Skip to main content
Erschienen in: Cardiovascular Drugs and Therapy 1/2013

Open Access 01.02.2013 | REVIEW ARTICLE

The Late Na+ Current - Origin and Pathophysiological Relevance

verfasst von: Antonio Zaza, Marcella Rocchetti

Erschienen in: Cardiovascular Drugs and Therapy | Ausgabe 1/2013

In excitable tissues, voltage-dependent Na+ current (INa) is best known for supporting autoregenerative depolarization and impulse propagation. Its transient component (INaT), which is large and terminated within several milliseconds by channel inactivation, fulfils this role. Nevertheless, INa also includes a smaller sustained component, i.e. one persisting during prolonged membrane depolarization, which contributes to repolarization course. Sustained INa implies slow or incomplete inactivation of a proportion of the Na+ channels activated during the action potential upstroke. Several mechanisms may underlie this phenomenon and contribute to arrhythmogenesis in different conditions.

Sustained Na+ Currents

Window Currents

Truly steady-state currents may flow through otherwise inactivating channels if membrane potential is maintained in a restricted range (“window”). These currents are commonly referred to as “window currents”. The window corresponds to the region of overlap between “activation” and “inactivation” curves, in which the probabilities of channels to be open and not inactivated are both greater than 0 at steady-state. A window is normally present in cardiac myocytes for ICaL and, to a lesser extent for INa (Fig. 1), but it may be enhanced, or shifted to different potentials, by channel mutations (e.g. the Nav1.5 ΔKPQ mutation) [1].
When, during repolarization, membrane potential moves into the “window”, a fraction of channels may recover from inactivation and immediately reactivate. The resulting depolarization supports autoregenerative activation which, albeit concerning a small fraction of total channels, may substantially distort membrane potential course (early afterdepolarizations, EADs) and even trigger propagated activity [2, 3]. The INa window is narrow and more negative than the plateau phase in normal ventricular myocytes; thus, in the latter ICaL window more likely accounts for EADs generation [4] (Fig. 1). INa window, somewhat more prominent in normal Purkinje myocytes [5, 6], may be widened in all cell types by Na+ channel mutations.
A further mechanism of INa enhancement during repolarization is provided by an increase in the rate of channel recovery from inactivation relative to deactivation one. This mechanism, observed in some NaV1.5 mutations, is referred to as “reactivation under non-equilibrium conditions” [7]. This is because the excess Na+ current can be observed only during dynamic changes of membrane potential (non-equilibrium). This is not a proper steady-state component and, notably, it can escape detection by the standard “voltage step” clamp protocols used to assess the functional phenotype of channel mutations.

Late Na+ Current (INaL)

Figure 2a shows that sustained INa, identified as current sensitive to blockade by tetrodotoxin (TTX), exists also at potentials positive to the INa window (−65 to −50 mV; shown in in Fig. 1a for the same cell type). This current is provided by a component named “late” Na+ current (INaL). INaL, much smaller than INaT in normal ventricular myocytes, may be enhanced by more than 3-fold under pathological conditions [8]. Single-channel analysis reveals that the channel gating underlying INaL is complex and, in addition to late “scattered” openings, it includes a “burst mode” not observed during the transient component of INa (INaT) [9, 10]. This led to hypothesize that INaL might be carried by channel variants other than the one prevailing in cardiac myocytes (NaV1.5), whose expression would be enhanced under pathological conditions (a contribution of NaV1.8 channels to INaL in mouse and rabbit cardiomyocytes has been indeed reported [11]). However, INaL with all its gating modes, can be detected by expression of Nav1.5 channels in cell lines (Fig. 2b), where no other Na+ channel isoforms are present [10, 12]. This proves that INaL can be carried by the same molecular entity accounting for INaT; according to this view, INaL enhancement reflects a gating abnormality. Observations crucial for the development of this interpretation came from single channel analysis of a mutation (ΔKPQ) associated with LQT3 syndrome [13]. The mutant channel had increased probability of burst openings during sustained depolarization, a behaviour interpreted as instability of the inactivated state [1, 13]. The interpretation of INaL enhancement as a gating abnormality of Nav1.5 channels may hold true also for acquired conditions, as proven in the case of heart failure [1416]. Nevertheless, we cannot rule out that changes in isoform expression may contribute in some of the diverse conditions in which INaL enhancement occurs.
Because of recovery from inactivation upon repolarization takes some time, INaT availability depends on the interval between excitations. This is true also for INaL, thus conferring to this current “reverse” rate-dependency [17], which is partial at physiological rates (approx 50 % reduction from quiescence to 120 b/min) [18].
Mutations prolonging action potential duration (APD) cause enhancement of plateau Na+ current through one or more of the mechanisms described above. For instance, all the above (widened voltage window, inactivation instability and accelerated recovery) may be operative in ΔKPQ mutants [1], but only non-equilibrium reactivation appears to account for the APD prolonging effect observed with the Il768V mutant [7].

Conditions and Mechanisms of INaL Enhancement

Although the interest in INaL pathophysiological role was mostly triggered by identification of Na+ channel mutations leading to arrhythmogenic QT prolongation [1], secondary INaL enhancement occurs in association to a surprisingly large number of common disease states (reviewed in [19]), including cardiac hypertrophy/failure and ischemia, not necessarily related to each other in terms of primary pathogenesis. Such a pattern suggests that INaL enhancement may be a common response to cell stress/dysfunction. Accordingly, in cardiac myocytes, INaL is enhanced by reactive oxygen species (ROS) [20], whose generation is generally associated to cell distress. The question of whether ischemia can enhance INaL can be answered only indirectly, because membrane currents can be recorded only in isolated myocytes, which cannot be subjected to ischemia. Nevertheless, INaL is enhanced by myocyte exposure to hypoxia [21] or ischemic metabolites [22] and INaL blockade effectively prevents ischemia/reperfusion damage in the intact heart [23].
The search of a common mechanism, mediating INaL enhancement by cellular stress of heterogeneous etiology, has highlighted the role of Ca2+-calmodulin kinase (CaMKIIδ) activation [24]. This is a Ca2+- and ROS-activated cytosolic enzyme which may, among other targets, phosphorylates NaV1.5 channels. Converging evidence indicates that CaMKIIδ overexpression (or activation) enhances INaL and generates cardiac abnormalities that are reversed by INaL blockade [25]. Upstream components of the CaMKII activation pathway, i.e. calmodulin (CaM) and Ca2+ itself, have also been shown to enhance INaL directly, with differences between normal and failing myocytes [26]. While this does not rule out further modes of INaL enhancement (see ref [27]. for review), CaMKIIδ activation is particularly relevant because its relation with INaL may set up a vicious feed-back loop (Fig. 3), very likely to contribute to evolution of cell dysfunction and damage in response to stress. Whereas CaMKII inhibition is not available for therapeutic use yet, INaL blockade may break such a vicious loop [25].
To summarize, except for the case of primary (genetic) Na+ channel defects, INaL enhancement might be viewed as a generic response to cellular stress, that is secondary in origin, but has a pivotal role in mediating functional derangements and disease progression.
INaL enhancement can also result from abnormalities in proteins other than the Na+ channel itself, but interacting with it to form macromolecular complexes [28, 29]. For instance, mutations of scaffold and adaptor proteins, as ankyrin-B [30] and caveolin-3 [31] respectively, have been recently identified in LQT3 patients. A detailed review of the factors potentially contributing to INaL enhancement in heart failure has been provided by Matsev et al. [27].

INaL Impact on Electrical Activity and Ionic Homeostasis

INaL directly affects electrical activity (it promotes depolarization or counters repolarization) and provides a route of Na+ influx. Considering the role of Na+ gradient in transmembrane transport, the latter may affect cellular solute homeostasis, thus generating a host of indirect consequences of physiological and pathophysiological relevance.

Electrophysiological Effects

INaL flows during repolarization and may directly affect its course. In normal canine ventricles INaL is differentially expressed across the wall (M-cells and Purkinje cells > subendocardial cells > subepicardial cells) [17, 32, 33]. Thus, it is conceivably a player in the physiological transmural repolarization gradient and in its rate-dependency in the dog (APD restitution) [34]. In normal ventricular myocytes, INaL inhibition by ranolazine (INa blocker with selectivity for INaL vs INaT) causes negligible APD changes. On the other hand, the remarkable effects of IKr blockade on APD, on its rate-depencency and, most importantly, on repolarization stability are all reversed by ranolazine [18, 35, 36]. These apparently contrasting findings may be reconciled by considering that ranolazine also partially blocks IKr [37]. Under basal conditions, this may offset the effect of INaL inhibition on APD; in turn, concomitant INaL inhibition limits the effect of IKr inhibition, thus preventing repolarization instability. If this interpretation is correct, we can conclude that INaL and IKr are physiologically in balance during normal repolarization; whenever this balance is altered, by either INaL enhancement or IKr blockade, repolarization stability is compromised. An extreme example of this condition is advanced heart failure, in which INaL enhancement and IKr downregulation coexist and are associated with dramatic repolarization instability [15]. Although the concept of IKr - INaL balance is valid in a broad sense, the effects of IKr blockade and INaL enhancement on action potential contour are not identical, an observation which may have its counterpart in the differences of clinical presentation between the LQT2 (IKr deficiency) and LQT3 (INaL enhancement) syndromes [38].
The direct contribution of INaL to repolarization course provides a first powerful mechanism linking arrhythmogenesis to INaL enhancement. Nevertheless, the latter may also facilitate arrhythmias through Ca2+ handling abnormalities (see below) and it is difficult to establish which mechanism prevails in a specific condition. The mutual interplay between Ca2+ handling and repolarization course [39] may actually make this distinction pointless.
A role of INaL in arrhythmogenesis is often inferred from the antiarrhythmic effect of its selective blocker ranolazine. While this may be considered legitimate in many cases, there are exceptions due to specificities of drug action. The best example is ranolazine efficacy on atrial arrhythmias, to which mechanisms other than INaL inhibition may also contribute [40].

Effects on Ionic Homeostasis

INa represents the main source of Na+ entry during the cardiac cycle. Abeit INaL amplitude is normally 1/1000 of that of INaT [10], INaL persists throughout repolarization; as a result, INaT and INaL may similarly contribute to Na+ influx during a cardiac cycle [41]. Nevertheless, INaL inhibition (by TTX or ranolazine) marginally affects Ca2+ cycling [42] and contractility [43] in normal hearts, thus suggesting that the attending changes in Na+ influx are effectively buffered by matching changes in Na+ extrusion. However, cellular homeostasis can be compromised by the marked increase in Na+ influx resulting from pathological INaL enhancement.
Na+ is normally extruded from the cell by the ATP powered Na+/K+ pump. Therefore excess Na+ influx, even when successfully buffered, may increase ATP consumption. Furthermore, if influx exceeds the maximal extrusion rate, Na+ accumulates in the cytosol, thus partially dissipating its transmembrane gradient. Because the latter energizes many secondary membrane transport mechanisms, most importantly the Na+/Ca2+ exchanger (NCX) and the Na+/H+ exchanger (NHE), a pivotal consequence of INaL enhancement is perturbed homeostasis of intracellular Ca2+ and H+ (Fig. 3).
NCX is the main mechanism of Ca2+ extrusion from the cell. Increased cytosolic Na+ moves its electrochemical equilibrium potential in the negative direction, thus reducing the driving force for its forward operation during diastole and possibly reversing the direction of transport during systole (i.e. Ca2+ entry through NCX). The resulting increase in intracellular Ca2+ may re-establish NCX driving force, but the system equilibrium is now moved to higher cytosolic Ca2+ levels. Under conditions of INaL enhancement (e.g heart failure) NCX expression may be upregulated [44] and, provided that a driving force still exists, this may increase Ca2+ transport rate. However, the effect of this change in sustaining Ca2+ extrusion can only be partial, because it vanishes as NCX electrochemical equilibrium is approached. Accordingly, unless maximal Na+/K+ pump transport rate is also incremented, an increase in total cellular Ca2+ content is a necessary consequence of INaL enhancement. A further aspect of interest is the distribution of such an increment between subcellular compartments. Stimulation of Ca2+ uptake by the sarcoplasmic reticulum (SR) (by SERCA2) is a central component of physiological stimuli meant to increase cell Ca2+ content (e.g. by β-adrenergic activation). This ensures that most of the gain concerns SR luminal Ca2+, which results in larger amplitude of Ca2+ transient (larger developed force) and lower diastolic Ca2+ (accelerated relaxation). This is not the case for INaL enhancement, in which the Ca2+ increment concerns primarily the cytosol, which may only secondarily increase Ca2+ in the SR. Although the latter may increase maximal developed force, this is expectedly associated with higher diastolic Ca2+. The consequences of cytosolic Ca2+ being persistently elevated are multiple and of pathophysiological relevance.
Opening of SR Ca2+ release channels (RyRs) is directly triggered by cytosolic Ca2+, a process facilitated by high Ca2+ in the SR lumen [45]. Increased cytosolic Ca2+ also activates CaMKII-dependent RyRs phosphorylation, a further mechanism of RyRs facilitation (see below, Fig. 3). It is therefore unsurprising that INaL enhancement may lead to facilitation of diastolic “Ca2+ waves” and the resulting electrical disturbances (delayed afterdepolarizations, DADs) [46]. This represents an important arrhythmogenic mechanism, probably the one prevailing under conditions of altered Ca2+ handling (e.g. heart failure).
With persistently elevated cytosolic Ca2+, diastolic function may be hampered by delayed and incomplete sarcomere relaxation. A contribution of INaL to diastolic dysfunction has been demonstrated in various experimental models of heart failure [25, 42] and ranolazine improved diastolic relaxation in human ischemic heart disease [47]. In addition to its direct hemodynamic impact, diastolic dysfunction may limit coronary flow by extrinsic compression of intramural vessels. Significance of this effect is indirectly demonstrated by the ability of ranolazine to improve myocardial perfusion in the setting of coronary artery disease (likely INaL enhancement) [48]. Indeed, being ranolazine devoid of significant direct vasodilator effects, the perfusion improvement is likely to result from accelerated diastolic relaxation, a well known factor in modulation of coronary flow [49].
Abnormally elevated cytosolic Ca2+ may also activate a number of signalling pathways involved in modulation of cell function and, in the long run, structure and fate. CaMKIIδ activation, is at the same time, a cause (see above) and a consequence [50] of INaL enhancement. Phosphorylation of SR Ca2+ release channels (RyRs) by this kinase increases their open probability [51], ultimately leading to destabilization of the Ca2+ store. This accounts for the facilitation of spontaneous Ca2+ release events and delayed afterpotentials (DADs, their arrhythmogenic electrical consequence) induced by either INaL enhancement or CaMKIIδ overexpression [52] and suppressed in both cases by INaL blockade [25]. Calcineurin, a cytosolic Ca2+-activated phosphatase, regulates translocation to the nucleus of NFAT, a transcriptional regulator centrally involved in hypertrophic remodelling [53]. While there is still no direct evidence for the involvement of this specific pathway in INaL-induced damage, ranolazine has been shown to affect downstream gene transcription and histomorphometric parameters in heart failure induced by coronary microembolization [54].
A further consequence of pathological INaL enhancement is a derangement in cell energy balance. The main ATP-consuming mechanism involved in the control of cellular homeostasis (the Na+/K+ and SERCA pumps) are conceivably short-circuited by enhanced sarcolemmal Na+ influx and increased Ca2+ leak from the SR (by RyRs facilitation) (Fig. 3). Therefore, INaL enhancement is expected to increase ATP consumption by mechanisms not directly involved in force generation, which would translate into decreased mechanical efficiency. This view is supported by the observation that INaL blockade prevents the drop in myocardial ATP, but not the positive inotropic effect, of ouabain [55]. In addition to improved coronary perfusion, increased myocardial efficiency might partly account for the ability of INaL blockade to improve exercise capacity of ischemic patients without altering systolic mechanical work [56].
It has been reported that increased cytosolic Na+ may also jeopardize mitochondrial function (Fig. 4). Concomitant cytosolic Na+ loading (to 15 mM) impaired Ca2+-triggered mitochondrial energetic adaptation in patch-clamped myocytes, thereby causing an abnormal drop in NADH (i.e metabolic energy) upon catecholamine challenge [57]. On the other hand, ranolazine pre-treatment failed to modify NADH course during global ischemia/reperfusion in isolated hearts [43]. Notably, in the same preparation, ranolazine opposed ischemia-induced rise in cytosolic and mitochondrial Ca2+; this was associated, as expected, with diminished ROS generation and delayed opening of mitochondrial permeability transition pore (mPTP) [43]. A plausible network of mechanisms in accord with these observations is illustrated in Fig. 4. Altogether, this preliminary evidence suggests that, during ischemia/reperfusion, INaL inhibition may be more effective in preserving integrity of mitochondria than in improving their function. Although ranolazine effects on energy metabolism can be accounted for by INaL inhibition, the drug has been also reported to inhibit fatty acid oxidation [58, 59], a further action potentially protecting mitochondria during metabolic stress. However, this action was observed with drug dosage (60–200 mg/Kg/day) far from that required for the anti-ischemic effect in humans (10–15 mg/Kg/day).
Under normal conditions, cellular H+ homeostasis may be less sensitive than Ca2+ homeostasis to INaL enhancement, because mechanisms other than NHE contribute to H+ extrusion. During abrupt reperfusion following acute coronary occlusion, massive H+ extrusion through NHE contributes to intracellular Na+ loading, which explains why direct NHE inhibition paradoxically improves Ca2+ handling and limits myocardial injury [60]. Nevertheless, the situation might be different in the presence of chronic partial ischemia, when reduced H+ export by NHE might lead to persistent intracellular acidosis, potentially contributing to impair force development and relaxation. Although awaiting experimental confirmation, this hypothesis is supported by the observation that the increment in intracellular Na+ caused by Na+/K+ pump blockade does produce intracellular acidosis, which can be prevented by concomitant INaL blockade [55].

Functional Interaction Between INaL and K+ Currents

Several observations suggest a complex interplay, with partly unexplained mechanisms, between INaL and K+ currents during repolarization. Such an interplay may have consequences on electrophysiology and ionic homeostasis.
The impact on repolarization of blocking endogenous INaL in the presence of K+ current inhibition (IKr, Ito or IK1) [36] may look disproportionately large to the small magnitude of the inward current removed by INaL block. Although ranolazine, often used as the INaL blocker, shares the channel binding domain on HERG channels (carrying IKr) with E-4031, it cannot competitively displace E-4031 [36]. Moreover, ranolazine effects are largely mimicked by TTX, which does not bind to HERG channels. Therefore, this disproportion cannot be attributed to ancillary ranolazine properties and remains largely unexplained.
A further relevant aspect of INaL blockade is its ability to interfere with the intrinsic reverse rate-dependency of APD and of its amplification by K+ channel blockers [18]. Because INaL decreases at faster rates (see above), INaL block shortens APD more at slower ones, thus dampening the intrinsic reverse rate-dependency of repolarization [18]. Rate-dependency of INaL may also account for the ability of INaL block to flatten “APD restitution” curves, an action strongly correlated with antifibrillatory effect [34].
Recent work on olfactory neurons highlights a structurally organized relationship between Na+ channels and the Na+-activated K+ current (IKNa) [61], whereby the latter is specifically activated by Na+ influx through INaL. IKNa activation may oppose INaL-induced membrane depolarization but, by doing this, it would increases the driving force for Na+ influx. Thus, coupling to IKNa might partially dissociate the effects of INaL enhancement on membrane potential from those on intracellular ionic homeostasis. IKNa is expressed in (guinea-pig) ventricular myocytes [62] and contributes to regulate APD [63]. However, its structural association with cardiac INa channels and functional interaction with INaL enhancement has not been investigated thus far.

Conclusions

Although of small magnitude, endogenous INaL contributes in setting repolarization course of normal myocytes, being in a delicate balance with K+ currents. Disruption of this balance may strongly affect repolarization and its stability. Furthermore, INaL enhancement, occurring in many pathological conditions as a general response to cell stress, can profoundly alter intracellular ionic homeostasis with consequences on contractile function, electrical stability and cell fate. Accordingly, INaL represents a therapeutic target with an expectedly pleiotropic effect, which is being gradually unveiled by experimental and clinical studies.
Open Access This article is distributed under the terms of the Creative Commons Attribution License which permits any use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Wang DW, Yazawa K, George Jr AL, Bennett PB. Characterization of human cardiac Na+ channel mutations in the congenital long QT syndrome. Proc Natl Acad Sci U S A. 1996;93:13200–5.PubMedCrossRef Wang DW, Yazawa K, George Jr AL, Bennett PB. Characterization of human cardiac Na+ channel mutations in the congenital long QT syndrome. Proc Natl Acad Sci U S A. 1996;93:13200–5.PubMedCrossRef
2.
Zurück zum Zitat January CT, Riddle JM. Early afterdepolarizations: mechanism of induction and block. A role for L-type Ca2+ current. Circ Res. 1989;64:977–90.PubMedCrossRef January CT, Riddle JM. Early afterdepolarizations: mechanism of induction and block. A role for L-type Ca2+ current. Circ Res. 1989;64:977–90.PubMedCrossRef
3.
Zurück zum Zitat Zeng J, Rudy Y. Early afterdepolarizations in cardiac myocytes: mechanism and rate dependence. Biophys J. 1995;68:949–64.PubMedCrossRef Zeng J, Rudy Y. Early afterdepolarizations in cardiac myocytes: mechanism and rate dependence. Biophys J. 1995;68:949–64.PubMedCrossRef
4.
Zurück zum Zitat Hirano Y, Moscucci A, January CT. Direct measurement of L-type Ca2+ window current in heart cells. Circ Res. 1992;70:445–55.PubMedCrossRef Hirano Y, Moscucci A, January CT. Direct measurement of L-type Ca2+ window current in heart cells. Circ Res. 1992;70:445–55.PubMedCrossRef
5.
Zurück zum Zitat Attwell D, Cohen IS, Eisner DA, Ohba M, Ojeda C. The steady state TTX-sensitive (“window”) sodium current in cardiac Purkinje fibres. Pflügers Arch. 1979;379:137–42.PubMedCrossRef Attwell D, Cohen IS, Eisner DA, Ohba M, Ojeda C. The steady state TTX-sensitive (“window”) sodium current in cardiac Purkinje fibres. Pflügers Arch. 1979;379:137–42.PubMedCrossRef
6.
Zurück zum Zitat Colatsky TJ. Mechanism of action of lidocaine and quinidine on action potential duration in rabbit cardiac Purkinje fibers. Effects on steady state Na+ currents? Circ Res. 1982;50:17–27.PubMedCrossRef Colatsky TJ. Mechanism of action of lidocaine and quinidine on action potential duration in rabbit cardiac Purkinje fibers. Effects on steady state Na+ currents? Circ Res. 1982;50:17–27.PubMedCrossRef
7.
Zurück zum Zitat Clancy CE, Tateyama M, Liu H, Wehrens XH, Kass RS. Non-equilibrium gating in cardiac Na+ channels: an original mechanism of arrhythmia. Circulation. 2003;107:2233–7.PubMedCrossRef Clancy CE, Tateyama M, Liu H, Wehrens XH, Kass RS. Non-equilibrium gating in cardiac Na+ channels: an original mechanism of arrhythmia. Circulation. 2003;107:2233–7.PubMedCrossRef
8.
Zurück zum Zitat Berecki G, Zegers JG, Bhuiyan ZA, Verkerk AO, Wilders R, van Ginneken AC. Long-QT syndrome-related sodium channel mutations probed by the dynamic action potential clamp technique. J Physiol. 2006;570:237–50.PubMed Berecki G, Zegers JG, Bhuiyan ZA, Verkerk AO, Wilders R, van Ginneken AC. Long-QT syndrome-related sodium channel mutations probed by the dynamic action potential clamp technique. J Physiol. 2006;570:237–50.PubMed
9.
Zurück zum Zitat Patlak JB, Ortiz M. Slow currents through single sodium channels of the adult rat heart. J Gen Physiol. 1985;86:89–104.PubMedCrossRef Patlak JB, Ortiz M. Slow currents through single sodium channels of the adult rat heart. J Gen Physiol. 1985;86:89–104.PubMedCrossRef
10.
Zurück zum Zitat Maltsev VA, Undrovinas AI. A multi-modal composition of the late Na+ current in human ventricular cardiomyocytes. Cardiovasc Res. 2006;69:116–27.PubMedCrossRef Maltsev VA, Undrovinas AI. A multi-modal composition of the late Na+ current in human ventricular cardiomyocytes. Cardiovasc Res. 2006;69:116–27.PubMedCrossRef
11.
Zurück zum Zitat Yang T, Atack TC, Stroud DM, Zhang W, Hall L, Roden DM. Blocking Scn10a channels in heart reduces late sodium current and is antiarrhythmic. Circ Res. 2012;111:322–32.PubMedCrossRef Yang T, Atack TC, Stroud DM, Zhang W, Hall L, Roden DM. Blocking Scn10a channels in heart reduces late sodium current and is antiarrhythmic. Circ Res. 2012;111:322–32.PubMedCrossRef
12.
Zurück zum Zitat Marangoni S, Di Resta C, Rocchetti M, Barile L, Rizzetto R, Summa A, et al. A Brugada syndrome mutation (p.S216L) and its modulation by p.H558R polymorphism: standard and dynamic characterization. Cardiovasc Res. 2011;91:606–16.PubMedCrossRef Marangoni S, Di Resta C, Rocchetti M, Barile L, Rizzetto R, Summa A, et al. A Brugada syndrome mutation (p.S216L) and its modulation by p.H558R polymorphism: standard and dynamic characterization. Cardiovasc Res. 2011;91:606–16.PubMedCrossRef
13.
Zurück zum Zitat Bennett PB, Yazawa K, Makita N, George ALJ. Molecular mechanism for an inherited cardiac arrhythmia. Nature. 1995;376:683–5.PubMedCrossRef Bennett PB, Yazawa K, Makita N, George ALJ. Molecular mechanism for an inherited cardiac arrhythmia. Nature. 1995;376:683–5.PubMedCrossRef
14.
Zurück zum Zitat Undrovinas AI, Maltsev VA, Kyle JW, Silverman N, Sabbah HN. Gating of the late Na+ channel in normal and failing human myocardium. J Mol Cell Cardiol. 2002;34:1477–89.PubMedCrossRef Undrovinas AI, Maltsev VA, Kyle JW, Silverman N, Sabbah HN. Gating of the late Na+ channel in normal and failing human myocardium. J Mol Cell Cardiol. 2002;34:1477–89.PubMedCrossRef
15.
Zurück zum Zitat Maltsev VA, Sabbah HN, Higgins RS, Silverman N, Lesch M, Undrovinas AI. Novel, ultraslow inactivating sodium current in human ventricular cardiomyocytes. Circulation. 1998;98:2545–52.PubMedCrossRef Maltsev VA, Sabbah HN, Higgins RS, Silverman N, Lesch M, Undrovinas AI. Novel, ultraslow inactivating sodium current in human ventricular cardiomyocytes. Circulation. 1998;98:2545–52.PubMedCrossRef
16.
Zurück zum Zitat Maltsev VA, Silverman N, Sabbah HN, Undrovinas AI. Chronic heart failure slows late sodium current in human and canine ventricular myocytes: implications for repolarization variability. Eur J Heart Fail. 2007;9:219–27.PubMedCrossRef Maltsev VA, Silverman N, Sabbah HN, Undrovinas AI. Chronic heart failure slows late sodium current in human and canine ventricular myocytes: implications for repolarization variability. Eur J Heart Fail. 2007;9:219–27.PubMedCrossRef
17.
Zurück zum Zitat Zygmunt AC, Eddlestone GT, Thomas GP, Nesterenko VV, Antzelevitch C. Larger late sodium conductance in M cells contributes to electrical heterogeneity in canine ventricle. Am J Physiol Heart Circ Physiol. 2001;281:H689–97.PubMed Zygmunt AC, Eddlestone GT, Thomas GP, Nesterenko VV, Antzelevitch C. Larger late sodium conductance in M cells contributes to electrical heterogeneity in canine ventricle. Am J Physiol Heart Circ Physiol. 2001;281:H689–97.PubMed
18.
Zurück zum Zitat Wu L, Ma J, Li H, Wang C, Grandi E, Zhang P, et al. Late sodium current contributes to the reverse rate-dependent effect of IKr inhibition on ventricular repolarization. Circulation. 2011;123:1713–20.PubMedCrossRef Wu L, Ma J, Li H, Wang C, Grandi E, Zhang P, et al. Late sodium current contributes to the reverse rate-dependent effect of IKr inhibition on ventricular repolarization. Circulation. 2011;123:1713–20.PubMedCrossRef
19.
Zurück zum Zitat Zaza A, Belardinelli L, Shryock JC. Pathophysiology and pharmacology of the cardiac “late sodium current”. Pharmacol Ther. 2008;119:326–39.PubMedCrossRef Zaza A, Belardinelli L, Shryock JC. Pathophysiology and pharmacology of the cardiac “late sodium current”. Pharmacol Ther. 2008;119:326–39.PubMedCrossRef
20.
Zurück zum Zitat Ward CA, Giles WR. Ionic mechanism of the effects of hydrogen peroxide in rat ventricular myocytes. J Physiol. 1997;500(Pt 3):631–42.PubMed Ward CA, Giles WR. Ionic mechanism of the effects of hydrogen peroxide in rat ventricular myocytes. J Physiol. 1997;500(Pt 3):631–42.PubMed
21.
Zurück zum Zitat Ju YK, Saint DA, Gage PW. Hypoxia increases persistent sodium current in rat ventricular myocytes. J Physiol. 1996;497(Pt 2):337–47.PubMed Ju YK, Saint DA, Gage PW. Hypoxia increases persistent sodium current in rat ventricular myocytes. J Physiol. 1996;497(Pt 2):337–47.PubMed
22.
Zurück zum Zitat Wu J, Corr PB. Palmitoyl carnitine modifies sodium currents and induces transient inward current in ventricular myocytes. Am J Physiol. 1994;266:H1034–46.PubMed Wu J, Corr PB. Palmitoyl carnitine modifies sodium currents and induces transient inward current in ventricular myocytes. Am J Physiol. 1994;266:H1034–46.PubMed
23.
Zurück zum Zitat Wang P, Fraser H, Lloyd SG, McVeigh JJ, Belardinelli L, Chatham JC. A comparison between ranolazine and CVT-4325, a novel inhibitor of fatty acid oxidation, on cardiac metabolism and left ventricular function in rat isolated perfused heart during ischemia and reperfusion. J Pharmacol Exp Ther. 2007;321:213–20.PubMedCrossRef Wang P, Fraser H, Lloyd SG, McVeigh JJ, Belardinelli L, Chatham JC. A comparison between ranolazine and CVT-4325, a novel inhibitor of fatty acid oxidation, on cardiac metabolism and left ventricular function in rat isolated perfused heart during ischemia and reperfusion. J Pharmacol Exp Ther. 2007;321:213–20.PubMedCrossRef
24.
Zurück zum Zitat Wagner S, Dybkova N, Rasenack EC, Jacobshagen C, Fabritz L, Kirchhof P, et al. Ca/calmodulin-dependent protein kinase II regulates cardiac Na channels. J Clin Invest. 2006;116:3127–38.PubMedCrossRef Wagner S, Dybkova N, Rasenack EC, Jacobshagen C, Fabritz L, Kirchhof P, et al. Ca/calmodulin-dependent protein kinase II regulates cardiac Na channels. J Clin Invest. 2006;116:3127–38.PubMedCrossRef
25.
Zurück zum Zitat Sossalla S, Maurer U, Schotola H, Hartmann N, Didie M, Zimmermann WH, et al. Diastolic dysfunction and arrhythmias caused by overexpression of CaMKIIdelta(C) can be reversed by inhibition of late Na (+) current. Basic Res Cardiol. 2011;106:263–72.PubMedCrossRef Sossalla S, Maurer U, Schotola H, Hartmann N, Didie M, Zimmermann WH, et al. Diastolic dysfunction and arrhythmias caused by overexpression of CaMKIIdelta(C) can be reversed by inhibition of late Na (+) current. Basic Res Cardiol. 2011;106:263–72.PubMedCrossRef
26.
Zurück zum Zitat Maltsev VA, Reznikov V, Undrovinas NA, Sabbah HN, Undrovinas A. Modulation of late sodium current by Ca2+, calmodulin, and CaMKII in normal and failing dog cardiomyocytes: similarities and differences. Am J Physiol Heart Circ Physiol. 2008;294:H1597–608.PubMedCrossRef Maltsev VA, Reznikov V, Undrovinas NA, Sabbah HN, Undrovinas A. Modulation of late sodium current by Ca2+, calmodulin, and CaMKII in normal and failing dog cardiomyocytes: similarities and differences. Am J Physiol Heart Circ Physiol. 2008;294:H1597–608.PubMedCrossRef
27.
Zurück zum Zitat Maltsev VA, Undrovinas A. Late sodium current in failing heart: friend or foe? Prog Biophys Mol Biol. 2008;96:421–51.PubMedCrossRef Maltsev VA, Undrovinas A. Late sodium current in failing heart: friend or foe? Prog Biophys Mol Biol. 2008;96:421–51.PubMedCrossRef
28.
Zurück zum Zitat Abriel H, Kass RS. Regulation of the voltage-gated cardiac sodium channel Nav1.5 by interacting proteins. Trends Cardiovasc Med. 2005;15:35–40.PubMedCrossRef Abriel H, Kass RS. Regulation of the voltage-gated cardiac sodium channel Nav1.5 by interacting proteins. Trends Cardiovasc Med. 2005;15:35–40.PubMedCrossRef
29.
Zurück zum Zitat Meadows LS, Isom LL. Sodium channels as macromolecular complexes: implications for inherited arrhythmia syndromes. Cardiovasc Res. 2005;67:448–58.PubMedCrossRef Meadows LS, Isom LL. Sodium channels as macromolecular complexes: implications for inherited arrhythmia syndromes. Cardiovasc Res. 2005;67:448–58.PubMedCrossRef
30.
Zurück zum Zitat Mohler PJ, Schott JJ, Gramolini AO, Dilly KW, Guatimosim S, duBell WH, et al. Ankyrin-B mutation causes type 4 long-QT cardiac arrhythmia and sudden cardiac death. Nature. 2003;421:634–9.PubMedCrossRef Mohler PJ, Schott JJ, Gramolini AO, Dilly KW, Guatimosim S, duBell WH, et al. Ankyrin-B mutation causes type 4 long-QT cardiac arrhythmia and sudden cardiac death. Nature. 2003;421:634–9.PubMedCrossRef
31.
Zurück zum Zitat Vatta M, Ackerman MJ, Ye B, Makielski JC, Ughanze EE, Taylor EW, et al. Mutant caveolin-3 induces persistent late sodium current and is associated with long-QT syndrome. Circulation. 2006;114:2104–12.PubMedCrossRef Vatta M, Ackerman MJ, Ye B, Makielski JC, Ughanze EE, Taylor EW, et al. Mutant caveolin-3 induces persistent late sodium current and is associated with long-QT syndrome. Circulation. 2006;114:2104–12.PubMedCrossRef
32.
Zurück zum Zitat Gintant GA, Datyner NB, Cohen IS. Slow inactivation of a tetrodotoxin-sensitive current in canine cardiac Purkinje fibers. Biophys J. 1984;45:509–12.PubMedCrossRef Gintant GA, Datyner NB, Cohen IS. Slow inactivation of a tetrodotoxin-sensitive current in canine cardiac Purkinje fibers. Biophys J. 1984;45:509–12.PubMedCrossRef
33.
Zurück zum Zitat Vassalle M, Bocchi L, Du F. A slowly inactivating sodium current (INa2) in the plateau range in canine cardiac Purkinje single cells. Exp Physiol. 2007;92:161–73.PubMedCrossRef Vassalle M, Bocchi L, Du F. A slowly inactivating sodium current (INa2) in the plateau range in canine cardiac Purkinje single cells. Exp Physiol. 2007;92:161–73.PubMedCrossRef
34.
Zurück zum Zitat Morita N, Lee JH, Xie Y, Sovari A, Qu Z, Weiss JN, et al. Suppression of re-entrant and multifocal ventricular fibrillation by the late sodium current blocker ranolazine. J Am Coll Cardiol. 2011;57:366–75.PubMedCrossRef Morita N, Lee JH, Xie Y, Sovari A, Qu Z, Weiss JN, et al. Suppression of re-entrant and multifocal ventricular fibrillation by the late sodium current blocker ranolazine. J Am Coll Cardiol. 2011;57:366–75.PubMedCrossRef
35.
Zurück zum Zitat Wu L, Shryock JC, Song Y, Li Y, Antzelevitch C, Belardinelli L. Antiarrhythmic effects of ranolazine in a guinea pig in vitro model of long-QT syndrome. J Pharmacol Exp Ther. 2004;310:599–605.PubMedCrossRef Wu L, Shryock JC, Song Y, Li Y, Antzelevitch C, Belardinelli L. Antiarrhythmic effects of ranolazine in a guinea pig in vitro model of long-QT syndrome. J Pharmacol Exp Ther. 2004;310:599–605.PubMedCrossRef
36.
Zurück zum Zitat Wu L, Rajamani S, Li H, January CT, Shryock JC, Belardinelli L. Reduction of repolarization reserve unmasks the proarrhythmic role of endogenous late Na(+) current in the heart. Am J Physiol Heart Circ Physiol. 2009;297:H1048–57.PubMedCrossRef Wu L, Rajamani S, Li H, January CT, Shryock JC, Belardinelli L. Reduction of repolarization reserve unmasks the proarrhythmic role of endogenous late Na(+) current in the heart. Am J Physiol Heart Circ Physiol. 2009;297:H1048–57.PubMedCrossRef
37.
Zurück zum Zitat Antzelevitch C, Belardinelli L, Zygmunt AC, Burashnikov A, Di Diego JM, Fish JM, et al. Electrophysiological effects of ranolazine, a novel antianginal agent with antiarrhythmic properties. Circulation. 2004;110:904–10.PubMedCrossRef Antzelevitch C, Belardinelli L, Zygmunt AC, Burashnikov A, Di Diego JM, Fish JM, et al. Electrophysiological effects of ranolazine, a novel antianginal agent with antiarrhythmic properties. Circulation. 2004;110:904–10.PubMedCrossRef
38.
Zurück zum Zitat Studenik CR, Zhou Z, January CT. Differences in action potential and early afterdepolarization properties in LQT2 and LQT3 models of long QT syndrome. Br J Pharmacol. 2001;132:85–92.PubMedCrossRef Studenik CR, Zhou Z, January CT. Differences in action potential and early afterdepolarization properties in LQT2 and LQT3 models of long QT syndrome. Br J Pharmacol. 2001;132:85–92.PubMedCrossRef
39.
Zurück zum Zitat Wilson LD, Wan X, Rosenbaum DS. Cellular alternans: a mechanism linking calcium cycling proteins to cardiac arrhythmogenesis. Ann N Y Acad Sci. 2006;1080:216–34.PubMedCrossRef Wilson LD, Wan X, Rosenbaum DS. Cellular alternans: a mechanism linking calcium cycling proteins to cardiac arrhythmogenesis. Ann N Y Acad Sci. 2006;1080:216–34.PubMedCrossRef
40.
Zurück zum Zitat Antzelevitch C, Burashnikov A, Sicouri S, Belardinelli L. Electrophysiological basis for the antiarrhythmic actions of ranolazine. Heart Rhythm. 2011;8:1281–90.PubMedCrossRef Antzelevitch C, Burashnikov A, Sicouri S, Belardinelli L. Electrophysiological basis for the antiarrhythmic actions of ranolazine. Heart Rhythm. 2011;8:1281–90.PubMedCrossRef
41.
Zurück zum Zitat Makielski JC, Farley AL. Na(+) current in human ventricle: implications for sodium loading and homeostasis. J Cardiovasc Electrophysiol. 2006;17 Suppl 1:S15–20.PubMedCrossRef Makielski JC, Farley AL. Na(+) current in human ventricle: implications for sodium loading and homeostasis. J Cardiovasc Electrophysiol. 2006;17 Suppl 1:S15–20.PubMedCrossRef
42.
Zurück zum Zitat Undrovinas NA, Maltsev VA, Belardinelli L, Sabbah HN, Undrovinas A. Late sodium current contributes to diastolic cell Ca2+ accumulation in chronic heart failure. J Physiol Sci. 2010;60:245–57.PubMedCrossRef Undrovinas NA, Maltsev VA, Belardinelli L, Sabbah HN, Undrovinas A. Late sodium current contributes to diastolic cell Ca2+ accumulation in chronic heart failure. J Physiol Sci. 2010;60:245–57.PubMedCrossRef
43.
Zurück zum Zitat Aldakkak M, Camara AK, Heisner JS, Yang M, Stowe DF. Ranolazine reduces Ca2+ overload and oxidative stress and improves mitochondrial integrity to protect against ischemia reperfusion injury in isolated hearts. Pharmacol Res. 2011;64:381–92.PubMedCrossRef Aldakkak M, Camara AK, Heisner JS, Yang M, Stowe DF. Ranolazine reduces Ca2+ overload and oxidative stress and improves mitochondrial integrity to protect against ischemia reperfusion injury in isolated hearts. Pharmacol Res. 2011;64:381–92.PubMedCrossRef
44.
Zurück zum Zitat Bers DM, Despa S, Bossuyt J. Regulation of Ca2+ and Na+ in normal and failing cardiac myocytes. Ann N Y Acad Sci. 2006;1080:165–77.PubMedCrossRef Bers DM, Despa S, Bossuyt J. Regulation of Ca2+ and Na+ in normal and failing cardiac myocytes. Ann N Y Acad Sci. 2006;1080:165–77.PubMedCrossRef
45.
Zurück zum Zitat Gyorke S, Gyorke I, Lukyanenko V, Terentyev D, Viatchenko-Karpinski S, Wiesner TF. Regulation of sarcoplasmic reticulum calcium release by luminal calcium in cardiac muscle. Front Biosci. 2002;7:d1454–63.PubMedCrossRef Gyorke S, Gyorke I, Lukyanenko V, Terentyev D, Viatchenko-Karpinski S, Wiesner TF. Regulation of sarcoplasmic reticulum calcium release by luminal calcium in cardiac muscle. Front Biosci. 2002;7:d1454–63.PubMedCrossRef
46.
Zurück zum Zitat Fredj S, Lindegger N, Sampson KJ, Carmeliet P, Kass RS. Altered Na+ channels promote pause-induced spontaneous diastolic activity in long QT syndrome type 3 myocytes. Circ Res. 2006;99:1225–32.PubMedCrossRef Fredj S, Lindegger N, Sampson KJ, Carmeliet P, Kass RS. Altered Na+ channels promote pause-induced spontaneous diastolic activity in long QT syndrome type 3 myocytes. Circ Res. 2006;99:1225–32.PubMedCrossRef
47.
Zurück zum Zitat Figueredo VM, Pressman GS, Romero-Corral A, Murdock E, Holderbach P, Morris DL. Improvement in left ventricular systolic and diastolic performance during ranolazine treatment in patients with stable angina. J Cardiovasc Pharmacol Ther. 2010. Figueredo VM, Pressman GS, Romero-Corral A, Murdock E, Holderbach P, Morris DL. Improvement in left ventricular systolic and diastolic performance during ranolazine treatment in patients with stable angina. J Cardiovasc Pharmacol Ther. 2010.
48.
Zurück zum Zitat Venkataraman R, Belardinelli L, Blackburn B, Heo J, Iskandrian AE. A study of the effects of ranolazine using automated quantitative analysis of serial myocardial perfusion images. JACC Cardiovasc Imaging. 2009;2:1301–9.PubMedCrossRef Venkataraman R, Belardinelli L, Blackburn B, Heo J, Iskandrian AE. A study of the effects of ranolazine using automated quantitative analysis of serial myocardial perfusion images. JACC Cardiovasc Imaging. 2009;2:1301–9.PubMedCrossRef
49.
Zurück zum Zitat Duncker DJ, Bache RJ. Regulation of coronary blood flow during exercise. Physiol Rev. 2008;88:1009–86.PubMedCrossRef Duncker DJ, Bache RJ. Regulation of coronary blood flow during exercise. Physiol Rev. 2008;88:1009–86.PubMedCrossRef
50.
Zurück zum Zitat Yao L, Fan P, Jiang Z, Viatchenko-Karpinski S, Wu Y, Kornyeyev D, et al. Nav1.5-dependent persistent Na+ influx activates CaMKII in rat ventricular myocytes and N1325S mice. Am J Physiol Cell Physiol. 2011;301:C577–86.PubMedCrossRef Yao L, Fan P, Jiang Z, Viatchenko-Karpinski S, Wu Y, Kornyeyev D, et al. Nav1.5-dependent persistent Na+ influx activates CaMKII in rat ventricular myocytes and N1325S mice. Am J Physiol Cell Physiol. 2011;301:C577–86.PubMedCrossRef
51.
Zurück zum Zitat Maier LS, Bers DM. Role of Ca2+/calmodulin-dependent protein kinase (CaMK) in excitation-contraction coupling in the heart. Cardiovasc Res. 2007;73:631–40.PubMedCrossRef Maier LS, Bers DM. Role of Ca2+/calmodulin-dependent protein kinase (CaMK) in excitation-contraction coupling in the heart. Cardiovasc Res. 2007;73:631–40.PubMedCrossRef
52.
Zurück zum Zitat Maier LS, Zhang T, Chen L, DeSantiago J, Brown JH, Bers DM. Transgenic CaMKIIdeltaC overexpression uniquely alters cardiac myocyte Ca2+ handling: reduced SR Ca2+ load and activated SR Ca2+ release. Circ Res. 2003;92:904–11.PubMedCrossRef Maier LS, Zhang T, Chen L, DeSantiago J, Brown JH, Bers DM. Transgenic CaMKIIdeltaC overexpression uniquely alters cardiac myocyte Ca2+ handling: reduced SR Ca2+ load and activated SR Ca2+ release. Circ Res. 2003;92:904–11.PubMedCrossRef
53.
Zurück zum Zitat Heineke J, Molkentin JD. Regulation of cardiac hypertrophy by intracellular signalling pathways. Nat Rev Mol Cell Biol. 2006;7:589–600.PubMedCrossRef Heineke J, Molkentin JD. Regulation of cardiac hypertrophy by intracellular signalling pathways. Nat Rev Mol Cell Biol. 2006;7:589–600.PubMedCrossRef
54.
Zurück zum Zitat Rastogi S, Sharov VG, Mishra S, Gupta RC, Blackburn B, Belardinelli L, et al. Ranolazine combined with enalapril or metoprolol prevents progressive LV dysfunction and remodeling in dogs with moderate heart failure. Am J Physiol Heart Circ Physiol. 2008;295:H2149–55.PubMedCrossRef Rastogi S, Sharov VG, Mishra S, Gupta RC, Blackburn B, Belardinelli L, et al. Ranolazine combined with enalapril or metoprolol prevents progressive LV dysfunction and remodeling in dogs with moderate heart failure. Am J Physiol Heart Circ Physiol. 2008;295:H2149–55.PubMedCrossRef
55.
Zurück zum Zitat Hoyer K, Song Y, Wang D, Phan D, Balschi J, Ingwall JS, et al. Reducing the late sodium current improves cardiac function during sodium pump inhibition by ouabain. J Pharmacol Exp Ther. 2011;337:513–23.PubMedCrossRef Hoyer K, Song Y, Wang D, Phan D, Balschi J, Ingwall JS, et al. Reducing the late sodium current improves cardiac function during sodium pump inhibition by ouabain. J Pharmacol Exp Ther. 2011;337:513–23.PubMedCrossRef
56.
Zurück zum Zitat Stone PH, Chaitman BR, Stocke K, Sano J, DeVault A, Koch GG. The anti-ischemic mechanism of action of ranolazine in stable ischemic heart disease. J Am Coll Cardiol. 2010;56:934–42.PubMedCrossRef Stone PH, Chaitman BR, Stocke K, Sano J, DeVault A, Koch GG. The anti-ischemic mechanism of action of ranolazine in stable ischemic heart disease. J Am Coll Cardiol. 2010;56:934–42.PubMedCrossRef
57.
Zurück zum Zitat Maack C, Cortassa S, Aon MA, Ganesan AN, Liu T, O’Rourke B. Elevated cytosolic Na+ decreases mitochondrial Ca2+ uptake during excitation-contraction coupling and impairs energetic adaptation in cardiac myocytes. Circ Res. 2006;99:172–82.PubMedCrossRef Maack C, Cortassa S, Aon MA, Ganesan AN, Liu T, O’Rourke B. Elevated cytosolic Na+ decreases mitochondrial Ca2+ uptake during excitation-contraction coupling and impairs energetic adaptation in cardiac myocytes. Circ Res. 2006;99:172–82.PubMedCrossRef
58.
Zurück zum Zitat Abdalla S, Fu X, Elzahwy SS, Klaetschke K, Streichert T, Quitterer U. Up-regulation of the cardiac lipid metabolism at the onset of heart failure. Cardiovasc Hematol Agents Med Chem. 2011;9:190–206.PubMedCrossRef Abdalla S, Fu X, Elzahwy SS, Klaetschke K, Streichert T, Quitterer U. Up-regulation of the cardiac lipid metabolism at the onset of heart failure. Cardiovasc Hematol Agents Med Chem. 2011;9:190–206.PubMedCrossRef
59.
Zurück zum Zitat Fang YH, Piao L, Hong Z, Toth PT, Marsboom G, Bache-Wiig P, et al. Therapeutic inhibition of fatty acid oxidation in right ventricular hypertrophy: exploiting Randle’s cycle. J Mol Med (Berl). 2012;90:31–43.CrossRef Fang YH, Piao L, Hong Z, Toth PT, Marsboom G, Bache-Wiig P, et al. Therapeutic inhibition of fatty acid oxidation in right ventricular hypertrophy: exploiting Randle’s cycle. J Mol Med (Berl). 2012;90:31–43.CrossRef
60.
Zurück zum Zitat Vaughan-Jones RD, Spitzer KW, Swietach P. Intracellular pH regulation in heart. J Mol Cell Cardiol. 2009;46:318–31.PubMedCrossRef Vaughan-Jones RD, Spitzer KW, Swietach P. Intracellular pH regulation in heart. J Mol Cell Cardiol. 2009;46:318–31.PubMedCrossRef
61.
Zurück zum Zitat Hage TA, Salkoff L. Sodium-activated potassium channels are functionally coupled to persistent sodium currents. J Neurosci. 2012;32:2714–21.PubMedCrossRef Hage TA, Salkoff L. Sodium-activated potassium channels are functionally coupled to persistent sodium currents. J Neurosci. 2012;32:2714–21.PubMedCrossRef
62.
Zurück zum Zitat Luk HN, Carmeliet E. Na(+)-activated K+ current in cardiac cells: rectification, open probability, block and role in digitalis toxicity. Pflugers Arch. 1990;416:766–8.PubMedCrossRef Luk HN, Carmeliet E. Na(+)-activated K+ current in cardiac cells: rectification, open probability, block and role in digitalis toxicity. Pflugers Arch. 1990;416:766–8.PubMedCrossRef
63.
Zurück zum Zitat Rodrigo GC, Chapman RA. A sodium-activated potassium current in intact ventricular myocytes isolated from the guinea-pig heart. Exp Physiol. 1990;75:839–42.PubMed Rodrigo GC, Chapman RA. A sodium-activated potassium current in intact ventricular myocytes isolated from the guinea-pig heart. Exp Physiol. 1990;75:839–42.PubMed
64.
Zurück zum Zitat Murgia M, Giorgi C, Pinton P, Rizzuto R. Controlling metabolism and cell death: at the heart of mitochondrial calcium signalling. J Mol Cell Cardiol. 2009;46:781–8.PubMedCrossRef Murgia M, Giorgi C, Pinton P, Rizzuto R. Controlling metabolism and cell death: at the heart of mitochondrial calcium signalling. J Mol Cell Cardiol. 2009;46:781–8.PubMedCrossRef
Metadaten
Titel
The Late Na+ Current - Origin and Pathophysiological Relevance
verfasst von
Antonio Zaza
Marcella Rocchetti
Publikationsdatum
01.02.2013
Verlag
Springer US
Erschienen in
Cardiovascular Drugs and Therapy / Ausgabe 1/2013
Print ISSN: 0920-3206
Elektronische ISSN: 1573-7241
DOI
https://doi.org/10.1007/s10557-012-6430-0

Weitere Artikel der Ausgabe 1/2013

Cardiovascular Drugs and Therapy 1/2013 Zur Ausgabe

Update Kardiologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.