Next Article in Journal
Perspective and Therapeutic Potential of the Noncoding RNA–Connexin Axis
Previous Article in Journal
Post-Translational Modifications of Proteins of Malaria Parasites during the Life Cycle
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

NaV1.8 as Proarrhythmic Target in a Ventricular Cardiac Stem Cell Model

by
Nico Hartmann
1,2,*,†,
Maria Knierim
2,3,†,
Wiebke Maurer
1,2,
Nataliya Dybkova
1,2,
Florian Zeman
4,
Gerd Hasenfuß
1,2,
Samuel Sossalla
1,2,5,‡ and
Katrin Streckfuss-Bömeke
1,2,6,*,‡
1
Clinic for Cardiology and Pneumology, University Medical Center, 37075 Göttingen, Germany
2
DZHK (German Center for Cardiovascular Research), Partner Site Göttingen and Rhein Main, 61231 Bad Nauheim, Germany
3
Clinic for Cardio-Thoracic and Vascular Surgery, University Medical Center, 37075 Göttingen, Germany
4
Center for Clinicial Trials, University of Regensburg, 93042 Regensburg, Germany
5
Medical Clinic I, Cardiology and Angiology, Giessen and Department of Cardiology at Kerckhoff Heart and Lung Center, Justus-Liebig-University, 61231 Bad Nauheim, Germany
6
Institute of Pharmacology and Toxicology, University of Würzburg, 97078 Würzburg, Germany
*
Authors to whom correspondence should be addressed.
These authors contributed equally (first authorship).
These authors contributed equally (senior authorship).
Int. J. Mol. Sci. 2024, 25(11), 6144; https://doi.org/10.3390/ijms25116144
Submission received: 3 March 2024 / Revised: 25 May 2024 / Accepted: 30 May 2024 / Published: 2 June 2024
(This article belongs to the Section Molecular Biology)

Abstract

:
The sodium channel NaV1.8, encoded by the SCN10A gene, has recently emerged as a potential regulator of cardiac electrophysiology. We have previously shown that NaV1.8 contributes to arrhythmogenesis by inducing a persistent Na+ current (late Na+ current, INaL) in human atrial and ventricular cardiomyocytes (CM). We now aim to further investigate the contribution of NaV1.8 to human ventricular arrhythmogenesis at the CM-specific level using pharmacological inhibition as well as a genetic knockout (KO) of SCN10A in induced pluripotent stem cell CM (iPSC-CM). In functional voltage-clamp experiments, we demonstrate that INaL was significantly reduced in ventricular SCN10A-KO iPSC-CM and in control CM after a specific pharmacological inhibition of NaV1.8. In contrast, we did not find any effects on ventricular APD90. The frequency of spontaneous sarcoplasmic reticulum Ca2+ sparks and waves were reduced in SCN10A-KO iPSC-CM and control cells following the pharmacological inhibition of NaV1.8. We further analyzed potential triggers of arrhythmias and found reduced delayed afterdepolarizations (DAD) in SCN10A-KO iPSC-CM and after the specific inhibition of NaV1.8 in control cells. In conclusion, we show that NaV1.8-induced INaL primarily impacts arrhythmogenesis at a subcellular level, with minimal effects on systolic cellular Ca2+ release. The inhibition or knockout of NaV1.8 diminishes proarrhythmic triggers in ventricular CM. In conjunction with our previously published results, this work confirms NaV1.8 as a proarrhythmic target that may be useful in an anti-arrhythmic therapeutic strategy.

1. Introduction

Cardiac electrophysiology is a complex and precisely regulated process crucial for the normal functioning of the heart. Sodium channels are essential for the rapid depolarization phase of the cardiac action potential (AP) and contribute to initiating and propagating electrical signals [1,2]. However, under certain pathological conditions, such as heart failure (HF), ischemia, and atrial fibrillation (AF), voltage-gated (NaV) sodium channels exhibit increased reactivation or sustained opening, resulting in the relevant augmentation of a persistent inward current known as the late sodium current (INaL) [3,4,5,6,7,8,9,10,11]. One particular sodium channel isoform, NaV1.8, encoded by the SCN10A gene, has gained attention due to its potential involvement in cardiac electrophysiology. Extensive research has shown that NaV1.8 contributes to nociceptive signaling and pain perception in peripheral sensory neurons [12,13,14,15]. However, genome-wide association studies reported that the SCN10A gene is associated with cardiac arrhythmias [16,17,18]. Although several studies have investigated the expression of SCN10A in ventricular myocardium [19,20,21,22,23,24], there is an ongoing debate regarding the mechanism responsible for the effects associated with NaV1.8 and the specific cellular localization of these effects, whether it is in cardiac ganglia or cardiomyocytes (CM) [20,23]. The present study addresses this knowledge gap using genetic ablation or knockout (KO) of SCN10A in human induced pluripotent stem cell-CM (iPSC-CM) as a tool to supplement the use of pharmacological agents that inhibit the channel. This approach can reveal the inherent impact of NaV1.8 on INaL in human CM without any confounding or unspecific effects of pharmacological inhibitors. This SCN10A iPSC-CM KO model was used in our previous study and demonstrated that NaV1.8 is expressed in HF CM and contributes to INaL formation in ventricular iPSC-CM [22]. Since it is still unclear how NaV1.8-dependent INaL contributes to cellular arrhythmias, the objective of this study was to investigate potential triggers of arrhythmias in human ventricular CM. Therefore, we conducted additional cellular electrophysiological measurements, including the spontaneous release of Ca2+ from the sarcoplasmic reticulum (SR), action potential duration (APD), delayed afterdepolarizations (DADs), and Ca2+ transients to further characterize the arrhythmogenic potential of NaV1.8 in the heart. By utilizing CRISPR/Cas9-based KO technology, we provide compelling evidence for the existence of a NaV1.8-mediated INaL in human CM, contributing valuable information to the existing body of knowledge [22].

2. Results

2.1. Effects of NaV1.8 on INaL in Human Ventricular iPSC-Cardiomyocytes

Homozygous SCN10A/NaV1.8 KO iPSC were differentiated into 2-month-old ventricular CM as described elsewhere [25]. In our previous study, we demonstrated that the knockout of NaV1.8 in ventricular iPSC-CM after mild isoproterenol stimulation (Iso, 50 nmol/L) resulted in a substantial reduction (~70%) in the amplitude of INaL compared to control iPSC-CM [22]. The application of a NaV1.8-specific blocker (PF-01247324) did not produce any additional effect on INaL in NaV1.8 KO iPSC-CM compared to untreated NaV1.8 knockout cells [22]. In the present study, we aimed to validate and expand upon these initial findings by conducting a larger-scale investigation involving a higher number of single ventricular iPSC-CM generated from a higher number of cardiac differentiations. To increase the relatively small amplitude of INaL in healthy human iPSC-CM, we used 50 nmol/L Iso, which is consistent with our previous experiments. To assess potential non-specific effects associated with either genetic KO or pharmacological intervention, we utilized the specific NaV1.8 blocker PF-01247324 at a concentration of 1 µmol/L [22]. In functional voltage-clamp experiments, we showed that INaL was significantly reduced by pharmacological inhibition and the genetic KO of NaV1.8, validating the impact of both approaches on INaL (Figure 1). The increased INaL induced by Iso in control ventricular iPSC-CM (−108.4 ± 9.5 A*ms*F−1) was significantly reduced in KO iPSC-CM by 70.8% (−32.5 ± 4.4 A*ms*F−1, p < 0.0001, Figure 1b) and by the specific NaV1.8 inhibitor, PF-01247324 (1 µmol/L), by 70.0% (−31.6 ± 6.7 A*ms*F−1; p < 0.0001; Figure 1b). No further effects on INaL were observed in KO iPSC-CM after the application of PF-01247324 (−36.3 ± 3.0 A*ms*F−1; p < 0.981; Figure 1b). These results demonstrated in a broader set of experiments based on statistical analyses of multiple cardiac differentiations and not on single iPSC-CM, confirm our previous results that NaV1.8 contributes to INaL formation in ventricular iPSC-CM, demonstrated in a broader set of experiments based on statistical analyses of multiple cardiac differentiations and not single iPSC-CM.

2.2. Effects of NaV1.8 on APD and Arrhythmic Triggers in Ventricular Cells

To explore the potential effects of NaV1.8 on the action potential in human ventricular iPSC-CM, we conducted current-clamp experiments on whole cells. The APD of KO iPSC-CM was not significantly different from that of control cells, both with and without additional application of the specific NaV1.8 blocker PF-01247324 (Figure 2a,b, control at 0.5 Hz, APD90, 395.4 ± 42.0 ms vs. control + PF-01247324 420.9 ± 46.5 ms, an increase of 6%; KO control 340.4 ± 34.6 ms, a decrease of 13% vs. KO + PF-01247324 376.4 ± 34.8 ms, a reduction of 4.8%). Furthermore, the APD in KO cells at 20% repolarization (APD20), at 50% repolarization (APD50), and at 70% repolarization (APD70) were also similar to that of control cells (Supplemental Figure S1, Supplemental Table S1). Potential non-specific effects associated with KO or pharmacological inhibition of NaV1.8 were analyzed by comparing resting membrane potential (RMP), action potential amplitude (APA), and upstroke velocity (Vmax) between all groups. No significant effects of SCN10A-KO or pharmacological inhibition of NaV1.8 were observed on these parameters.

2.3. Role of NaV1.8 in the Generation of SR Ca2+ Leak in the Ventricle

It is known that INaL can induce arrhythmogenic diastolic SR Ca2+ release [26]. Furthermore, we demonstrated in our previous studies that the arrhythmogenic potential of NaV1.8 in the ventricle is exerted through the enhancement of INaL [22,23] and, consequently, the induction of spontaneous SR Ca2+ release on a subcellular level. To further investigate these effects on a standardized larger scale at the CM-specific level and ultimately by KO of NaV1.8 in human CM, we conducted confocal microscopy experiments in ventricular human control and NaV1.8 KO iPSC-CM.
Diastolic confocal line scans using Fluo 4-AM revealed that KO of SCN10A in ventricular human iPSC-CM resulted in a decrease in the frequency of spontaneous arrhythmogenic Ca2+ sparks compared to the corresponding control cells (control: 4.18 ± 0.25 vs. KO: 2.72 ± 0.19 sparks/100 µm/s, p = 0.02). When NaV1.8 was pharmacologically inhibited using PF-01247324, a significant reduction in diastolic Ca2+ sparks was observed in control cells (control + PF-01247324: 2.50 ± 0.22, p = 0.03) but not in KO cells (KO + PF-01247324: 2.36 ± 0.25, p = 0.99) (Figure 3a,b). Thus, the reduction in both INaL and diastolic SR Ca2+ release in NaV1.8 KO or after NaV1.8 inhibition confirms a significant role of this sodium channel in cellular arrhythmogenesis in the human ventricle.
However, the amount of systolic Ca2+ release and the dynamics of Ca2+ sequestration, as investigated in epifluorescence experiments using Fura 2-AM, were left unaffected by KO or pharmacological inhibition of NaV1.8 (Supplemental Figure S2).
We further investigated the effects of pharmacological inhibition and genetic ablation of NaV1.8 on the incidence of diastolic Ca2+ waves, which constitute primary proarrhythmic triggers. The percentage of cells exhibiting diastolic Ca2+ waves was significantly reduced in control ventricular iPSC-CM by inhibition of NaV1.8 with PF-01247324 (percent of all cells exhibiting Ca2+ waves: control 17% vs. control + PF-01247324 7% of all cells exhibiting Ca2+ waves, p < 0.05). Genetic ablation of NaV1.8 showed a slight tendency to reduce Ca2+ wave frequency, but the effect was not statistically significant (control: 17% vs. KO: 9%; p = 0.14; Figure 3c,d).

2.4. NaV1.8 Inhibition or KO Reduces Proarrhythmic Triggers in Ventricular Cardiomyocytes

To gain further insights into the contribution of INaL to cellular arrhythmias, we investigated potential triggers of arrhythmias. Because afterdepolarizations occur infrequently under basal conditions, we augmented cellular SR Ca2+ stores with isoproterenol (Iso) to increase cellular arrhythmias [27]. Under these conditions, we observed a pronounced number of DADs in control iPSC-CM. This effect was significantly reduced by inhibiting NaV1.8 with PF-01247324 and by genetically depleting NaV1.8 (DADs/min., Figure 4a,b, at 0.5 Hz control iPSC-CM 12.9 ± 1.4 DADs/min.; control iPSC-CM + PF-01247324: 2.8 ± 0.7 DADs/min.; KO iPSC-CM control 5.4 ± 1.2 DADs/min.; KO iPSC-CM + PF-01247324: 4.1 ± 1.2 DADs/min.; Figure 4a,b). The application of PF-01247324 had no significant effect on NaV1.8 KO lines, which showed an already reduced incidence of arrhythmic events. Thus, genetic KO or inhibition of NaV1.8 markedly prevented cellular arrhythmias in human ventricular CM. We did not observe any early afterdepolarizations in control or NaV1.8 KO iPSC-CM.

3. Discussion

In this study, we employed CRISPR/Cas9 KO technology in human ventricular CM to demonstrate that the genetic KO of NaV1.8 resulted in a substantial reduction in INaL. No significant effects on APD were observed. The heightened frequency of spontaneous arrhythmogenic SR Ca2+ sparks, and Ca2+ waves elicited by Iso was significantly diminished through pharmacological intervention or genetic KO of NaV1.8. Additionally, DADs and spontaneous action potentials, indirectly elicited by an increased SR Ca2+ leak, were effectively mitigated through either pharmacological intervention or genetic KO of NaV1.8.

3.1. Role of NaV1.8 on INaL Generation

In the current study, we validated the involvement of NaV1.8 in Iso-augmented INaL in human control ventricular iPSC-CM by observing a reduction in INaL through the specific NaV1.8 blocker PF-01247324 or genetic ablation. Our data support the findings of other studies including a report by Yang et al. (2012) [28]. These authors inhibited SCN10A channels with a specific NaV1.8 blocker in adult SCN10A-/- mice and rabbit ventricular CM. Their results showed a reduction in INaL and anti-arrhythmic effects on ventricular arrhythmias after pharmacologic inhibition of NaV1.8 [28]. In our earlier work, Dybkova et al. (2018) identified NaV1.8 in human failing ventricular myocardium and demonstrated its significant contribution to INaL formation [23]. We also exhibited successful modulation of INaL by NaV1.8 inhibitors [23]. Additionally, in another study, NaV1.8 expression was significantly upregulated in human left ventricular hypertrophy (LVH), and increased NaV1.8 expression was associated with enhanced INaL, potentially contributing to the arrhythmogenic substrate and impaired cardiac function observed in LVH [20]. Of note, in addition to its demonstrated presence in atrial and ventricular human CM (adult and iPSC-CM), NaV1.8 associated INaL has also been characterized in various other species (murine, canine and porcine models); thus, some species- and model-dependent differences should be considered [9,10,26,27,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56]
Most of the previous work regarding NaV1.8-related characteristics of INaL was obtained on the basis of pharmacological interventions. Drugs may have additional, unspecific effects, leaving the mechanism by which NaV1.8 generates INaL unclear. The present study addresses this gap in knowledge and confirms our previous results in human SCN10A-KO iPSC-CM by demonstrating that NaV1.8 plays a significant role in generating INaL by taking an alternative approach that is not purely pharmacological. Furthermore, our results demonstrate that NaV1.8-induced INaL is not limited to ganglia, as assumed in the past, but is also present in ventricular CM, as shown by the results of our previous project [22].
Since INaL is typically very small under healthy or basal conditions, we employed isoproterenol to enhance INaL in our cellular model in order to facilitate a comparison between control and KO iPSC-healthy-CM. It is worth noting that without beta-adrenergic stimulation or structural disease, Casini et al. (2019) did not detect any NaV1.8-dependent INaL in non-diseased rabbit ventricular CM by using specific blockers [57]. This underscores the dependence of NaV1.8-associated INaL enhancement on stimulation by pharmacological (beta-adrenergic activation) or pathological circumstances (e.g., chronic CAMKIIδc overexpression [22]), which likely explains the absence of NaV1.8 effects in their study. The fact that INaL is only enhanced under pathological conditions has to be taken into account in the interpretation of experiments and establishes INaL as a disease-specific target.

3.2. Significance of NaV1.8 in INaL and Arrhythmogenesis

Previous studies have established the significant role of INaL in determining the duration of APs in ventricular CM [27,45,58,59,60,61,62]. Considering that NaV1.8 expression is upregulated in human HF, we investigated the impact of NaV1.8-induced INaL on various AP parameters. Consistent with our previous findings, our current study demonstrates that NaV1.8 has only minimal effects on ventricular AP parameters in ventricular control CM or in SCN10A-KO iPSC-CM. At first glance, the results seem to contradict previous findings regarding ventricular NaV1.8 channels, their effect on APD, and their contribution to arrhythmias in the context of HF and hypertrophy. In the failing human heart and in a mouse model with a genetic SCN10A KO, Dybkova et al. (2018) demonstrated that NaV1.8 expression was higher and that this was associated with a minimally prolonged APD, contributing to INaL-induced arrhythmias [23]. The contribution of NaV1.8 to Na+- and Ca2+-dependent cellular arrhythmias revealed that NaV1.8 channels can promote abnormal electrical activity and Ca2+ overload in CM. These proarrhythmic effects were mediated by enhanced Na+ and Ca2+ influx through NaV1.8 channels [21]. Moreover, the interaction between CaMKIIδc and NaV1.8 in HF leads to CaMKIIδc phosphorylation of NaV1.8 channels, exacerbating the proarrhythmic effects in the failing hearts [22]. In human LVH, Ahmad et al. (2019) demonstrated that enhanced NaV1.8 function was associated with AP prolongation [20]. Overall, these studies suggest that the increased expression and activity of NaV1.8 channels in failing hearts and hypertrophied ventricles, in contrast to the healthy CM investigated in the present study, may contribute to APD prolongation and proarrhythmic effects. Thus, further investigation of NaV1.8 and its inhibition and of the characteristics of INaL in injured or diseased human iPSC-CM models will be of interest to help clarify mechanisms that play a role under pathological conditions.
We show that the inhibition of NaV1.8 by PF-01247324 of INaL or Vmax was absent in NaV1.8 KO ventricular iPSC-CM. Given that Vmax (dv/dt) serves as an indicator of the rapid influx of Na+ and the peak Na+ current, these findings indicate that NaV1.8 does not play a relevant role in the peak Na+ current in ventricular iPSC CM.
The association between enhanced INaL and an increased susceptibility to arrhythmias is intricate [3,9,45]. This complexity stems from the increased leakage of Ca2+ from the SR, which can instigate a transient inward current, subsequently giving rise to arrhythmogenic DADs. In the current study, we observed a decrease in the frequency of spontaneous SR Ca2+ sparks and spontaneous Ca2+ waves in human ventricular iPSC-CM with SCN10A KO as well as in control CM after pharmacological inhibition of NaV1.8. Since Ca2+ waves are known to act as potent proarrhythmic triggers, our findings provide evidence for the involvement of NaV1.8-induced INaL in cellular arrhythmogenesis in isolated human ventricular iPSC-CM. This is in line with our previous investigations, which demonstrated that the INaL, mediated by the influx of Na+, can induce the influx of Ca2+ via the reverse mode of the sodium-calcium exchanger (NCX) in the human ventricle. This process can lead to an elevation in the cytosolic concentration of Ca2+ and an increased occurrence of ryanodine receptor (RyR2) Ca2+ release events [21]. Furthermore, inhibiting INaL by using specific NaV1.8 channel inhibitors has been shown to reduce the reverse mode of the NCX and, consequently, mitigate diastolic proarrhythmogenic SR Ca2+ leakage [20,21,23]. Interestingly, we did not observe any effects on intracellular Ca2+ transients in SCN10A-KO CM or following NaV1.8 inhibition in control CM. This implies there are no negative inotropic effects of NaV1.8 inhibition, which is particularly noteworthy considering the situation in HF patients.
Since INaL-dependent diastolic SR Ca2+ release has the potential to induce an NCX-mediated depolarizing current (ITi), resulting in cellular arrhythmias [63,64,65], we examined the impact of NaV1.8 on afterdepolarizations and spontaneous action potentials. In our current investigation, a substantial number of DADs was noted in control iPSC-CM following Iso application. This effect was significantly mitigated by either inhibiting NaV1.8 with PF-01247324 or through genetic depletion.
It is widely recognized that an augmented INaL results in cellular Na+ accumulation [66,67], subsequently leading to Ca2+ overload through activation of the NCX reverse mode [9,21,26,68,69]. Our earlier investigations have elucidated that this sequence of events may initiate the diastolic release of Ca2+ from the SR and the subsequent occurrence of DADs. These effects are attributed to an intensified phosphorylation of RyR2, regulated by CaMKIIδc [27]; this may explain our current findings and how NaV1.8 influences arrhythmogenesis via INaL.

3.3. Clinical Relevance

Ventricular arrhythmias, such as ventricular tachycardia and ventricular fibrillation, are life-threatening conditions associated with a high risk of sudden cardiac death [60,70]. Studies have shown that increased NaV1.8-dependent INaL plays a role in the development of arrhythmia in various pathological conditions, including HF, LVH, and other structural heart diseases in ventricular CM [20,22,23]. Unfortunately, in HF patients, current anti-arrhythmic medications, such as amiodarone, have shown severe adverse effects, potentially including potentially organ toxicity [71]. Therefore, there is a crucial need for safer and more effective compounds to treat arrhythmias, especially in patients with HF. In the context of trials testing oral selective inhibitors in patients, the findings of the aforementioned article suggest a promising and easily implemented strategy for drug repurposing [71]. Targeting NaV1.8 and specifically inhibiting INaL emerges as a potential therapeutic approach to prevent or treat ventricular arrhythmias. The current study shows that either genetic elimination of NaV1.8 through SCN10A KO in iPSC-CM or pharmacological inhibition of NaV1.8 can effectively reverse the proarrhythmic effects at the cellular level in ventricular cells.

4. Materials and Methods

All procedures conducted in this study adhered to the principles outlined in the Declaration of Helsinki and received approval from the local ethics committee of the University Medical Center of Göttingen (Az-10/9/15). Informed consent was signed by all tissue donors.

4.1. Generating Homozygous Knockout iPSC

The generation of the homozygous SCN10A-KO iPSC line was carried out following the previously described protocols [22,25]. Briefly, we used a previously described wildtype iPSC line [72] as a template and edited it with the CRISPR-Cas9 technique as described in Maurer et al. [25]. The wildtype iPSC were electroporated with a mixture of two guideRNAs, a tracrRNA, and the Cas9 protein using the Human Stem Cell Nucleofector Kit (Amaxa VPH-5022) and the Amaxa Nucleofection II Device (Lonza, program B-016). An electroporation enhancer was added to enhance electroporation efficiency. After electroporation, 72 colonies were expanded, and genomic DNA was analyzed by Sanger sequencing. A promising iPSC mixed clone was chosen for further singularisation. After successful singularisation and additional Sanger sequencing of genomic DNA, two homozygously edited SCN10A knockout iPSC clones were obtained. These clones were regularly sequenced to check for maintenance of the knockout gene editing.

4.2. IPSC-Cardiomyocyte Differentiation

Two NaV1.8 KO iPSC lines (K62.1 and K62.4) and the corresponding isogenic control iPSC line [72] were cultured and differentiated into ventricular iPSC-CM as described previously [22,72]. In brief, iPSCs were cultured in a monolayer on Geltrex®-coated dishes in chemically defined E8 medium (Life Technologies) until 80–90% confluency. Then, we initiated cardiac differentiation by manipulating Wnt signaling. Wnt signaling was activated by a medium change to cardio differentiation medium (RPMI 1640 with GlutaMAX medium (Gibco) supplemented with 0.02% L-ascorbic acid 2-phosphate (Sigma-Aldrich) and 0.05% albumin (Sigma-Aldrich)), supplemented with the GSK3 inhibitor CHIR99021 (4 µmol/L, Millipore). Subsequently, Wnt signaling was inhibited by changing the medium 48 h later to cardio differentiation medium supplemented with 5 µmol/L of the inhibitor of Wnt production-2 (IWP2, Millipore) for another 48 h. The medium was changed to cardio differentiation medium without adding small molecules for 48 h. From day 6 onward, the medium was changed to cardio culture medium (RPMI 1640 with GlutaMAX medium supplemented with 2% B27 with insulin (Gibco)), which was changed every 2 to 3 days. From day 14 to 21, the cells began beating. They were then passaged as 600,000 cells/well onto 6-well culture dishes and metabolically selected for CM for 4 to 5 days by depletion of glucose but supplementation of lactate (4 mmol/L) in a specified cardio selection medium (RPMI 1640 w/o glucose supplemented with 0.02% L-ascorbic acid 2-phosphate (Sigma-Aldrich) and 0.05% albumin (Sigma-Aldrich)). IPSC-CM were matured in cardio culture medium, with a medium change twice a week. The purity of the cells was determined by flow cytometry (>90% cTNT+ cells), immunofluorescence staining, and qPCR analysis for cardiac ventricular sub-type markers. Measurements were performed roughly 60 to 90 days after the initiation of the differentiation process. For functional experiments, iPSC-CM were plated on glass dishes with different cell numbers (World Precision Instruments).

4.3. Standard Operating Procedures

All electrophysiological measurements were conducted following a standardized protocol. A total of 35,000 ventricular iPSC-CM were plated on glass-bottomed Fluoro Dishes. Spontaneously beating ventricular iPSC-CM were stimulated at 0.5 Hz during experiments to achieve a steady state. Single-cell measurements were performed at room temperature with a stimulation frequency of 0.5 Hz/1 Hz/2 Hz. To selectively inhibit sodium currents induced by NaV1.8, a specific NaV1.8 blocker, PF-01247324 (1 µmol/L, Sigma-Aldrich, Taufkirchen, Germany), was utilized. Cellular electrophysiological measurements were carried out with mild beta-adrenergic stimulation using isoproterenol (Iso) at a concentration of 50 nmol/L (Sigma-Aldrich, Taufkirchen, Germany). iPSC-CM were incubated with either Iso (50 nmol/L, or Iso + PF-01247324 (1 µmol/L, Sigma-Aldrich, Taufkirchen, Germany) for 15 min before starting measurements. Regarding the specificity of NaV1.8 blockers, there are already published data that have demonstrated the specificity of PF-01247324 and showed that it is an efficacious inhibitor of NaV1.8 [21,22,73,74].

4.4. Patch-Clamp-Experiments

Patch-clamp experiments were performed as previously described [21,22,74,75]. For action potential recordings, we employed the whole-cell patch-clamp technique in the current clamp configuration. The microelectrodes (3–5 MΩ) were filled with the following solution (in mmol/L): 92 K-aspartate, 48 KCl, 1 Mg-ATP, 10 HEPES, 0.02 EGTA, 0.1 GTP-Tris, and 4 Na2-ATP (pH 7.2, adjusted with KOH). The bath solution consisted of the following components (in mmol/L): 140 NaCl, 4 KCl, 1 MgCl2, 2 CaCl2, 10 glucose, and 10 HEPES (pH 7.4, adjusted with NaOH). Action potentials were continuously elicited by square current pulses with an amplitude of 1–2 nA and a duration of 1–5 ms at a frequency ranging from 0.5 to 2 Hz. The access resistance typically ranged from ~5 to 15 MΩ after patch rupture. Fast capacitance was compensated for in a cell-attached configuration, while membrane capacitance and series resistance were compensated after patch rupture. Signals were filtered using 2.9 and 10 kHz Bessel filters and recorded with an EPC10 from HEKA Elektronik Dr. Schulze GmbH, Lambrecht, Germany. Both the measuring electrode and the bath electrode (earth conductor of the bath solution) were made of chlorinated silver wire (HEKA Elektronik Dr. Schulze GmbH, Lambrecht, Germany).
To measure the INaL ruptured patch, whole-cell patch-clamping was performed at room temperature. The resistance of the pipette was between 2 and 3 mega-Ohm (MΩ) when filled with pipette solution containing (in mmol/L): 95 CsCl, 40 Cs-glutamate, 10 NaCl, 0.92 MgCl2, 5 Mg-ATP, 0.3 Li-GTP, 5 HEPES, 0.03 niflumic acid (to block Ca2+-activated chloride current), 0.02 nifedipine (to block Ca2+ current), 0.004 strophanthidine (to block Na+/K+ ATPase current) 1 EGTA, and 0.36 CaCl2 (free[Ca2+]i, 100 nmol/L) (pH 7.2 with CsOH at room temperature). iPSC-CM were maintained in the bath solution containing (in mmol/L): 135 NaCl, 5 tetramethylammonium chloride, 4 CsCl, 2 MgCl2, 10 glucose, and 10 HEPES (pH 7.4 with CsOH at room temperature). To minimize contaminating Ca2+ currents during INaL measurements, Ca2+ was omitted from the bath solution. INaL was measured only in those iPSC-CM where a seal of more than 1 giga-Ohm was achieved, and the access resistance remained < 7 MΩ. When the whole-cell patch configuration was achieved, iPSC-CM were given a period of 3 min to be stabilized before conducting measurements. Thereafter, iPSC-CM were held at −120 mV before depolarization to −35 mV for a duration of 1000 ms with 10 pulses and a basic cycle length of 2 s. INaL was measured as integral current amplitude between 100 and 500 ms and was normalized to the membrane capacitance.

4.5. Confocal Ca2+ Imaging and Epifluorescence Microscopy

Ca2+ imaging and epifluorescence microscopy were performed as previously described [74].
In detail, for confocal Ca2+ imaging, ventricular iPSC-CM plated on glass-bottom FluoroDishes were incubated with the Ca2+ indicator Fluo 4-AM (10 µmol/L, Invitrogen) for 15 min at room temperature for de-esterification of the dye. The solution was substituted with Tyrode’s solution and the respective pharmacological agents and incubated for 15 min. Confocal line scans were obtained with a laser scanning confocal microscope (LSM 5 Pascal, Zeiss). Scans were conducted after continuous electrical field stimulation at 0.5 Hz during a pause in stimulation. Ca2+ release events were analyzed using the SparkMaster plugin for ImageJ. The mean Ca2+ spark frequency was calculated from the number of sparks normalized to scan width, duration, and scan rate (100 µm/s). Cells exhibiting major Ca2+ release events (Ca2+ wavelets or waves) were excluded from the calculation of Ca2+ spark frequency and separately classified as proarrhythmic cells as a proportion of all cells.
For epifluorescence microscopy, ventricular iPSC-CM were dissociated and plated as described above and loaded with the ratiometric Ca2+ indicator Fura 2-AM (5 µmol/L, Invitrogen) for 15 min at RT. Subsequently, cells were washed with Tyrode’s solution for de-esterification and incubated with pharmacological agents as described above. Measurements were performed using a fluorescence detection system (IonOptix) connected to an inverted microscope with oil immersion lens (40×). CM were subjected to electrical field stimulation at 0.5 Hz for the duration of the experiment to ensure steady intracellular Ca2+ concentration. Recording of Ca2+ transients for analysis was performed at 0.5 Hz at steady state. The stimulation was paused for 30 s to evaluate the spontaneous beating frequency of iPSC-CM (Supplemental Figure S3). Ca2+ transients were analyzed using the software IonWizard (IonOptix).

4.6. Statistical Analysis

The data are reported as mean ± SEM unless otherwise stated. Analysis was carried out with Prism 9 software (Graphpad, San Diego, CA, USA). Three or more groups, including more than one differentiation experiment, were compared using nested one-way ANOVA to account for potential variance between iPSC-CM differentiations. The results were corrected for multiple comparisons by Sidak’s correction. Fisher’s exact test was used to statistically compare proportions. P values are two-sided and considered statistically significant if p < 0.05.

5. Limitations

Using an iPSC-CM model, it is not possible to discriminate between specific cell types located at different parts of the ventricle (epi-, mid-, endocardial). All experiments were performed on a single-cell level, limiting a direct translation of our results and conclusions regarding arrhythmias into an in-vivo situation (e.g., no electrical cell-to-cell conduction).

6. Conclusions

In conclusion, our study demonstrated that NaV1.8 is involved in the formation of INaL in human ventricular iPSC-CM under pathological conditions. Genetic knockout or pharmacological inhibition of NaV1.8 effectively modulated proarrhythmogenic triggers, including INaL, diastolic SR Ca2+ leak, Ca2+ waves, and DADs in human ventricular CM.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/ijms25116144/s1.

Author Contributions

Conceptualization, N.H. and S.S.; Data curation, N.H. and M.K.; Formal analysis, N.H., M.K., F.Z. and N.D.; Funding acquisition, N.H., G.H., S.S. and K.S.-B.; Investigation, N.H. and M.K.; Methodology, N.H., M.K., W.M., N.D., F.Z., S.S. and K.S.-B.; Project administration, S.S.; Resources, N.D., G.H. and S.S.; Software, M.K. and N.D.; Supervision, G.H. and K.S.-B.; Validation, N.H., M.K., S.S. and K.S.-B.; Visualization, N.H., M.K., W.M. and N.D.; Writing—original draft preparation, N.H. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Else-Kröner-Fresenius Foundation to N.H. (2020_EKEA.56); German Heart Foundation/German Foundation of Heart Research (to S.S.) and SFB 1002 (to G.H.); and grants from the Deutsche Forschungsgemeinschaft (DFG) through the International Research Training Group Award (IRTG) 1816 (to K.S.-B.; W.M. is a fellow under IRTG 1816), and to K.S.-B. and S.S. (471241922).

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the local Ethics Committee of the University Medical Center Göttingen (Az-10/9/15). Date of approval: 18/10/16; number of approval: 10/9/15.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The original contributions presented in the study are included in the article/Supplementary Materials, further inquiries can be directed to the corresponding authors.

Acknowledgments

We thank Yvonne Metz and Johanna Heine for their excellent technical assistance.

Conflicts of Interest

N.H. and M.K. declare no conflicts of interest. K.S.-B. received research support from Novartis, BionTECH, and speaker honoraria from Novartis. S.S. received speaker’s/consultancy honoraria from Boehringer Ingelheim, AstraZeneca, Berlin-Chemie, Novartis, Bristol Myers Squibb, and Lilly.

References

  1. Bers, D.M. Excitation-Contraction Coupling and Cardiac Contractile Force, 2nd ed.; Kluwer Academic Publishers: Dordrecht, The Netherlands; Boston, MA, USA, 2001; p. xxiv. 427p. [Google Scholar]
  2. Antzelevitch, C.; Burashnikov, A. Overview of Basic Mechanisms of Cardiac Arrhythmia. Card. Electrophysiol. Clin. 2011, 3, 23–45. [Google Scholar] [CrossRef] [PubMed]
  3. Undrovinas, A.I.; Fleidervish, I.A.; Makielski, J.C. Inward sodium current at resting potentials in single cardiac myocytes induced by the ischemic metabolite lysophosphatidylcholine. Circ. Res. 1992, 71, 1231–1241. [Google Scholar] [CrossRef] [PubMed]
  4. Wu, J.; Corr, P.B. Palmitoyl carnitine modifies sodium currents and induces transient inward current in ventricular myocytes. Am. J. Physiol. 1994, 266, H1034–H1046. [Google Scholar] [CrossRef] [PubMed]
  5. Ju, Y.K.; Saint, D.A.; Gage, P.W. Hypoxia increases persistent sodium current in rat ventricular myocytes. J. Physiol. 1996, 497 Pt 2, 337–347. [Google Scholar] [CrossRef] [PubMed]
  6. Ward, C.A.; Giles, W.R. Ionic mechanism of the effects of hydrogen peroxide in rat ventricular myocytes. J. Physiol. 1997, 500 Pt 3, 631–642. [Google Scholar] [CrossRef] [PubMed]
  7. Maltsev, V.A.; Undrovinas, A.I. Relationship between steady-state activation and availability of cardiac sodium channel: Evidence of uncoupling. Cell. Mol. Life Sci. 1998, 54, 148–151. [Google Scholar] [CrossRef] [PubMed]
  8. Undrovinas, A.I.; Maltsev, V.A.; Sabbah, H.N. Repolarization abnormalities in cardiomyocytes of dogs with chronic heart failure: Role of sustained inward current. Cell. Mol. Life Sci. 1999, 55, 494–505. [Google Scholar] [CrossRef] [PubMed]
  9. Song, Y.; Shryock, J.C.; Belardinelli, L. An increase of late sodium current induces delayed afterdepolarizations and sustained triggered activity in atrial myocytes. Am. J. Physiol. Heart Circ. Physiol. 2008, 294, H2031–H2039. [Google Scholar] [CrossRef] [PubMed]
  10. Valdivia, C.R.; Chu, W.W.; Pu, J.; Foell, J.D.; Haworth, R.A.; Wolff, M.R.; Kamp, T.J.; Makielski, J.C. Increased late sodium current in myocytes from a canine heart failure model and from failing human heart. J. Mol. Cell. Cardiol. 2005, 38, 475–483. [Google Scholar] [CrossRef]
  11. Ronchi, C.; Torre, E.; Rizzetto, R.; Bernardi, J.; Rocchetti, M.; Zaza, A. Late sodium current and intracellular ionic homeostasis in acute ischemia. Basic Res. Cardiol. 2017, 112, 12. [Google Scholar] [CrossRef]
  12. Djouhri, L.; Fang, X.; Okuse, K.; Wood, J.N.; Berry, C.M.; Lawson, S.N. The TTX-resistant sodium channel Nav1.8 (SNS/PN3): Expression and correlation with membrane properties in rat nociceptive primary afferent neurons. J. Physiol. 2003, 550, 739–752. [Google Scholar] [CrossRef] [PubMed]
  13. Dib-Hajj, S.D.; Binshtok, A.M.; Cummins, T.R.; Jarvis, M.F.; Samad, T.; Zimmermann, K. Voltage-gated sodium channels in pain states: Role in pathophysiology and targets for treatment. Brain Res. Rev. 2009, 60, 65–83. [Google Scholar] [CrossRef] [PubMed]
  14. Liu, M.; Wood, J.N. The roles of sodium channels in nociception: Implications for mechanisms of neuropathic pain. Pain Med. 2011, 12 (Suppl. S3), S93–S99. [Google Scholar] [CrossRef] [PubMed]
  15. Akopian, A.N.; Sivilotti, L.; Wood, J.N. A tetrodotoxin-resistant voltage-gated sodium channel expressed by sensory neurons. Nature 1996, 379, 257–262. [Google Scholar] [CrossRef] [PubMed]
  16. Ritchie, M.D.; Denny, J.C.; Zuvich, R.L.; Crawford, D.C.; Schildcrout, J.S.; Bastarache, L.; Ramirez, A.H.; Mosley, J.D.; Pulley, J.M.; Basford, M.A.; et al. Genome- and phenome-wide analyses of cardiac conduction identifies markers of arrhythmia risk. Circulation 2013, 127, 1377–1385. [Google Scholar] [CrossRef] [PubMed]
  17. Bezzina, C.R.; Barc, J.; Mizusawa, Y.; Remme, C.A.; Gourraud, J.B.; Simonet, F.; Verkerk, A.O.; Schwartz, P.J.; Crotti, L.; Dagradi, F.; et al. Common variants at SCN5A-SCN10A and HEY2 are associated with Brugada syndrome, a rare disease with high risk of sudden cardiac death. Nat. Genet. 2013, 45, 1044–1049. [Google Scholar] [CrossRef]
  18. Hu, D.; Barajas-Martinez, H.; Pfeiffer, R.; Dezi, F.; Pfeiffer, J.; Buch, T.; Betzenhauser, M.J.; Belardinelli, L.; Kahlig, K.M.; Rajamani, S.; et al. Mutations in SCN10A are responsible for a large fraction of cases of Brugada syndrome. J. Am. Coll. Cardiol. 2014, 64, 66–79. [Google Scholar] [CrossRef] [PubMed]
  19. Yang, T.; Atack, T.C.; Stroud, D.M.; Zhang, W.; Hall, L.; Roden, D.M. Blocking Scn10a channels in heart reduces late sodium current and is antiarrhythmic. Circ. Res. 2012, 111, 322–332. [Google Scholar] [CrossRef] [PubMed]
  20. Ahmad, S.; Tirilomis, P.; Pabel, S.; Dybkova, N.; Hartmann, N.; Molina, C.E.; Tirilomis, T.; Kutschka, I.; Frey, N.; Maier, L.S.; et al. The functional consequences of sodium channel Na(V) 1.8 in human left ventricular hypertrophy. ESC Heart Fail. 2019, 6, 154–163. [Google Scholar] [CrossRef]
  21. Bengel, P.; Ahmad, S.; Tirilomis, P.; Trum, M.; Dybkova, N.; Wagner, S.; Maier, L.S.; Hasenfuss, G.; Sossalla, S. Contribution of the neuronal sodium channel NaV1.8 to sodium- and calcium-dependent cellular proarrhythmia. J. Mol. Cell. Cardiol. 2020, 144, 35–46. [Google Scholar] [CrossRef]
  22. Bengel, P.; Dybkova, N.; Tirilomis, P.; Ahmad, S.; Hartmann, N.; Mohamed, B.A.; Krekeler, M.C.; Maurer, W.; Pabel, S.; Trum, M.; et al. Detrimental proarrhythmogenic interaction of Ca2+/calmodulin-dependent protein kinase II and NaV1.8 in heart failure. Nat. Commun. 2021, 12, 6586. [Google Scholar] [CrossRef] [PubMed]
  23. Dybkova, N.; Ahmad, S.; Pabel, S.; Tirilomis, P.; Hartmann, N.; Fischer, T.H.; Bengel, P.; Tirilomis, T.; Ljubojevic, S.; Renner, A.; et al. Differential regulation of sodium channels as a novel proarrhythmic mechanism in the human failing heart. Cardiovasc. Res. 2018, 114, 1728–1737. [Google Scholar] [CrossRef] [PubMed]
  24. Stroud, D.M.; Yang, T.; Bersell, K.; Kryshtal, D.O.; Nagao, S.; Shaffer, C.; Short, L.; Hall, L.; Atack, T.C.; Zhang, W.; et al. Contrasting Nav1.8 Activity in Scn10a-/- Ventricular Myocytes and the Intact Heart. J. Am. Heart Assoc. 2016, 5, e002946. [Google Scholar] [CrossRef] [PubMed]
  25. Maurer, W.; Hartmann, N.; Argyriou, L.; Sossalla, S.; Streckfuss-Bomeke, K. Generation of homozygous Na(v)1.8 knock-out iPSC lines by CRISPR Cas9 genome editing to investigate a potential new antiarrhythmic strategy. Stem Cell Res. 2022, 60, 102677. [Google Scholar] [CrossRef] [PubMed]
  26. Undrovinas, N.A.; Maltsev, V.A.; Belardinelli, L.; Sabbah, H.N.; Undrovinas, A. Late sodium current contributes to diastolic cell Ca2+ accumulation in chronic heart failure. J. Physiol. Sci. 2010, 60, 245–257. [Google Scholar] [CrossRef] [PubMed]
  27. Toischer, K.; Hartmann, N.; Wagner, S.; Fischer, T.H.; Herting, J.; Danner, B.C.; Sag, C.M.; Hund, T.J.; Mohler, P.J.; Belardinelli, L.; et al. Role of late sodium current as a potential arrhythmogenic mechanism in the progression of pressure-induced heart disease. J. Mol. Cell. Cardiol. 2013, 61, 111–122. [Google Scholar] [CrossRef] [PubMed]
  28. Lu, Y.Y.; Cheng, C.C.; Chen, Y.C.; Chen, S.A.; Chen, Y.J. ATX-II-induced pulmonary vein arrhythmogenesis related to atrial fibrillation and long QT syndrome. Eur. J. Clin. Investig. 2012, 42, 823–831. [Google Scholar] [CrossRef]
  29. Biet, M.; Barajas-Martinez, H.; Ton, A.T.; Delabre, J.F.; Morin, N.; Dumaine, R. About half of the late sodium current in cardiac myocytes from dog ventricle is due to non-cardiac-type Na(+) channels. J. Mol. Cell. Cardiol. 2012, 53, 593–598. [Google Scholar] [CrossRef] [PubMed]
  30. Bunch, T.J.; Mahapatra, S.; Murdock, D.; Molden, J.; Weiss, J.P.; May, H.T.; Bair, T.L.; Mader, K.M.; Crandall, B.G.; Day, J.D.; et al. Ranolazine reduces ventricular tachycardia burden and ICD shocks in patients with drug-refractory ICD shocks. Pacing Clin. Electrophysiol. PACE 2011, 34, 1600–1606. [Google Scholar] [CrossRef]
  31. Burashnikov, A.; Di Diego, J.M.; Zygmunt, A.C.; Belardinelli, L.; Antzelevitch, C. Atrium-selective sodium channel block as a strategy for suppression of atrial fibrillation: Differences in sodium channel inactivation between atria and ventricles and the role of ranolazine. Circulation 2007, 116, 1449–1457. [Google Scholar] [CrossRef]
  32. Burashnikov, A.; Sicouri, S.; Di Diego, J.M.; Belardinelli, L.; Antzelevitch, C. Synergistic effect of the combination of ranolazine and dronedarone to suppress atrial fibrillation. J. Am. Coll. Cardiol. 2010, 56, 1216–1224. [Google Scholar] [CrossRef] [PubMed]
  33. Dybkova, N.; Wagner, S.; Backs, J.; Hund, T.J.; Mohler, P.J.; Sowa, T.; Nikolaev, V.O.; Maier, L.S. Tubulin polymerization disrupts cardiac beta-adrenergic regulation of late INa. Cardiovasc. Res. 2014, 103, 168–177. [Google Scholar] [CrossRef] [PubMed]
  34. Fischer, T.H.; Herting, J.; Mason, F.E.; Hartmann, N.; Watanabe, S.; Nikolaev, V.O.; Sprenger, J.U.; Fan, P.; Yao, L.; Popov, A.F.; et al. Late INa increases diastolic SR-Ca2+-leak in atrial myocardium by activating PKA and CaMKII. Cardiovasc. Res. 2015, 107, 184–196. [Google Scholar] [CrossRef]
  35. Frommeyer, G.; Milberg, P.; Maier, L.S.; Eckardt, L. Late sodium current inhibition: The most promising antiarrhythmic principle in the near future? Curr. Med. Chem. 2014, 21, 1271–1280. [Google Scholar] [CrossRef] [PubMed]
  36. Greer-Short, A.; Musa, H.; Alsina, K.M.; Ni, L.; Word, T.A.; Reynolds, J.O.; Gratz, D.; Lane, C.; El-Refaey, M.; Unudurthi, S.; et al. Calmodulin kinase II regulates atrial myocyte late sodium current, calcium handling, and atrial arrhythmia. Heart Rhythm Off. J. Heart Rhythm. Soc. 2020, 17, 503–511. [Google Scholar] [CrossRef] [PubMed]
  37. Hale, S.L.; Shryock, J.C.; Belardinelli, L.; Sweeney, M.; Kloner, R.A. Late sodium current inhibition as a new cardioprotective approach. J. Mol. Cell. Cardiol. 2008, 44, 954–967. [Google Scholar] [CrossRef] [PubMed]
  38. Horvath, B.; Hezso, T.; Kiss, D.; Kistamas, K.; Magyar, J.; Nanasi, P.P.; Banyasz, T. Late Sodium Current Inhibitors as Potential Antiarrhythmic Agents. Front. Pharmacol. 2020, 11, 413. [Google Scholar] [CrossRef] [PubMed]
  39. Hwang, H.; Arcidi, J.M., Jr.; Hale, S.L.; Simkhovich, B.Z.; Belardinelli, L.; Dhalla, A.K.; Shryock, J.C.; Kloner, R.A. Ranolazine as a cardioplegia additive improves recovery of diastolic function in isolated rat hearts. Circulation 2009, 120, S16–S21. [Google Scholar] [CrossRef] [PubMed]
  40. Le Grand, B.; Pignier, C.; Letienne, R.; Cuisiat, F.; Rolland, F.; Mas, A.; Vacher, B. Sodium late current blockers in ischemia reperfusion: Is the bullet magic? J. Med. Chem. 2008, 51, 3856–3866. [Google Scholar] [CrossRef]
  41. Maier, L.S. New treatment options for late Na current, arrhythmias, and diastolic dysfunction. Curr. Heart Fail. Rep. 2012, 9, 183–191. [Google Scholar] [CrossRef]
  42. Maier, L.S.; Sossalla, S. The late Na current as a therapeutic target: Where are we? J. Mol. Cell. Cardiol. 2013, 61, 44–50. [Google Scholar] [CrossRef] [PubMed]
  43. Maltsev, V.A.; Kyle, J.W.; Mishra, S.; Undrovinas, A. Molecular identity of the late sodium current in adult dog cardiomyocytes identified by Nav1.5 antisense inhibition. Am. J. Physiol. Heart Circ. Physiol. 2008, 295, H667–H676. [Google Scholar] [CrossRef] [PubMed]
  44. Maltsev, V.A.; Reznikov, V.; Undrovinas, N.A.; Sabbah, H.N.; 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–H1608. [Google Scholar] [CrossRef] [PubMed]
  45. Maltsev, V.A.; Undrovinas, A. Late sodium current in failing heart: Friend or foe? Prog. Biophys. Mol. Biol. 2008, 96, 421–451. [Google Scholar] [CrossRef] [PubMed]
  46. Morita, N.; Lee, J.H.; Xie, Y.; Sovari, A.; Qu, Z.; Weiss, J.N.; Karagueuzian, H.S. Suppression of re-entrant and multifocal ventricular fibrillation by the late sodium current blocker ranolazine. J. Am. Coll. Cardiol. 2011, 57, 366–375. [Google Scholar] [CrossRef] [PubMed]
  47. Noble, D.; Noble, P.J. Late sodium current in the pathophysiology of cardiovascular disease: Consequences of sodium-calcium overload. Heart 2006, 92 (Suppl. S4), iv1–iv5. [Google Scholar] [CrossRef] [PubMed]
  48. Poulet, C.; Wettwer, E.; Grunnet, M.; Jespersen, T.; Fabritz, L.; Matschke, K.; Knaut, M.; Ravens, U. Late Sodium Current in Human Atrial Cardiomyocytes from Patients in Sinus Rhythm and Atrial Fibrillation. PLoS ONE 2015, 10, e01314322015. [Google Scholar] [CrossRef] [PubMed]
  49. Sag, C.M.; Mallwitz, A.; Wagner, S.; Hartmann, N.; Schotola, H.; Fischer, T.H.; Ungeheuer, N.; Herting, J.; Shah, A.M.; Maier, L.S.; et al. Enhanced late INa induces proarrhythmogenic SR Ca leak in a CaMKII-dependent manner. J. Mol. Cell. Cardiol. 2014, 76, 94–105. [Google Scholar] [CrossRef] [PubMed]
  50. Schotten, U.; Verheule, S.; Kerfant, B.G.; Greiser, M. Enhanced late Na(+) currents in atrial fibrillation new drug target or just an epiphenomenon? J. Am. Coll. Cardiol. 2010, 55, 2343–2345. [Google Scholar] [CrossRef]
  51. Song, Y.; Shryock, J.C.; Wagner, S.; Maier, L.S.; Belardinelli, L. Blocking late sodium current reduces hydrogen peroxide-induced arrhythmogenic activity and contractile dysfunction. J. Pharmacol. Exp. Ther. 2006, 318, 214–222. [Google Scholar] [CrossRef]
  52. Song, Y.; Shryock, J.C.; Wu, L.; Belardinelli, L. Antagonism by ranolazine of the pro-arrhythmic effects of increasing late INa in guinea pig ventricular myocytes. J. Cardiovasc. Pharmacol. 2004, 44, 192–199. [Google Scholar] [CrossRef]
  53. Wu, Y.; Song, Y.; Belardinelli, L.; Shryock, J.C. The late Na+ current (INa) inhibitor ranolazine attenuates effects of palmitoyl-L-carnitine to increase late INa and cause ventricular diastolic dysfunction. J. Pharmacol. Exp. Ther. 2009, 330, 550–557. [Google Scholar] [CrossRef] [PubMed]
  54. Xi, Y.; Wu, G.; Yang, L.; Han, K.; Du, Y.; Wang, T.; Lei, X.; Bai, X.; Ma, A. Increased late sodium currents are related to transcription of neuronal isoforms in a pressure-overload model. Eur. J. Heart Fail. 2009, 11, 749–757. [Google Scholar] [CrossRef]
  55. Zygmunt, A.C.; Eddlestone, G.T.; Thomas, G.P.; Nesterenko, V.V.; 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–H697. [Google Scholar] [CrossRef] [PubMed]
  56. Zang, Y.; Dai, L.; Zhan, H.; Dou, J.; Xia, L.; Zhang, H. Theoretical investigation of the mechanism of heart failure using a canine ventricular cell model: Especially the role of up-regulated CaMKII and SR Ca2+ leak. J. Mol. Cell. Cardiol. 2013, 56, 34–43. [Google Scholar] [CrossRef] [PubMed]
  57. Casini, S.; Marchal, G.A.; Kawasaki, M.; Nariswari, F.A.; Portero, V.; van den Berg, N.W.E.; Guan, K.; Driessen, A.H.G.; Veldkamp, M.W.; Mengarelli, I.; et al. Absence of Functional Nav1.8 Channels in Non-diseased Atrial and Ventricular Cardiomyocytes. Cardiovasc. Drugs Ther. 2019, 33, 649–660. [Google Scholar] [CrossRef]
  58. Maltsev, V.A.; Silverman, N.; Sabbah, H.N.; Undrovinas, A.I. Chronic heart failure slows late sodium current in human and canine ventricular myocytes: Implications for repolarization variability. Eur. J. Heart Fail. 2007, 9, 219–227. [Google Scholar] [CrossRef] [PubMed]
  59. Antzelevitch, C.; Belardinelli, L. The role of sodium channel current in modulating transmural dispersion of repolarization and arrhythmogenesis. J. Cardiovasc. Electrophysiol. 2006, 17 (Suppl. S1), S79–S85. [Google Scholar] [CrossRef]
  60. Wagner, S.; Maier, L.S.; Bers, D.M. Role of sodium and calcium dysregulation in tachyarrhythmias in sudden cardiac death. Circ. Res. 2015, 116, 1956–1970. [Google Scholar] [CrossRef]
  61. Maltsev, V.A.; Sabbah, H.N.; Higgins, R.S.; Silverman, N.; Lesch, M.; Undrovinas, A.I. Novel, ultraslow inactivating sodium current in human ventricular cardiomyocytes. Circulation 1998, 98, 2545–2552. [Google Scholar] [CrossRef]
  62. Dai, L.; Zang, Y.; Zheng, D.; Xia, L.; Gong, Y. Role of CaMKII and PKA in Early Afterdepolarization of Human Ventricular Myocardium Cell: A Computational Model Study. Comput. Math Methods Med. 2016, 2016, 4576313. [Google Scholar] [CrossRef] [PubMed]
  63. Lederer, W.J.; Tsien, R.W. Transient inward current underlying arrhythmogenic effects of cardiotonic steroids in Purkinje fibres. J. Physiol. 1976, 263, 73–100. [Google Scholar] [CrossRef]
  64. Cheng, H.; Lederer, W.J. Calcium sparks. Physiol. Rev. 2008, 88, 1491–1545. [Google Scholar] [CrossRef] [PubMed]
  65. Voigt, N.; Li, N.; Wang, Q.; Wang, W.; Trafford, A.W.; Abu-Taha, I.; Sun, Q.; Wieland, T.; Ravens, U.; Nattel, S.; et al. Enhanced sarcoplasmic reticulum Ca2+ leak and increased Na+-Ca2+ exchanger function underlie delayed afterdepolarizations in patients with chronic atrial fibrillation. Circulation 2012, 125, 2059–2070. [Google Scholar] [CrossRef] [PubMed]
  66. Belardinelli, L.; Shryock, J.C.; Fraser, H. Inhibition of the late sodium current as a potential cardioprotective principle: Effects of the late sodium current inhibitor ranolazine. Heart 2006, 92 (Suppl. S4), iv6–iv14. [Google Scholar] [CrossRef] [PubMed]
  67. Makielski, J.C.; Farley, A.L. Na(+) current in human ventricle: Implications for sodium loading and homeostasis. J. Cardiovasc. Electrophysiol. 2006, 17 (Suppl. S1), S15–S20. [Google Scholar] [CrossRef] [PubMed]
  68. Despa, S.; Islam, M.A.; Weber, C.R.; Pogwizd, S.M.; Bers, D.M. Intracellular Na(+) concentration is elevated in heart failure but Na/K pump function is unchanged. Circulation 2002, 105, 2543–2548. [Google Scholar] [CrossRef] [PubMed]
  69. Bers, D.M. Calcium cycling and signaling in cardiac myocytes. Annu. Rev. Physiol. 2008, 70, 23–49. [Google Scholar] [CrossRef] [PubMed]
  70. Zeppenfeld, K.; Tfelt-Hansen, J.; de Riva, M.; Winkel, B.G.; Behr, E.R.; Blom, N.A.; Charron, P.; Corrado, D.; Dagres, N.; de Chillou, C.; et al. 2022 ESC Guidelines for the management of patients with ventricular arrhythmias and the prevention of sudden cardiac death. Eur. Heart J. 2022, 43, 3997–4126. [Google Scholar] [CrossRef]
  71. Echt, D.S.; Liebson, P.R.; Mitchell, L.B.; Peters, R.W.; Obias-Manno, D.; Barker, A.H.; Arensberg, D.; Baker, A.; Friedman, L.; Greene, H.L.; et al. Mortality and morbidity in patients receiving encainide, flecainide, or placebo. The Cardiac Arrhythmia Suppression Trial. N. Engl. J. Med. 1991, 324, 781–788. [Google Scholar] [CrossRef]
  72. Borchert, T.; Hubscher, D.; Guessoum, C.I.; Lam, T.D.; Ghadri, J.R.; Schellinger, I.N.; Tiburcy, M.; Liaw, N.Y.; Li, Y.; Haas, J.; et al. Catecholamine-Dependent beta-Adrenergic Signaling in a Pluripotent Stem Cell Model of Takotsubo Cardiomyopathy. J. Am. Coll. Cardiol. 2017, 70, 975–991. [Google Scholar] [CrossRef] [PubMed]
  73. Payne, C.E.; Brown, A.R.; Theile, J.W.; Loucif, A.J.; Alexandrou, A.J.; Fuller, M.D.; Mahoney, J.H.; Antonio, B.M.; Gerlach, A.C.; Printzenhoff, D.M.; et al. A novel selective and orally bioavailable Nav 1.8 channel blocker, PF-01247324, attenuates nociception and sensory neuron excitability. Br. J. Pharmacol. 2015, 172, 2654–2670. [Google Scholar] [CrossRef] [PubMed]
  74. Hartmann, N.; Knierim, M.; Maurer, W.; Dybkova, N.; Hasenfuss, G.; Sossalla, S.; Streckfuss-Bomeke, K. Molecular and Functional Relevance of Na(V)1.8-Induced Atrial Arrhythmogenic Triggers in a Human SCN10A Knock-Out Stem Cell Model. Int. J. Mol. Sci. 2023, 24, 10189. [Google Scholar] [CrossRef]
  75. Pabel, S.; Ahmad, S.; Tirilomis, P.; Stehle, T.; Mustroph, J.; Knierim, M.; Dybkova, N.; Bengel, P.; Holzamer, A.; Hilker, M.; et al. Inhibition of NaV1.8 prevents atrial arrhythmogenesis in human and mice. Basic Res. Cardiol. 2020, 115, 20. [Google Scholar] [CrossRef] [PubMed]
Figure 1. NaV1.8 contributes to INaL formation in ventricular iPSC-CM. (a) Original traces of INaL in control iPSC-CM vs. SCN10A KO-iPSC-CM cells according to the inserted protocol. (b) Mean values per differentiation ± SEM of INaL (control n = 31 cells/5 differentiations; control + PF n = 14 cells/4 differentiations; SCN10A KO iPSC-CM control n = 18 cells/5 differentiations, SCN10A KO-iPSC-CM + PF n = 24 cells/4 differentiations). Single cells are presented as individual symbols and different iPSC-CM differentiations are color-coded. The small horizontal lines indicate the mean values per differentiation. Data were compared using nested one-way ANOVA with Sidak’s test for multiple comparisons to calculate p values.
Figure 1. NaV1.8 contributes to INaL formation in ventricular iPSC-CM. (a) Original traces of INaL in control iPSC-CM vs. SCN10A KO-iPSC-CM cells according to the inserted protocol. (b) Mean values per differentiation ± SEM of INaL (control n = 31 cells/5 differentiations; control + PF n = 14 cells/4 differentiations; SCN10A KO iPSC-CM control n = 18 cells/5 differentiations, SCN10A KO-iPSC-CM + PF n = 24 cells/4 differentiations). Single cells are presented as individual symbols and different iPSC-CM differentiations are color-coded. The small horizontal lines indicate the mean values per differentiation. Data were compared using nested one-way ANOVA with Sidak’s test for multiple comparisons to calculate p values.
Ijms 25 06144 g001
Figure 2. NaV1.8 inhibition or KO has no significant impact on action potential parameters of iPSC-CM. (a) Original traces of APD90 in ventricular control iPSC-CM vs. SCN10A-KO iPSC-CM at 0.5 Hz. Mean data ± SEM of (b) APD90, (c) AP amplitude, (d) RMP and (e) upstroke velocity (Vmax) of control and SCN10A-KO iPSCM (control n = 18 cells/4 differentiations; control + PF-01247324 [PF] n = 12 cells/3 differentiations; SCN10A-KO control n = 12 cells/3 differentiations, SCN10A-KO + PF n = 16 cells/3 differentiations). Single cells are presented as individual symbols and different iPSC-CM differentiations are color-coded. The small horizontal lines indicate the mean values per differentiation. Data were compared using nested one-way ANOVA with Sidak’s test for multiple comparisons to calculate p values.
Figure 2. NaV1.8 inhibition or KO has no significant impact on action potential parameters of iPSC-CM. (a) Original traces of APD90 in ventricular control iPSC-CM vs. SCN10A-KO iPSC-CM at 0.5 Hz. Mean data ± SEM of (b) APD90, (c) AP amplitude, (d) RMP and (e) upstroke velocity (Vmax) of control and SCN10A-KO iPSCM (control n = 18 cells/4 differentiations; control + PF-01247324 [PF] n = 12 cells/3 differentiations; SCN10A-KO control n = 12 cells/3 differentiations, SCN10A-KO + PF n = 16 cells/3 differentiations). Single cells are presented as individual symbols and different iPSC-CM differentiations are color-coded. The small horizontal lines indicate the mean values per differentiation. Data were compared using nested one-way ANOVA with Sidak’s test for multiple comparisons to calculate p values.
Ijms 25 06144 g002
Figure 3. Contribution of NaV1.8 to spontaneous diastolic SR Ca2+ release in ventricular iPSC-CM. (a) Representative surface plots showing spontaneous diastolic Ca2+ sparks (green) in ventricular iPSC-CM. (b) Mean values of Ca2+ spark frequency (CaSpF) normalized to scan width and duration. Numbers indicate the total cell count of control CM (control, n = 75 cells/4 differentiations), control cells treated with NaV1.8 inhibitor PF-01247324 (control + PF-01247324 [PF], n = 85/4), and KO of NaV1.8 after control treatment and treatment with PF-01247324 (n = 68/4 vs. 67/4). (c) Original representative line-scans of ventricular iPSC-CM illustrating a spontaneous proarrhythmogenic diastolic Ca2+ wave (green). (d) Percentage of cells exhibiting diastolic Ca2+ waves (control: 17% vs. control + PF-01247324: 7% of cells exhibiting Ca2+ waves, p < 0.05; KO: 9% vs. KO + PF-01247324: 7%). Values are presented as mean ± SEM or absolute numbers. Single cells are presented as individual symbols and different iPSC-CM differentiations are color-coded. The small horizontal lines indicate the mean values per differentiation. Data were compared using nested one-way ANOVA with Sidak’s test for multiple comparisons to calculate p values. Proportions were compared using Fisher’s exact test.
Figure 3. Contribution of NaV1.8 to spontaneous diastolic SR Ca2+ release in ventricular iPSC-CM. (a) Representative surface plots showing spontaneous diastolic Ca2+ sparks (green) in ventricular iPSC-CM. (b) Mean values of Ca2+ spark frequency (CaSpF) normalized to scan width and duration. Numbers indicate the total cell count of control CM (control, n = 75 cells/4 differentiations), control cells treated with NaV1.8 inhibitor PF-01247324 (control + PF-01247324 [PF], n = 85/4), and KO of NaV1.8 after control treatment and treatment with PF-01247324 (n = 68/4 vs. 67/4). (c) Original representative line-scans of ventricular iPSC-CM illustrating a spontaneous proarrhythmogenic diastolic Ca2+ wave (green). (d) Percentage of cells exhibiting diastolic Ca2+ waves (control: 17% vs. control + PF-01247324: 7% of cells exhibiting Ca2+ waves, p < 0.05; KO: 9% vs. KO + PF-01247324: 7%). Values are presented as mean ± SEM or absolute numbers. Single cells are presented as individual symbols and different iPSC-CM differentiations are color-coded. The small horizontal lines indicate the mean values per differentiation. Data were compared using nested one-way ANOVA with Sidak’s test for multiple comparisons to calculate p values. Proportions were compared using Fisher’s exact test.
Ijms 25 06144 g003
Figure 4. NaV1.8 KO and inhibition reduce cellular arrhythmogenic triggers (a) Original traces of DADs and triggered activity in control iPSC-CM vs. SCN10A-KO iPSC-CM at 0.5 Hz. (b) Mean values ± SEM of DAD/min. (control n = 18 cells/4 differentiations; ventricular control + PF n = 11 cells/3 differentiations; SCN10A-KO control n = 10 cells/3 differentiations, SCN10A-KO + PF n = 16 cells/3 differentiations). Single cells are presented as individual symbols and different iPSC-CM differentiations are color-coded. The small horizontal lines indicate the mean values per differentiation. Data were compared using nested one-way ANOVA with Sidak’s test for multiple comparisons to calculate p values.
Figure 4. NaV1.8 KO and inhibition reduce cellular arrhythmogenic triggers (a) Original traces of DADs and triggered activity in control iPSC-CM vs. SCN10A-KO iPSC-CM at 0.5 Hz. (b) Mean values ± SEM of DAD/min. (control n = 18 cells/4 differentiations; ventricular control + PF n = 11 cells/3 differentiations; SCN10A-KO control n = 10 cells/3 differentiations, SCN10A-KO + PF n = 16 cells/3 differentiations). Single cells are presented as individual symbols and different iPSC-CM differentiations are color-coded. The small horizontal lines indicate the mean values per differentiation. Data were compared using nested one-way ANOVA with Sidak’s test for multiple comparisons to calculate p values.
Ijms 25 06144 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Hartmann, N.; Knierim, M.; Maurer, W.; Dybkova, N.; Zeman, F.; Hasenfuß, G.; Sossalla, S.; Streckfuss-Bömeke, K. NaV1.8 as Proarrhythmic Target in a Ventricular Cardiac Stem Cell Model. Int. J. Mol. Sci. 2024, 25, 6144. https://doi.org/10.3390/ijms25116144

AMA Style

Hartmann N, Knierim M, Maurer W, Dybkova N, Zeman F, Hasenfuß G, Sossalla S, Streckfuss-Bömeke K. NaV1.8 as Proarrhythmic Target in a Ventricular Cardiac Stem Cell Model. International Journal of Molecular Sciences. 2024; 25(11):6144. https://doi.org/10.3390/ijms25116144

Chicago/Turabian Style

Hartmann, Nico, Maria Knierim, Wiebke Maurer, Nataliya Dybkova, Florian Zeman, Gerd Hasenfuß, Samuel Sossalla, and Katrin Streckfuss-Bömeke. 2024. "NaV1.8 as Proarrhythmic Target in a Ventricular Cardiac Stem Cell Model" International Journal of Molecular Sciences 25, no. 11: 6144. https://doi.org/10.3390/ijms25116144

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop