 100.Huang BS, Leenen FH. Brain ‘ouabain’ and angiotensin II in salt-sensitive hypertension in spontaneously hypertensive rats. Hypertension. 1996;28:1005–1012. doi: 10.1161/01.hyp.28.6.1005. [DOI] [PubMed] [Google Scholar]
  • 101.Huang BS, Leenen FH. Both brain angiotensin II and ‘ouabain’ contribute to sympathoexcitation and hypertension in Dahl S rats on high salt intake. Hypertension. 1998;32:1028–1033. doi: 10.1161/01.hyp.32.6.1028. [DOI] [PubMed] [Google Scholar]
  • 102.Fedorova OV, et al. Intrahippocampal microinjection of an exquisitely low dose of ouabain mimics NaCl loading and stimulates a bufadienolide Na/K-ATPase inhibitor. J Hypertens. 2007;25:1834–1844. doi: 10.1097/HJH.0b013e328200497a. [DOI] [PubMed] [Google Scholar]
  • 103.Fedorova OV, et al. Brain ouabain stimulates peripheral marinobufagenin via angiotensin II signalling in NaCl loaded Dahl-S rats. J Hypertens. 2005;23:1515–1523. doi: 10.1097/01.hjh.0000174969.79836.8b. [DOI] [PubMed] [Google Scholar]
  • 104.Takahashi H, et al. Centrally-induced vasopressor responses to sodium-potassium adenosine triphosphatase inhibitor, ouabain, may be mediated via angiotensin II in the anteroventral third ventricle in the brain. Jpn Circ J. 1984;48:1243–1250. doi: 10.1253/jcj.48.1243. [DOI] [PubMed] [Google Scholar]
  • 105.Huang BS, Leenen FHH. Sympathoexcitatory and pressor responses to increased brain sodium and ouabain are mediated via brain ANGII. Am J Physiol. 1996;270:H275–H280. doi: 10.1152/ajpheart.1996.270.1.H275. [DOI] [PubMed] [Google Scholar]
  • 106.Huang BS, et al. Increases in CSF [Na+] precede the increases in blood pressure in Dahl S rats and SHR on a high-salt diet. Am J Physiol Heart Circ Physiol. 2004;287:H1160–H1166. doi: 10.1152/ajpheart.00126.2004. [DOI] [PubMed] [Google Scholar]
  • 107.Wang H, Leenen FH. Brain sodium channels and central sodium-induced increases in brain ouabain-like compound and blood pressure. J Hypertens. 2003;21:1519–1524. doi: 10.1097/00004872-200308000-00016. [DOI] [PubMed] [Google Scholar]
  • 108.Amin MS, et al. Distribution of epithelial sodium channels and mineralocorticoid receptors in cardiovascular regulatory centers in rat brain. Am J Physiol Regul Integr Comp Physiol. 2005;289:R1787–R1797. doi: 10.1152/ajpregu.00063.2005. [DOI] [PubMed] [Google Scholar]
  • 109.Orlov SN, Mongin AA. Salt-sensing mechanisms in blood pressure regulation and hypertension. Am J Physiol Heart Circ Physiol. 2007;293:H2039–H2053. doi: 10.1152/ajpheart.00325.2007. [DOI] [PubMed] [Google Scholar]
  • 110.Fedorova OV, et al. Endogenous marinobufagenin-like factor in acute plasma volume expansion. Clin Exp Hypertens. 1998;20:581–591. doi: 10.3109/10641969809053236. [DOI] [PubMed] [Google Scholar]
  • 111.Periyasamy SM, et al. Salt loading induces redistribution of the plasmalemmal Na/K-ATPase in proximal tubule cells. Kidney Int. 2005;67:1868–1877. doi: 10.1111/j.1523-1755.2005.00285.x. [DOI] [PubMed] [Google Scholar]
  • 112.Fedorova OV, Bagrov AY. Inhibition of Na/K ATPase from rat aorta by two Na/K pump inhibitors, ouabain and marinobufagenin: evidence of interaction with different alpha-subunit isoforms. Am J Hypertens. 1997;10:929–935. doi: 10.1016/s0895-7061(97)00096-4. [DOI] [PubMed] [Google Scholar]
  • 113.Dahl LK, et al. Humoral transmission of hypertension: evidence from parabiosis. Circ Res. 1969;24(suppl):S21–S33. [PubMed] [Google Scholar]
  • 114.Liu J, Shapiro JI. Regulation of sodium pump endocytosis by cardiotonic steroids: Molecular mechanisms and physiological implications. Pathophysiology. 2007;14:171–181. doi: 10.1016/j.pathophys.2007.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Liu J, et al. Effects of cardiac glycosides on sodium pump expression and function in LLC-PK1 and MDCK cells. Kidney Int. 2002;62:2118–2125. doi: 10.1046/j.1523-1755.2002.00672.x. [DOI] [PubMed] [Google Scholar]
  • 116.Oweis S, et al. Cardiac glycoside downregulates NHE3 activity and expression in LLC-PK1 cells. Am J Physiol Renal Physiol. 2006;290:F997–F1008. doi: 10.1152/ajprenal.00322.2005. [DOI] [PubMed] [Google Scholar]
  • 117.Cai H, et al. Regulation of apical NHE3 trafficking by ouabain-induced activation of basolateral Na/K-ATPase receptor complex. Am J Physiol Cell Physiol. 2008;294:C555–C563. doi: 10.1152/ajpcell.00475.2007. [DOI] [PubMed] [Google Scholar]
  • 118.Schrier RW, Abraham WT. Hormones and hemodynamics in heart failure. N Engl J Med. 1999;341:577–585. doi: 10.1056/NEJM199908193410806. [DOI] [PubMed] [Google Scholar]
  • 119.Schreiber V, et al. Digoxin-like immunoreactivity in the serum of rats with cardiac overload. J Mol Cell Cardiol. 1981;13:107–110. doi: 10.1016/0022-2828(81)90232-7. [DOI] [PubMed] [Google Scholar]
  • 120.Morise T, et al. Biological activity of partially purified digitalis-like substance and Na-K-ATPase inhibitor in rats. Jpn Circ J. 1988;52:1309–1316. doi: 10.1253/jcj.52.1309. [DOI] [PubMed] [Google Scholar]
  • 121.Liu ZQ, et al. Intra-cellular electrolyte changes and levels of endogenous digoxin-like substance within the plasma in patients with congestive heart failure. Int J Cardiol. 1990;27:47–53. doi: 10.1016/0167-5273(90)90190-g. [DOI] [PubMed] [Google Scholar]
  • 122.Gottlieb SS, et al. Elevated concentrations of endogenous ouabain in patients with congestive heart failure. Circulation. 1992;86:420–425. doi: 10.1161/01.cir.86.2.420. [DOI] [PubMed] [Google Scholar]
  • 123.Manunta P, et al. Left ventricular mass, stroke volume, and ouabain-like factor in essential hypertension. Hypertension. 1999;34:450–456. doi: 10.1161/01.hyp.34.3.450. [DOI] [PubMed] [Google Scholar]
  • 124.Pierdomenico SD, et al. Endogenous ouabain and hemodynamic and left ventricular geometric patterns in essential hypertension. Am J Hypertens. 2001;14:44–50. doi: 10.1016/s0895-7061(00)01225-5. [DOI] [PubMed] [Google Scholar]
  • 125.Balzan S, et al. Increased circulating levels of ouabain-like factor in patients with asymptomatic left ventricular dysfunction. Eur J Heart Fail. 2001;3:165–171. doi: 10.1016/s1388-9842(00)00132-x. [DOI] [PubMed] [Google Scholar]
  • 126.Pitzalis MV, et al. Independent and incremental prognostic value of endogenous ouabain in idiopathic dilated cardiomyopathy. Eur J Heart Fail. 2006;8:179–186. doi: 10.1016/j.ejheart.2005.07.010. [DOI] [PubMed] [Google Scholar]
  • 127.Stella P, et al. Endogenous ouabain and cardiomyopathy in dialysis patients. J Intern Med. 2008;263:274–280. doi: 10.1111/j.1365-2796.2007.01883.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Fedorova OV, et al. Coordinated shifts in Na/K-ATPase isoforms and their endogenous ligands during cardiac hypertrophy and failure in NaCl-sensitive hypertension. J Hypertens. 2004;22:389–397. doi: 10.1097/00004872-200402000-00025. [DOI] [PubMed] [Google Scholar]
  • 129.Akimova OA, et al. Cardiotonic steroids differentially affect intracellular Na+ and [Na+]i/[K+]i-independent signaling in C7-MDCK cells. J Biol Chem. 2005;280:832–839. doi: 10.1074/jbc.M411011200. [DOI] [PubMed] [Google Scholar]
  • 130.Neuss M, et al. Apoptosis in cardiac disease-what is it-how does it occur. Cardiovasc Drugs Ther. 2001;15:507–523. doi: 10.1023/a:1013715704835. [DOI] [PubMed] [Google Scholar]
  • 131.Mohmand B, et al. Uremic cardiomyopathy: role of circulating digitalis like substances. Front Biosci. 2005;10:2036–2044. doi: 10.2741/1679. [DOI] [PubMed] [Google Scholar]
  • 132.Middleton RJ, et al. Left ventricular hypertrophy in the renal patient. J Am Soc Nephrol. 2001;12:1079–1084. doi: 10.1681/ASN.V1251079. [DOI] [PubMed] [Google Scholar]
  • 133.Kennedy DJ, et al. Effect of chronic renal failure on cardiac contractile function, calcium cycling, and gene expression of proteins important for calcium homeostasis in the rat. J Am Soc Nephrol. 2003;14:90–97. doi: 10.1097/01.asn.0000037403.95126.03. [DOI] [PubMed] [Google Scholar]
  • 134.Kennedy DJ, et al. Partial nephrectomy as a model for uremic cardiomyopathy in the mouse. Am J Physiol Renal Physiol. 2008;294:F450–F454. doi: 10.1152/ajprenal.00472.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Elkareh J, et al. Marinobufagenin stimulates fibroblast collagen production and causes fibrosis in experimental uremic cardiomyopathy. Hypertension. 2007;49:215–224. doi: 10.1161/01.HYP.0000252409.36927.05. [DOI] [PubMed] [Google Scholar]
  • 136.Kennedy DJ, et al. Central role for the cardiotonic steroid marinobufagenin in the pathogenesis of experimental uremic cardiomyopathy. Hypertension. 2006;47:488–495. doi: 10.1161/01.HYP.0000202594.82271.92. [DOI] [PubMed] [Google Scholar]
  • 137.London GM. Left ventricular alterations and end-stage renal disease. Nephrol Dial Transplant. 2002;17(suppl 1):S29–S36. doi: 10.1093/ndt/17.suppl_1.29. [DOI] [PubMed] [Google Scholar]
  • 138.Gallery ED, et al. Plasma volume contraction: a significant factor in both pregnancy-associated hypertension (pre-eclampsia) and chronic hypertension in pregnancy. Q J Med. 1979;192:593–602. [PubMed] [Google Scholar]
  • 139.Masilamani S, Baylis C. Pregnant rats are refractory to the natriuretic actions of ANP. Am J Physiol. 1994;267:R1611–R1616. doi: 10.1152/ajpregu.1994.267.6.R1611. [DOI] [PubMed] [Google Scholar]
  • 140.Graves SW. The possible role of digitalislike factors in pregnancy-induced hypertension. Hypertension. 1987;10:I84–I86. doi: 10.1161/01.hyp.10.5_pt_2.i84. [DOI] [PubMed] [Google Scholar]
  • 141.Graves SW, et al. Endogenous digoxin-immunoreactive substance in human pregnancies. J Clin Endocrinol Metab. 1984;58:748–751. doi: 10.1210/jcem-58-4-748. [DOI] [PubMed] [Google Scholar]
  • 142.Averina IV, et al. Endogenous Na/K-ATPase inhibitors in patients with preeclampsia. Cell Mol Biol (Noisy-le-grand) 2006;52:19–23. [PubMed] [Google Scholar]
  • 143.Goodlin RC. Antidigoxin antibodies in eclampsia. N Engl J Med. 1988;318:518–519. doi: 10.1056/NEJM198802253180815. [DOI] [PubMed] [Google Scholar]
  • 144.Adair CD, et al. Elevated endoxin-like factor complicating a multifetal second trimester pregnancy: treatment with digoxin-binding immunoglobulin. Am J Nephrol. 1996;16:529–531. doi: 10.1159/000169054. [DOI] [PubMed] [Google Scholar]
  • 145.Adair D, et al. Effects of Fab digoxin-specific antibodies on mean arterial pressure in severe preeclampsia [abstract] Am J Hypertens. 1997;10:11A. [Google Scholar]
  • 146.Di Grande A, et al. Release of a substance from the human placenta having digoxin-like immunoreactivity. Clin Exp Pharmacol Physiol. 1993;20:603–607. doi: 10.1111/j.1440-1681.1993.tb01747.x. [DOI] [PubMed] [Google Scholar]
  • 147.Amler E, et al. Human hypertensive placenta contains an increased amount of Na,K-ATPase with higher affinity for cardiac glycosides. Cell Biol Int. 1994;18:723–727. doi: 10.1006/cbir.1994.1101. [DOI] [PubMed] [Google Scholar]
  • 148.Dmitrieva RI, et al. Mammalian bufadienolide is synthesized from cholesterol in the adrenal cortex by a pathway that is independent of cholesterol side-chain cleavage. Hypertension. 2000;36:442–448. doi: 10.1161/01.hyp.36.3.442. [DOI] [PubMed] [Google Scholar]
  • 149.Fedorova OV, et al. Antibody to marinobufagenin lowers blood pressure in pregnant rats on a high NaCl intake. J Hypertens. 2005;23:835–842. doi: 10.1097/01.hjh.0000163153.27954.33. [DOI] [PubMed] [Google Scholar]
  • 150.Vu HV, et al. Involvement of marinobufagenin in a rat model of human preeclampsia. Am J Nephrol. 2005;25:520–528. doi: 10.1159/000088461. [DOI] [PubMed] [Google Scholar]
  • 151.LaMarca HL, et al. Marinobufagenin impairs first trimester cytotrophoblast differentiation. Placenta. 2006;27:984–988. doi: 10.1016/j.placenta.2005.12.004. [DOI] [PubMed] [Google Scholar]
  • 152.Weidemann H, et al. Diverse effects of stress and additional adrenocorticotropic hormone on digitalis-like compounds in normal and nude mice. J Neuroendocrinol. 2004;16:458–463. doi: 10.1111/j.1365-2826.2004.01181.x. [DOI] [PubMed] [Google Scholar]
  • 153.Bauer N, et al. Ouabain-like compound changes rapidly on physical exercise in humans and dogs: effects of beta-blockade and angiotensin-converting enzyme inhibition. Hypertension. 2005;45:1024–1028. doi: 10.1161/01.HYP.0000165024.47728.f7. [DOI] [PubMed] [Google Scholar]
  • 154.Bagrov AY, et al. Effect of antidigoxin antibody on myocardial Na,K-pump activity and of endogenous digoxin-like factor in acute myocardial ischemia in rats. Cardiovasc Res. 1993;27:1045–1050. doi: 10.1093/cvr/27.6.1045. [DOI] [PubMed] [Google Scholar]
  • 155.Grider G, et al. Endogenous digoxin-like immunoreactive factor (DLIF) serum concentrations are decreased in manic bipolar patients compared to normal controls. J Affect Disord. 1999;54:261–267. doi: 10.1016/s0165-0327(98)00208-0. [DOI] [PubMed] [Google Scholar]
  • 156.Goldstein I, et al. Involvement of Na(+), K(+)-ATPase and endogenous digitalis-like compounds in depressive disorders. Biol Psychiatry. 2006;60:491–499. doi: 10.1016/j.biopsych.2005.12.021. [DOI] [PubMed] [Google Scholar]
  • 157.Bagrov YY, et al. Involvement of endogenous digitalis-like factors in voluntary selection of alcohol by rats. Life Sci. 1999;64:PL219–PL225. doi: 10.1016/s0024-3205(99)00131-9. [DOI] [PubMed] [Google Scholar]
  • 158.Clerico A, Giampietro O. Is the endogenous digitalis-like factor the link between hypertension and metabolic disorders as diabetes mellitus, obesity and acromegaly? Clin Physiol Biochem. 1990;8:153–168. [PubMed] [Google Scholar]
  • 159.Chen S, et al. Role of digitalis-like substance in the hypertension of streptozotocin-induced diabetes in reduced renal mass rats. Am J Hypertens. 1993;6:397–406. doi: 10.1093/ajh/6.5.397. [DOI] [PubMed] [Google Scholar]
  • 160.Bagrov YY, et al. Endogenous digitalis-like ligands and Na/K-ATPase inhibition in experimental diabetes mellitus. Front Biosci. 2005;10:2257–2262. doi: 10.2741/1695. [DOI] [PubMed] [Google Scholar]
  • 161.Carroll JS, et al. Digitalis-like factor response to hyperinsulinemia accompanying a euglycemic hyperinsulinemic clamp or oral glucose tolerance test. Life Sci. 2001;69:829–837. doi: 10.1016/s0024-3205(01)01176-6. [DOI] [PubMed] [Google Scholar]
  • 162.Weidemann H. Na/K-ATPase, endogenous digitalis like compounds and cancer development—a hypothesis. Front Biosci. 2005;10:2165–2176. doi: 10.2741/1688. [DOI] [PubMed] [Google Scholar]
  • 163.Mijatovic T, et al. Cardiotonic steroids on the road to anti-cancer therapy. Biochim Biophys Acta. 2007;1776:32–57. doi: 10.1016/j.bbcan.2007.06.002. [DOI] [PubMed] [Google Scholar]
  • 164.Ferrandi M, et al. Ouabain antagonists as antihypertensive agents. Curr Pharm Des. 2005;11:3301–3305. doi: 10.2174/138161205774424735. [DOI] [PubMed] [Google Scholar]
  • 165.ClinicalTrials.gov [accessed 12 March 2008];(online 2006) Efficacy of Rostafuroxin in the treatment of essential hypertension. http://clinicaltrials.gov/ct2/show/NCT00415038.
  • 166.Finotti P, Palatini P. Canrenone as a partial agonist at the digitalis receptor site of sodium-potassium-activated adenosine triphosphatase. J Pharmacol Exp Ther. 1981;217:784–790. [PubMed] [Google Scholar]
  • 167.de Mendonça M, et al. Antihypertensive effect of canrenone in a model where endogenous ouabain-like factors are present. J Cardiovasc Pharmacol. 1988;11:75–83. doi: 10.1097/00005344-198801000-00012. [DOI] [PubMed] [Google Scholar]
  • 168.Waldorff S, Buch J. Canrenoate—a spironolactone metabolite: acute cardiac effects in digitalized patients. Eur J Cardiol. 1979;10:143–149. [PubMed] [Google Scholar]
  • 169.ClinicalTrials.gov [accessed 12 March 2008];(online 2005) Efficacy study of Digibind for treatment of severe preeclampsia. http://clinicaltrials.gov/show/NCT00158743.
  • 170.Menezes JC, et al. Digoxin antibody decreases natriuresis and diuresis in cerebral hemorrhage. Intensive Care Med. 2003;29:2291–2296. doi: 10.1007/s00134-003-1955-0. [DOI] [PubMed] [Google Scholar]
  • 171.Fedorova OV, et al. Reduction in myocardial PKC β2, Na/K-ATPase sensitivity to marinobufagenin and blood pressure in response to cicletanine. Hypertension. 2003;41:505–511. doi: 10.1161/01.HYP.0000053446.43894.9F. [DOI] [PubMed] [Google Scholar]
  • 172.Fedorova OV, et al. ANP differentially modulates marinobufagenin-induced sodium pump inhibition in kidney and aorta. Hypertension. 2006;48:1160–1168. doi: 10.1161/01.HYP.0000248129.20524.d0. [DOI] [PubMed] [Google Scholar]

RESOURCES