Next Article in Journal
Spheroid Culture Differentially Affects Cancer Cell Sensitivity to Drugs in Melanoma and RCC Models
Next Article in Special Issue
Zingerone Modulates Neuronal Voltage-Gated Na+ and L-Type Ca2+ Currents
Previous Article in Journal
Metallic Structures: Effective Agents to Fight Pathogenic Microorganisms
Previous Article in Special Issue
Angelman Syndrome and Angelman-like Syndromes Share the Same Calcium-Related Gene Signatures
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Evidence for Inhibitory Perturbations on the Amplitude, Gating, and Hysteresis of A-Type Potassium Current, Produced by Lacosamide, a Functionalized Amino Acid with Anticonvulsant Properties

1
Department of Physiology, College of Medicine, National Cheng Kung University, Tainan City 70101, Taiwan
2
Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City 70101, Taiwan
*
Author to whom correspondence should be addressed.
These authors contributed equally.
Int. J. Mol. Sci. 2022, 23(3), 1171; https://doi.org/10.3390/ijms23031171
Submission received: 8 December 2021 / Revised: 11 January 2022 / Accepted: 19 January 2022 / Published: 21 January 2022

Abstract

:
Lacosamide (Vimpat®, LCS) is widely known as a functionalized amino acid with promising anti-convulsant properties; however, adverse events during its use have gradually appeared. Despite its inhibitory effect on voltage-gated Na+ current (INa), the modifications on varying types of ionic currents caused by this drug remain largely unexplored. In pituitary tumor (GH3) cells, we found that the presence of LCS concentration-dependently decreased the amplitude of A-type K+ current (IK(A)) elicited in response to membrane depolarization. The IK(A) amplitude in these cells was sensitive to attenuation by the application of 4-aminopyridine, 4-aminopyridine-3-methanol, or capsaicin but not by that of tetraethylammonium chloride. The effective IC50 value required for its reduction in peak or sustained IK(A) was calculated to be 102 or 42 µM, respectively, while the value of the dissociation constant (KD) estimated from the slow component in IK(A) inactivation at varying LCS concentrations was 52 µM. By use of two-step voltage protocol, the presence of this drug resulted in a rightward shift in the steady-state inactivation curve of IK(A) as well as in a slowing in the recovery time course of the current block; however, no change in the gating charge of the inactivation curve was detected in its presence. Moreover, the LCS addition led to an attenuation in the degree of voltage-dependent hysteresis for IK(A) elicitation by long-duration triangular ramp voltage commands. Likewise, the IK(A) identified in mouse mHippoE-14 neurons was also sensitive to block by LCS, coincident with an elevation in the current inactivation rate. Collectively, apart from its canonical action on INa inhibition, LCS was effective at altering the amplitude, gating, and hysteresis of IK(A) in excitable cells. The modulatory actions on IK(A), caused by LCS, could interfere with the functional activities of electrically excitable cells (e.g., pituitary tumor cells or hippocampal neurons).

1. Introduction

Lacosamide (LCS, Vimpat®) is regarded as a functionalized amino acid (available orally and intravenously) for the treatment of a wide variety of seizures, such as focal-onset seizure or refractory status epilepticus [1,2,3,4,5,6,7,8,9,10,11,12,13,14,15,16,17]. However, of note, although LCS is safe and effective in anti-convulsant activities, the unwanted events following LCS treatment, such as dizziness, abnormal vision, diplopia, ataxia, personality changes, and cardiovascular adverse events (e.g., sinus node dysfunction), have gradually emerged [3,6,7,18,19,20,21,22,23,24]. The risk of myoclonic seizure was also reported to occur during the treatment with this drug [25]. On the other hand, a previous report by Hagenacker et al. (2013) has shown the ability of LCS to reduce analgesic effects and limit the effect on INa inhibition in dorsal root ganglion neurons in a model of peripheral neuropathic pain [26]. Therefore, it is worthwhile to reappraise the ionic mechanism of LCS actions in varying types of transmembrane ionic currents, although the voltage-gated Na+ (NaV) channels were, until recently, focused on its therapeutic effectiveness [27,28,29,30,31,32].
Voltage-gated K+ (KV) channels are recognized to have essential roles in determining membrane excitability. KV channels of the KV1.4 (KCNA4), KV3.3 (KCNC3), KV3.4 (KCNC4), and KV4.1-4.3 (KCND1-3) types have been regarded to be the main determinants of A-type K+ currents (IK(A)) (i.e., fast-inactivating K+ current or transient outward K+ currents [ITO]) [33,34]. The IK(A) is viewed to be voltage-gated K+ currents that are sensitive to block by 4-aminopyridine (4-AP) and that undergo fast activation and inactivation in response to step depolarization [33,35,36,37]. It is known as an important regulator of cell excitability and firing pattern, which have been characterized in varying types of neurons, heart cells, and endocrine cells [33,34,35,36,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52]. However, whether or how LCS and other related compounds cause any adjustments on the magnitude, gating kinetics, and voltage-dependent hysteresis (Vhys) of IK(A) has not been thoroughly elaborated, although its seeming ineffectiveness in altering the magnitude of K+ currents has been previously reported [30,53].
Therefore, in light of the above-mentioned considerations, the objective of the current study is to address the question of whether LCS can cause any possible modifications on other types of membrane ionic currents, such as IK(A) in pituitary tumor (GH3) cells and mHippoE-14 hippocampal neurons, and to determine the gating kinetics of IK(A) in the presence of LCS and to evaluate whether LCS interferes with the strength of voltage-dependent hysteresis (Vhys) of IK(A) activated by isosceles-triangular ramp pulse. GH3 cells are a clonal pituitary cell line that secretes both prolactin and growth hormone and have served as a useful model for investigations into the regulation of hormonal secretion [46,52], while the mHippoE-14 hippocampal cell line is known to possess the characteristics of embryonic hippocampal neurons and to enable accurate in-vitro assays for use in the discovery, development, and validation of new therapeutics targeted to central nervous system disorders [54]. Of importance, in this study, we provide substantial evidence to disclose that the exposure to LCS is capable of interacting with KA channels to perturb the modifications on the magnitude, gating kinetics, and Vhys strength of IK(A) present in electrically excitable cells (e.g., GH3 and mHippoE-14 cells). The inhibition by LCS of INa and IK(A) may synergistically act to perturb the functional activities of excitable cells in culture or occurring in vivo.

2. Results

2.1. Inhibitory Effect of Lacosamide (LCS) on A-type K+ Current (IK(A)) Measured from Pituitary Tumor (GH3) Cells

For the first stage of whole-cell current recordings, we intentionally kept GH3 cells in Ca2+-free, Tyrode’s solution containing 1 μM tetrodotoxin (TTX) and 0.5 mM CdCl2. The measurements were conducted in the recording electrode filled with a K+-containing (145 mM) solution. In order to elicit IK(A), we voltage-clamped the examined cell at a holding potential of −80 mV, and a depolarizing pulse to −30 mV with a duration of 1 sec was thereafter applied to it. The main reason why the test potential was set at −30 mV is that as the test pulse was set beyond +10 mV, different types of high-threshold delayed-rectifier K+ currents with a slowly inactivating property could be evoked. Consequently, the effect on K+ currents exerted by LCS would have been contaminated. In keeping with previous observations in different cell types [35,36,37,50], the IK(A), which biophysically displayed a rapidly activating and inactivating property, was consistently seen in GH3 cells. Upon membrane depolarization, the current residing in these cells was noticed to activate or inactivate in a monophasic or biphasic manner, respectively. Of interest, within one minute of exposing cells to LCS, the peak and sustained amplitudes of IK(A) in response to long sustained depolarization were progressively decreased (Figure 1A). The time course of LCS-mediated inhibition of IK(A) is illustrated in the Supplementary Materials file (Figure S1). For example, as cells were exposed to LCS at a concentration of 100 μM, the peak and sustained components of IK(A) were noticeably reduced to 306 ± 23 and 85 ± 11 pA (n = 8, p < 0.05) from control values of 592 ± 32 and 191 ± 18 pA (n = 8), respectively. When LCS was removed, peak amplitude returned to 545 ± 21 pA (n = 8). As demonstrated in Figure 1A,B, the inactivation time course of IK(A) in response to long step depolarization was evidently hastened. For example, during cell exposure to 100 μM LCS, the slow component of inactivation time constant (τinact(S)) in IK(A) was profoundly declined to 297 ± 35 ms (n = 8, p < 0.05) from a control value of 1004 ± 1021 ms (n = 8); however, no obvious change in the fast component of inactivation time constant of the current was demonstrated in its presence (56 ± 9 ms (in control) versus 53 ± 11 ms (in the presence of 100 μM LCS); n = 8, p > 0.05). The results reflect that the presence of LCS is able to exert a depressant action on IK(A) in these cells.

2.2. Kinetic Estimate of LCS-Induced Block on IK(A) in GH3 Cells

During cell exposure to LCS, in addition to the measured reduction in the amplitude of peak and sustained IK(A), the slow component of the current inactivation rate during long depolarizing command voltage tended to become faster. Therefore, we set out to evaluate the inactivation time course of LCS-mediated block in situations where cells were exposed to different LCS concentrations (3–300 μM). The concentration-dependence of the slow component in the IK(A) inactivation rate that we have obtained by adding different concentrations of this compound was then constructed and is illustrated in Figure 1B. The plot demonstrates data points tightly clustered around a straight line that rises up diagonally from the horizontal axis. In other words, its blocking effect on IK(A) led to a concentration-dependent increase in the slow component of the current inactivation rate (i.e., 1/τinact(S)). As a result, the LCS-perturbed effect on IK(A) residing in GH3 cells can be reliably explained by a state-dependent blocking mechanism through which the LCS molecule can preferentially bind to and block the open state (conformation) of the A-type K+ (KA) channels. The first-order blocking scheme is shown in the following:
C α   β   O   k + 1 * · L C S k 1   O · L C S
Alternatively, the dynamical system, yielding three equations, becomes:
d C d t = α × C + β × O
d O d t = α × C + k 1 × O · L C S O × β + k + 1 * · L C S
d O · L C S d t = k 1 × O · L C S + k + 1 * · L C S × O
where α or β is the kinetic constant for the opening or closing of the KA channels underlying the IK(A), respectively; k+1* or k−1 is the blocking (i.e., depending on the LCS concentration) or unblocking rate constant of the LCS binding, respectively; and [LCS] is the LCS concentration applied. C, O, or O·[LCS] indicates the closed (or resting), open, or open-bound state of the channel, respectively.
The forward (on, k+1*) or backward (off, k−1) rate constant was reliably estimated on the basis of the slow component in the inactivation time constant (τinact(S)) of IK(A) during cell exposure to different LCS concentrations, as described under the Materials and Methods (Figure 1C). Consequently, the forward or reverse rate constant was given to be 0.0213 μM−1s−1 or 1.112 s−1, respectively; thereafter, dividing k−1 by k+1* gave a dissociation constant (KD) of 52.3 μM.
As demonstrated in Figure 1D, the concentration-dependent relationship of LCS on peak and sustained IK(A) evoked by 1 s long sustained depolarization was plotted. According to a least-squares fit to the modified Hill equation, the effective IC50 value required for LCS-mediated inhibitory effect on peak or sustained IK(A) was estimated to be 102 or 42 μM, respectively, with a Hill coefficient of 1.2. Of note, the value needed to suppress the sustained IK(A) amplitude in GH3 cells was nearly close to the KD value computed on the basis of the first-order reaction scheme (Figure 1C), although the IC50 value for its inhibition of peak IK(A) was higher than the KD value (i.e., a two-fold increase).

2.3. Effect of LCS, Tetraethylammonium Chloride (TEA), 4-Aminopyridine-3-methanol (4-AP-3-MeOH), 4-Aminopyridine (4-AP), Capsaicin (Cap) on the Amplitude of IK(A) in GH3 Cells

We extended to test how the IK(A) magnitude in these cells can be affected by different blockers of KA channels. This set of experiments was conducted when the examined cells were depolarized from −80 to −30 mV with a duration of 1 s, and we thereafter measured current amplitude at the start of command voltage during cell exposure to different compounds. As shown in Figure 2, the presence of TEA (5 mM) failed to decrease the peak amplitude of IK(A) in GH3 cells, while that of LCS (100 μM), 4-AP-3-MeOH (100 μM), 4-AP (5 mM), or Cap (30 μM) suppressed peak IK(A) effectively. 4-AP-3-MeOH, a derivative of 4-AP, has been previously demonstrated to inhibit IK(A) and to restore axonal conduction in injured spinal cord white matter [55,56], while Cap was reported to inhibit IK(A) in heart cells [36,38]. Hence, similar to the effects of 4-AP, 4-AP-3-MeOH, or Cap, the ability of LCS to suppress the peak component of IK(A) is clearly demonstrated in GH3 cells.

2.4. Effect of LCS on the Current versus Voltage (I–V) Relationship of IK(A) Recorded from GH3 Cells

To characterize the inhibitory effect of LCS on peak and sustained IK(A), we continued to explore whether this drug might exert any adjustments on the steady-state I–V relationship of peak IK(A) inherently in these cells. The experiments were conducted in a voltage-clamp mode with a holding potential of −80 mV, and different rectangular voltages, ranging between −80 and +10 mV with a duration of 1 s, were subsequently applied to the examined cell. Figure 3A depicts representative IK(A) traces taken with or without the application of LCS at a concentration of 100 μM, and the mean I–V relationships of peak or sustained IK(A) in the absence and presence of this drug were established and are depicted in Figure 3B,C, respectively. The data indicated that no obvious adjustments in the overall I–V relationship of peak or sustained IK(A) were detected during GH3-cell exposure to LCS, although the conductance of peak or sustained IK(A) obtained in its presence was greatly reduced throughout the entire range of clamping potentials applied.

2.5. Steady-State Inactivation Curve of IK(A) Obtained in the Absence and Presence of LCS

To further characterize the inhibitory effect of LCS on IK(A), we tested the voltage dependence of LCS effect on IK(A) in GH3 cells by using a two-step voltage protocol. Figure 4A,B show the steady-state inactivation curve of IK(A), taken with or without the application of 100 μM LCS. This set of current measurements was conducted with the voltage protocol in which a 1-s conditioning pulse to different potentials was applied to precede the 1-s test pulse to −30 mV from a holding potential of −80 mV. We then constructed the relationship between the conditioning potential and the normalized amplitude of peak IK(A). The experimental data sets were collected and then least-squares fitted with a Boltzmann function (detailed in Materials and Methods). The values of either V1/2 or q (i.e., apparent gating charge) in the absence and presence of 100 μM LCS are either −71.4 ± 2.1 and −65.2 ± 1.9 mV or 10.2 ± 0.8 and 10.5 ± 0.9 e (n = 7), respectively. From these results, we were, therefore, able to show that the application of 100 μM LCS significantly shifted the midpoint of the inactivation curve toward the depolarizing voltage by approximately 6 mV (p < 0.05); conversely, no modifications in the gating charge of the curve were detected in the presence of this drug. Thus, in GH3 cells, there was a voltage dependence of the steady-state inactivation curve of IK(A) during exposure to LCS.

2.6. Effect of LCS on the Recovery of IK(A) Block Measured from GH3 Cells

In attempts to evaluate LCS-mediated inhibition of peak IK(A) in these cells, we further investigated the recovery time course of current inactivation acquired with or without the application of this drug. For this set of current recordings, we implemented another two-pulse protocol that comprises a first (conditioning pulse) depolarizing voltage command and a second depolarizing command (test pulse) applied following varying inter-pulse intervals (Figure 5). The ratios of the peak IK(A) in response to the second and first pulses were then taken as a measure of recovery from the IK(A) block, and they were constructed and plotted versus inter-pulse intervals. The results, showing IK(A) recovery from the block in the absence or presence of 100 μM LCS, are illustrated in Figure 5A,B. The time course, taken either from the control period (i.e., LCS was not present) or during exposure to 100 μM LCS, could be optimally described by a single exponential function with the recovery time constant of 1.01 ± 0.05 s (n = 8) or 1.66 ± 0.04 s (n = 8), respectively. It is, therefore, plausible to reflect, from the present observations, that the recovery of the IK(A) block was overly prolonged by cell exposure to LCS. The delayed recovery of the IK(A) block caused by LCS presence was most likely attributed to the open channel block.

2.7. Effect of LCS on Voltage-Dependent Hysteresis (Vhys) of IK(A) Identified in GH3

A number of studies have demonstrated the ability of IK(A) strength to have impacts on varying firing patterns or action potential configurations existing in different types of electrically excitable cells [33,34,36,38,43,44,45,47,48,50,51,52,55]. For these reasons, we sought to examine the presence of IK(A)-linked Vhys and to explore whether cell exposure to LCS is capable of modifying the Vhys extent of IK(A) activated in response to inverted isosceles-triangular ramp pulse with varying durations. In these experiments, in the absence and presence of LCS, we held the examined cell at −80 mV and a downsloping (forward) limb from +10 to −80 mV, followed by an upsloping (backward) limb back to +10 (i.e., inverted isosceles-triangular ramp pulse), with varying durations (600 or 800 ms) thereafter imposed on it (Figure 6A1,A2). Under these experimental conditions, the voltage-dependent hysteresis (Vhys) of IK(A) elicited by such a ramp waveform was robustly observed. Of note, the instantaneous IK(A) amplitudes measured at the descending (ramping from +10 to −80 mV) limb of the triangular ramp were actually higher than those at the equipotential level of the ascending (ramping from −80 to +10 mV) limb of the ramp, especially at the voltages ranging between −40 and +10 mV (Figure 6B). The inactivation process of the current resulted in residual currents at the end of the ramp pulse, and the I–V loop in Figure 6B tended to be open when the voltage was ramped back to +10 mV.
Additional, during cell exposure to 30 or 100 μM LCS, the Vhys strength (i.e., Δarea (the difference in the area encircled by the curve in the descending and ascending direction)) of IK(A) responding to both descending and ascending limbs of inverted triangular ramp pulse was progressively declined with increasing LCS concentration (Figure 6B,C). For example, as the duration of isosceles-triangular ramp applied was set at 800 ms (i.e., the ramp speed of ±112.5 mV/s), the value of Δarea for the Vhys emergence in the control period (i.e., LCS was not present) was 14.55 ± 0.72 nA·mV (n = 8), while in the same duration, the Δarea value was substantially declined to 6.13 ± 0.32 nA·mV during cell exposure to 100 μM LCS (n = 8, p < 0.05). Findings from these results, therefore, prompted us to indicate that there was apparently the emergence of Vhys for IK(A) activation in response to an inverted isosceles-triangular ramp pulse in GH3 cells and that the hysteretic strength of the current became evidently diminished with increasing LCS concentration.

2.8. Inhibitory Effect of LCS on IK(A) Present in mHippoE-14 Neurons

In a final set of whole-cell recordings, we decided to examine whether IK(A) was functionally expressed in other types of cells (e.g., mHippoE-14 neurons) and how its magnitude or gating can be influenced by the presence of LCS. Similar to the experimental protocol done above in GH3 cells, cells were bathed in Ca2+-free, Tyrode’s solution containing 1 μM TTX and 0.5 mM CdCl2. As demonstrated in Figure 7, when the examined cell was 1-s depolarized from −80 to −30 mV, the IK(A) with a rapid activating and inactivating property was robustly identified. This current was subject to inhibition by 5 mM 4-AP. Noticeably, when mHippoE-14 neurons were continually exposed to LCS (30 or 100 μM), the peak or sustained amplitude of IK(A) in response to 1-s membrane depolarization from −80 to −30 mV was reduced. Concurrently, the LCS’s inhibition was accompanied by an evident reduction in the τinact(S) value of IK(A), with no change in the fast component of the inactivation time constant. For example, one minute after LCS (100 μM) was added, sustained IK(A) amplitude was significantly reduced from 24.2 ± 4.2 to 7.8 ± 1.1 pA (n = 8, p < 0.05) and the τinact(S) value was concurrently diminished from 509 ± 32 to 169 ± 15 ms (n = 8, p < 0.05). After washout of the drug, sustained IK(A) was restored to 22.9 ± 3.9 pA (n = 7, p < 0.05). As such, consistent with the experimental observations described above in GH3 cells, we observed that the IK(A) found in mHippoE-14 neurons was readily evoked in response to long sustained depolarization and that the current was sensitive to block by LCS.

3. Discussion

In the current study, we found that the presence of LCS could depress IK(A) in a concentration-, time-, state-, and hysteresis-dependent manner in GH3 cells. The block of IK(A) produced by LCS apparently is noted to be not inherently instantaneous, but it develops over time after the KA channels become opened; thereafter, such a block produces a concurrent rise in the inactivation rate of the current activated by long-lasting membrane depolarization. During cell exposure to LCS, the IK(A) exhibited a blunted peak and hastened decay, which is consistent with the possibility that the opening of channels was retarded by the binding of LCS. It is thus most likely that the blocking site of this drug is located within the channel pore only when the KA channel is opened.
Distinguishable from previous observations [30,53], the present results demonstrated that LCS exerted a differential depressant action on the peak and sustained IK(A) natively expressed in GH3 cells. IK(A) in these cells is biophysically characterized by a rapidly activating and inactivating property during depolarizing voltage command, and it is sensitive to blockage by 4-AP, 4-AP-3-MeOH, or Cap but not by TEA (Figure 2). LCS tends to be selective for sustained over peak IK(A) in response to a long depolarizing pulse. The rate of IK(A) inactivation was considerably enhanced as the LCS concentration increased. Moreover, according to quantitative estimates from a heuristic reaction scheme elaborated above, the KD (i.e., k−1/k+1*) value on the basis of the LCS-perturbed inactivation time constant of IK(A) (i.e., τinact(S)) seen in GH3 cells was yielded to be 52.3 μM. This value agrees closely with the calculated IC50 value (42 μM), which was needed for LCS-mediated inhibition of sustained IK(A); however, it is apparently lower than the IC50 (102 μM) required for LCS’s ability to suppress peak IK(A) (Figure 1). Therefore, the results strongly reflect that there is a considerable and selective block of sustained IK(A) in the presence of LCS.
As reported previously, the effect of LCS on hNaV1.5 Na+ channels had an inhibitory effect, with a IC50 value of around 70–80 µM [29]. The estimated IC50 values required for inhibition by LCS of NaV1.7-, NaV1.3-, and NaV1.8-encoded currents were reported to be 182, 415, and 16 µM, respectively [57]. The IC50 value for LCS-mediated inhibition of IK(A) was thus noticed to overlap those for inhibition of the different isoforms of NaV channels. A previous report also showed that the mean LCS level in serum or CSF was around 8.2 µg/mL (33 µM) or 7.4 µg/mL (29 µM), respectively [58]. It is reasonable to assume, therefore, that the inhibitory effect on IK(A) caused by LCS could be of pharmacological or therapeutic relevance.
Another point that needs to be explored is that the inactivation time course of the IK(A) obtained in the presence of LCS was noticed to become faster in a concentration-dependent manner, although the initial rising phase of the current (i.e., activation time course) remained unaffected. That is, during exposure to LCS, the inactivation time course of IK(A), especially in the slow component, had the propensity to decay more rapidly, although the fast component of current inactivation in response to long step depolarization failed to be changed. This feature can therefore be incorporated into a simple kinetic scheme (i.e., closed ↔ open ↔ open-bound), as stated above in the Results section. The results, in turn, mean that aside from its inhibition of peak IK(A), the LCS molecule may primarily interact on the activation and/or inactivation process, presumably resulting in the adjustments of the magnitude and gating kinetics of the current, although the detailed underlying ionic mechanism of its action on IK(A) remains to be delineated. However, the possibility that LCS may have a higher affinity for the open conformation of KA channels than the closed (resting) conformation without evoking open channel block cannot be excluded.
The nonlinear Vhys phenomenon inherently in different types of voltage-gated ionic currents has been since demonstrated to play roles in affecting various electrical behavior of excitable cells [59,60]. In this study, we were able to identify the Vhys existence of IK(A) residing in GH3 cells (Figure 6), implying that the voltage sensitivity inherent in the gating charge movement of KA channels is thought to rely on the previous state of the channel [61,62]. In other words, the magnitude of IK(A) is most likely to be contingent on the pre-existing state(s) or conformation(s) of the KA channel. The Vhys strength of IK(A) is important, and it would be engaged in the regulation of electrical behaviors of excitable cells such as GH3 cells [62,63]. In other words, as the action potential develops, the voltage dependence of KA channels may shift the mode of Vhys to one in which activation occurs at more negative potentials, resulting in an increase in membrane repolarization, while as the membrane becomes negative, the voltage-dependence of IK(A) activation would switch to more positive voltages, therefore enhancing cell excitability [44,61].
In this study, we additionally evaluated the possible perturbations by LCS of the instantaneous and non-equilibrium property inherent in IK(A) activated by an inverted isosceles-triangular ramp pulse. The experimental observations that we have obtained led us to unravel the LCS’s effectiveness in reducing the Vhys-linked Δarea for IK(A) elicited by a triangular ramp pulse, suggesting that Vhys behavior is engaged in the voltage-dependent activation of the current. During the exposure to LCS, the steady-state inactivation curve of IK(A) was shifted to less hyperpolarized potential (Figure 4), and the recovery of the current block also became slowed (Figure 5). In this scenario, it is reasonable to assume that any modifications by LCS of IK(A) depend not only on the LCS concentration given but also on various confounding factors, such as the pre-existing level of the resting potential and the different firing patterns of action potentials or their combinations, assuming the magnitude of IK(A) is sufficiently present in the cells examined.
From the present observations, we show that LCS shifted the inactivation curve of IK(A) in the rightward direction, suggesting that it interacts with channels in the inactivated state. Consequently, neither the resting membrane potential nor the magnitude of INa could be seriously altered by its inhibitory effect on IK(A), although it per se inhibited INa directly [27,28,29,30,31,32,57]. It has been established that the KV channels, such as KA channels, are crucial in shaping action potentials. Indeed, as its subthreshold activation and transient inactivation, the overall properties of IK(A) make it an excellent target for any modulatory mechanism influencing membrane excitability and action potential firing. While the detailed mechanism of the LCS-induced suppression of IK(A) remains unknown, our study suggests that the LCS-induced decrease in IK(A) might increase excitability in electrically excitable cells, possibly leading to changes in neural plasticity [33,34,35,36,39,40,41,42,43,44,45,46,47,48,49,50,51,52].
The amplitude of IK(A) observed in GH3 or mHippoE14 cells is sensitive to inhibition by 4-AP or 4-AP-3-MeOH, yet not by TEA. The KA-channel currents demonstrated here are thus primarily coded by genes from the KV4 (KCND) subfamily (e.g., KV4.2 and KV4.3) [35,42,46,64,65], although it is also likely that KV1.4, KV3.3, KV3.4, KV4.1, KV4.2, and KV4.3 contribute to the formation of KA channels [46,52,65].
In this study, the IK(A) residing in mHippoE-14 hippocampal neurons was also observed to be sensitive to blockage by LCS, and the inactivation time course of the current in response to long sustained depolarization was accompanied by a rapid decay. (Figure 7). The magnitude of IK(A) has been previously demonstrated to be engaged in cell excitability, network synchronicity, and convulsant properties [33,34,43,45,47,48,49,51,55]. As a result, it is plausible to assume, from the present observations, that the LCS-mediated inhibition of IK(A) would be of clinical, therapeutic, pharmacological, or even toxicological relevance, although a previous report showed that neither hERG nor L-type Ca2+ currents were altered by LCS [66]. The extent to which the unwanted reactions linked to the LCS treatment was explained by its perturbations on the magnitude, gating, and Vhys of IK(A) and thus warrants further investigation. Findings from the present study tend to be informative as they highlight the proof-of-concept that needs to be taken into consideration since the wide spectrum of LCS’s beneficial effects has been clinically observed [1,2,3,4,5,6,7,9,10,11,12,13,14,15,16,17,18,19,20,21,24,25,32].

4. Materials and Methods

4.1. Chemicals, Drugs, and Solutions Used in This Study

Lacosamide (LCS, erlosamide, harkoseride, SPM 927, ADD 234034, Vimpat®, R-enantiomer of 2-acetamido-N-benzyl-3-methoxy-propioniamide, 2,3-diaminomaleonitrile, C13H18N2O3; https://pubchem.ncbi.nlm.nih.gov/compound/Lacosamide accessed on 10 January 2022), 4-aminopyridine (4-AP), capsaicin (Cap), tetraethylammonium chloride (TEA), and tetrodotoxin (TTX) were supplied by Sigma-Aldrich (Merck, Taipei, Taiwan), while 4-aminopyridine-3-methanol (4-AP-3-MeOH) was provided by Alfa Aesar (Uni-onward Corp., Tainan, Taiwan). For cell preparations, culture media, fetal bovine serum, L-glutamine, trypsin/EDTA, and penicillin–streptomycin were acquired from HyCloneTM (Thermo Fisher, Kaohsiung, Taiwan), whereas all other chemicals, such as aspartic acid, CdCl2, EGTA, and HEPES, were of laboratory grade and taken from standard sources.
The ion composition of the extracellular solution, (i.e., normal Tyrode’s solution buffered by HEPES) used for this study was as follows (in mM): NaCl 136.5, KCl 5.4, CaCl2 1.8, MgCl2 0.53, glucose 5.5, and HEPES-NaOH buffer 5 (pH 7.4). For the recordings of the flow through macroscopic IK(A), we kept cells bathed in Ca2+-free, Tyrode’s solution in order to avoid the contamination of Ca2+-activated K+ and voltage-gated Ca2+ currents, while the patch electrodes for recording were backfilled with the following intracellular solution (in mM): K-aspartate 130, KCl 20, MgCl2 1, KH2PO4 1, Na2ATP3, Na2GTP 0.1, EGTA 0.1, and HEPES-KOH buffer 5 (pH 7.2). To record voltage-gated Na+ current (INa), we substituted K+ ions in the internal solution for equimolar Cs+ ions, and the pH value in the solution was titrated to 7.2 by adding CsOH. To avoid the contamination of Cl currents, Cl ions inside the pipette solution were replaced with aspartate. All solutions used for this study were generally prepared in deionized water from a Milli-Q® water purification system (Merck Millipore, Taipei, Taiwan). The pipette solution and culture media were always filtered with an Acrodisc® syringe filter that contains a 0.2-μm Supor® nylon membrane (#4612; Pall Corp.; Genechain Biotechnology, Kaohsiung, Taiwan).

4.2. Cell Preparations

The GH3 pituitary cell line was supplied by the Bioresources Collection and Research Center (BCRC-60015; Hsinchu, Taiwan), whereas the embryonic mouse hippocampal cell line (mHippoE-14, CLU198) was by Codarlane CELLutions Biosystems, Inc. (Burlington, ON, Canada). The GH3 cell line was originally derived from the American Type Culture Collection (ATCC® [CCL-82.1TM]; Manassas, VA, USA). The GH3 cell line was maintained by growing cells in 50-mL plastic culture flasks in 5 mL of Ham’s F-12 medium, supplemented with 2.5% fetal calf serum (v/v), 15% horse serum (v/v), and 2 mM L-glutamine, while mHippoE-14 neurons were in Dulbecco’s modified Eagle’s medium, supplemented with 10% fetal bovine serum (v/v) and 2 mM L-glutamine. Flasks were kept at 37 °C in a humidified environment of 5% CO2/95% air. Growth medium was replaced twice a week, and cells were split into subcultures once a week. Electrophysiological measurements were made 6–10 days after the plating of the subcultures.

4.3. Electrophysiological Measurements

On the day of the experiments, we dispersed GH3 cells or mHippoE-14 neurons with 1% trypsin/EDTA solution, and a few drops of cell suspension was placed in a home-made recording chamber positioned on the stage of an inverted Olympus fluorescence microscope (CKX-41; Yuan Yu, Taipei, Taiwan). The microscope was set on an antI–Vibration air table contained within a Faraday’s cage, and it was also coupled to a digital video system (DCR-TR30; Sony, Tokyo, Japan), with a magnification of up to 1500×, to continuously monitor changes in both cell size and the electrode’s position. Cells were kept immersed at room temperature (20–25 °C) in normal Tyrode’s solution containing 1.8 mM CaCl2, and the solution’s composition is elaborated above. After the cells were allowed to adhere to the chamber’s bottom for several minutes, the recordings were carried out. The patch-clamp procedure in whole-cell mode (i.e., when membrane patch was broken by suction) was performed by using either an RK-400 (Biologic, Echirolles, France) or an Axopatch-200 amplifier (Molecular Devices; Bestogen Biotech, New Taipei City, Taiwan) [67]. When filled with internal solution, the pipettes used for recording had tip resistances ranging from 3 to 5 MΩ, and they were fabricated from Kimax-51 borosillicate capillaries (#34500 [1.5–1.8 mm in outer diameter]; Dogger, Tainan, Taiwan) by using either a PP-830 vertical puller (Narishige, Tokyo, Japan) or a P-97 programmable horizontal puller (Sutter, Novato, CA, USA) and the electrodes’ tips were fire-polished with an MF-83 microforge (Narishige). The potentials were corrected for the liquid-liquid junction potential, which develops when the composition of the pipette solution is different from that in the bath. Tested compounds were either applied through perfusion or added to the bath in the attempts to achieve the final concentration indicated.

4.4. Data Recordings

The signals were monitored on an HM-507 oscilloscope (Hameg, East Meadow, NY, USA) and digitally stored online in an ASUS ExpertBook laptop computer (P2451F; ASUS, Tainan, Taiwan) at 10 kHz, interfaced with a Digidata 1440A converter (Molecular Devices; Bestogen Biotech, New Taipei City, Taiwan). The latter device was used for efficient analog-to-digital/digital-to-analog (AD/DA) conversion. During the measurements, the process in data acquisition, equipped with this device, was controlled by pCLAMP 10.6 software (Molecular Devices) run under Windows 7 (Redmond, WA, USA), and the signals were simultaneously displayed on an LCD monitor through a USB type-C connection. Current signals that we achieved were low-pass-filtered at 2 kHz with an FL-4 four-pole Bessel filter (Dagan, Minneapolis, MN, USA) to minimize possible electrical interference. After the recorded data were digitally acquired, we off-line collated them using various analytical tools that included the LabChart 7.0 program (ADInstruments; Gerin, Tainan, Taiwan), OriginPro® 2021 (OriginLab; Scientific Formosa, Kaohsiung, Taiwan), and different custom-made macros built in Excel® 2021 under Office 365 (Redmond, WA, USA). pCLAMP-generated voltage-clamp profiles, in which either rectangular or ramp waveforms were created, were employed to examine the current-voltage (I–V) relationship, the steady-state inactivation curve, the recovery time course of current inactivation, or the voltage-dependent hysteresis (Vhys) of ionic currents (e.g., IK(A)).

4.5. Whole-Cell Current Analyses

To determine the concentration-dependent inhibition of LCS on the peak or sustained component of macroscopic IK(A), GH3 cells were immersed in Ca2+-free, Tyrode’s solution in which 1 μM TTX and 0.5 mM CdCl2 were added. The examined cell was held at −80 mV and 1 s depolarizing step from −80 until a clamping potential of −30 mV was delivered to it. Current amplitudes (at the beginning- and end-pulse of command voltage) evoked in response to depolarizing pulses to −30 mV were acquired in the control period (i.e., LCS was not present) and during the exposure to varying LCS concentrations (3 μM–1 mM). The concentration required to suppress 50% (i.e., IC50) of peak or sustained IK(A) was calculated with the goodness of fit by the use of a modified form of the Hill equation. That is,
R e l a t i v e   a m p l i t u d e = L C S n H × 1 a I C 50 n H + L C S n H + a
where [LCS] represents the different concentrations of LCS used; nH and IC50 are, respectively, the Hill coefficient inherent to the concentration–response response and the concentration at which 50% inhibition of peak or sustained IK(A) is observed. At this point, maximal inhibition (1−a) of peak or sustained IK(A) is also evaluated.
The time-dependent rate constants of the blocking (forward, on, k+1*) or unblocking (backward, off, k−1) reaction, taken with or without the application of different LCS concentrations, were evaluated from the slow component of the inactivation time constant (τinact(S)) of IK(A) evoked by depolarizing command voltage from −80 to −30 mV with a duration of 1 s. The τinact(S) values obtained in the presence of different LCS concentrations were approximated by fitting a two-exponential function (i.e., fast and slow components) to the decaying trajectory of each current trace. Since a Hill coefficient of about 1 was noticed according to the concentration-dependent relationship, the forward or backward rate constant was then extended, as determined using the following equation (i.e., simple linear regression model):
1 τ i n a c t S = k 1 + k + 1 * · L C S
in which [LCS] is the known concentration of LCS, and k+1* or k−1 is respectively achieved from the slope (i.e., Δ(1/τinact(S))/Δ[LCS]) or the vertical y-axis intercept at [LCS] = 0 of the interpolated regression line (i.e., the point at which the line crosses the y-axis), where the relation of the reciprocal time constant of IK(A) inactivation (i.e., 1/τinact(S)) versus the LCS concentration was constructed.
The relationship of the conditioning potential versus the IK(A) amplitude acquired with or without the LCS addition (i.e., quasi-steady-state inactivation curve of IK(A)) was approximated by a modified Boltzmann function (or the Fermi-Dirac distribution) of the following form:
I I m a x = 1 1 + e x p ( V V 1 2 ) q F R T
where Imax is the maximal amplitude of IK(A), V1/2 the voltage (in mV) at which half-maximal inactivation of the current is achieved, q the apparent gating charge in e (i.e., elementary charge), and F, R, and T the Faraday’s constant, the universal gas constant, and the absolute temperature, respectively.

4.6. Curve-Fitting Procedures and Statistical Analyses

Linear (e.g., relation of 1/τinact(S) versus the LCS concentration) or nonlinear (e.g., Hill or Boltzmann equation and double exponential) curve fitting to the experimental data sets presented herein was performed with the least-squares fit by using either the Solver add-in bundled with Excel® 2021 (Microsoft) or OriginPro® 2021 (OriginLab). The values are provided as means ± standard error of mean (SEM) with sample sizes (n), which denotes the cell number carefully collected. Student’s t-test (paired or unpaired samples) or analysis of variance (ANOVA-1 or ANOVA-2), followed by post-hoc Fisher’s least-significance difference test for multiple-range comparisons, was implemented for the statistical evaluation. Probability with p < 0.05 was considered statistically significant.

Supplementary Materials

The following are available online at www.mdpi.com/article/10.3390/ijms23031171/s1.

Author Contributions

Conceptualization, S.-N.W., H.-Y.C. and T.-H.C.; methodology, S.-N.W.; software, S.-N.W.; validation, H.-Y.C., T.-H.C. and S.-N.W.; formal analysis, S.-N.W.; investigation, H.-Y.C., T.-H.C. and S.-N.W.; resources, S.-N.W.; data curation, S.-N.W.; writing—original draft preparation, S.-N.W.; writing—review and editing, H.-Y.C., T.-H.C. and S.-N.W.; visualization, H.-Y.C., T.-H.C. and S.-N.W.; supervision, S.-N.W.; project administration, S.-N.W.; funding acquisition, S.-N.W. All authors have read and agreed to the published version of the manuscript.

Funding

This work was in part supported by a grant awarded by the Ministry of Science and Technology, Taiwan (MOST-110-2320-B-006-028) to SNW. The funders in this work are not involved in the study design, data collection, analyses, or interpretation.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The datasets used in this study are available on reasonable request.

Acknowledgments

The authors are grateful to Zi-Han Gao for the earlier work in the present study. The authors would like to thank Ching-Hsing Luo (Institute of Medical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan) for helpful encouragement during the initial stages of this work.

Conflicts of Interest

The authors declare no conflict of interest that is directly relevant to this study.

Abbreviations

ANOVA  analysis of variance
4-AP  4-aminopyridine
4-AP-3-MeOH  4-aminopyridine-3-methanol
Cap  capsaicin
IK(A)  A-type K+ current
I–V  current voltage
IC50  the concentration required for 50% inhibition
KA  A-type K+ channel
KD  dissociation constant
KV channel  voltage-gated K+ channel
LCS  lacosamide (Vimpat®)
NaV channel  voltage-gated Na+ channel
SEM  standard error of mean
TEA  tetraethylammonium chloride
τinact(S)  time constant in the slow component of current inactivation
TTX  tetrodotoxin
Vhys  voltage-dependent hysteresis

References

  1. Doty, P.; Rudd, G.D.; Stoehr, T.; Thomas, D. Lacosamide. Neurotherapeutics 2007, 4, 145–148. [Google Scholar] [CrossRef] [PubMed]
  2. Harris, J.A.; Murphy, J.A. Lacosamide and epilepsy. CNS Neurosci. Ther. 2011, 17, 678–682. [Google Scholar] [CrossRef] [PubMed]
  3. Strzelczyk, A.; Zöllner, J.P.; Willems, L.M.; Jost, J.; Paule, E.; Schubert-Bast, S.; Rosenow, F.; Bauer, S. Lacosamide in status epilepticus: Systematic review of current evidence. Epilepsia 2017, 58, 933–950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Hoy, S.M. Lacosamide: A Review in Focal-Onset Seizures in Patients with Epilepsy. CNS Drugs 2018, 32, 473–484. [Google Scholar] [CrossRef]
  5. Ortiz de la Rosa, J.S.; Ladino, L.D.; Rodríguez, P.J.; Rueda, M.C.; Polanía, J.P.; Castañeda, A.C. Efficacy of lacosamide in children and adolescents with drug-resistant epilepsy and refractory status epilepticus: A systematic review. Seizure 2018, 56, 34–40. [Google Scholar] [CrossRef] [Green Version]
  6. Rosenow, F.; Brandt, C.; Bozorg, A.; Dimova, S.; Steiniger-Brach, B.; Zhang, Y.; Ferrò, B.; Holmes, G.L.; Kälviäinen, R. Lacosamide in patients with epilepsy of cerebrovascular etiology. Acta Neurol. Scand. 2020, 141, 473–482. [Google Scholar] [CrossRef] [Green Version]
  7. Vossler, D.G.; Knake, S.; O’Brien, T.J.; Watanabe, M.; Brock, M.; Steiniger-Brach, B.; Williams, P.; Roebling, R. Efficacy and safety of adjunctive lacosamide in the treatment of primary generalised tonic-clonic seizures: A double-blind, randomised, placebo-controlled trial. J. Neurol. Neurosurg. Psychiatry 2020, 91, 1067–1075. [Google Scholar] [CrossRef]
  8. Alkhotani, A.; Abualula, H.; Almatrafi, Y.; Ghomiem, S.; Alaa, A. Efficacy and Tolerability of Lacosamide for Focal Epileptic Patients: Study from Epilepsy Clinic in Makkah. CNS Neurol. Disord. Drug Targets 2021, 20, 87–97. [Google Scholar] [CrossRef]
  9. Babar, R.K.; Bresnahan, R.; Gillespie, C.S.; Michael, B.D. Lacosamide add-on therapy for focal epilepsy. Cochrane Database Syst. Rev. 2021, 5, Cd008841. [Google Scholar] [CrossRef]
  10. Ben-Menachem, E.; Dominguez, J.; Szász, J.; Beller, C.; Howerton, C.; Jensen, L.; McClung, C.; Roebling, R.; Steiniger-Brach, B. Long-term safety and tolerability of lacosamide monotherapy in patients with epilepsy: Results from a multicenter, open-label trial. Epilepsia Open 2021, 6, 618–623. [Google Scholar] [CrossRef]
  11. Casciato, S.; Quarato, P.P.; Gialluisi, A.; D’Aniello, A.; Mascia, A.; Grammaldo, L.G.; Di Gennaro, G. Lacosamide as first add-on or conversion monotherapy: A retrospective real-life study. Epilepsy Behav. 2021, 122, 108128. [Google Scholar] [CrossRef]
  12. Eilam, A.; Khmeliov, N.; Penker, D.; Gilad, R. Intravenous Lacosamide in Seizure Clusters: Dose and Efficacy. Clin. Neuropharmacol. 2021, 44, 85–88. [Google Scholar] [CrossRef]
  13. Inoue, Y.; Liao, W.; Wang, X.; Du, X.; Tennigkeit, F.; Sasamoto, H.; Osakabe, T.; Hoshii, N.; Yuen, N.; Hong, Z. Safety and efficacy of adjunctive lacosamide in Chinese and Japanese adults with epilepsy and focal seizures: A long-term, open-label extension of a randomized, controlled trial. Epilepsy Res. 2021, 176, 106705. [Google Scholar] [CrossRef]
  14. Mascolo, A.P.; Marrama, F.; Mercuri, N.B.; Placidi, F. Efficacy and safety of lacosamide in the treatment of status epilepticus in a patient with comorbidities. Acta Biomed. 2021, 92, e2021090. [Google Scholar] [CrossRef]
  15. Panda, P.K.; Panda, P.; Dawman, L.; Sharawat, I.K. Efficacy of lacosamide and phenytoin in status epilepticus: A systematic review. Acta Neurol. Scand. 2021, 144, 366–374. [Google Scholar] [CrossRef]
  16. Rohracher, A.; Kalss, G.; Kuchukhidze, G.; Neuray, C.; Leitinger, M.; Höfler, J.; Kreidenhuber, R.; Rossini, F.; Volna, K.; Mauritz, M.; et al. New anti-seizure medication for elderly epilepsy patients—A critical narrative review. Expert Opin. Pharm. 2021, 22, 621–634. [Google Scholar] [CrossRef]
  17. Shin, Y.W.; Moon, J.; Cho, Y.W.; Kim, D.W.; Hong, S.B.; Kim, D.Y.; Chang, H.; Yoon, S.H.; Yu, K.S.; Jang, I.J.; et al. Tolerability of lacosamide rapid dose titration: A randomized, multicenter, prospective, open-label study. Epilepsy Behav. 2021, 115, 107663. [Google Scholar] [CrossRef]
  18. Li, J.; Sun, M.; Wang, X. The adverse-effect profile of lacosamide. Expert Opin. Drug. Saf. 2020, 19, 131–138. [Google Scholar] [CrossRef]
  19. Hamard, J.; Rigal, M.; Gony, M.; Bagheri, H. Lacosamide-induced personality changes: An unexpected adverse effect. Fundam. Clin. Pharmacol. 2021, 36, 224–226. [Google Scholar] [CrossRef]
  20. Kim, H.K.; Lee, H.; Bae, E.K.; Kim, D.W. Cardiac effects of rapid intravenous loading of lacosamide in patients with epilepsy. Epilepsy Res. 2021, 176, 106710. [Google Scholar] [CrossRef]
  21. Kishi, T.; Nomura, I.; Sakuma, K.; Okuya, M.; Oya, K.; Iwata, N. The Efficacy and Safety of Lacosamide for Bipolar Depression: A 12-Week Open-Label Pilot Trial. J. Clin. Psychopharmacol. 2021, 41, 204–206. [Google Scholar] [CrossRef] [PubMed]
  22. Minomo, S.; Amino, T.; Hara, N.; Ichijo, M.; Miyake, T.; Yasuura, A.; Kamata, T. Lacosamide-induced symptomatic sinus node dysfunction. Epileptic Disord. 2021, 23, 772–774. [Google Scholar] [CrossRef]
  23. Shibata, M.; Hoshino, R.; Shimizu, C.; Sato, M.; Furuta, N.; Ikeda, Y. Lacosamide-induced sinus node dysfunction followed by severe agranulocytosis. BMC Neurol. 2021, 21, 217. [Google Scholar] [CrossRef]
  24. Yang, C.; Peng, Y.; Zhang, L.; Zhao, L. Safety and Tolerability of Lacosamide in Patients With Epilepsy: A Systematic Review and Meta-Analysis. Front. Pharm. 2021, 12, 694381. [Google Scholar] [CrossRef]
  25. Bosak, M.; Wężyk, K.; Słowik, A. Lacosamide and myoclonic seizures: What is the risk of aggravation? Neurol. Neurochir. Pol. 2021, 55, 107–109. [Google Scholar] [CrossRef]
  26. Hagenacker, T.; Schäfer, N.; Büsselberg, D.; Schäfers, M. Analgesic ineffectiveness of lacosamide after spinal nerve ligation and its sodium channel activity in injured neurons. Eur. J. Pain 2013, 17, 881–892. [Google Scholar] [CrossRef]
  27. Beyreuther, B.K.; Freitag, J.; Heers, C.; Krebsfänger, N.; Scharfenecker, U.; Stöhr, T. Lacosamide: A review of preclinical properties. CNS Drug Rev. 2007, 13, 21–42. [Google Scholar] [CrossRef]
  28. Errington, A.C.; Stöhr, T.; Heers, C.; Lees, G. The investigational anticonvulsant lacosamide selectively enhances slow inactivation of voltage-gated sodium channels. Mol. Pharmacol. 2008, 73, 157–169. [Google Scholar] [CrossRef] [Green Version]
  29. Wang, G.K.; Wang, S.Y. Block of human cardiac sodium channels by lacosamide: Evidence for slow drug binding along the activation pathway. Mol. Pharmacol. 2014, 85, 692–702. [Google Scholar] [CrossRef] [Green Version]
  30. Huang, C.W.; Hung, T.Y.; Wu, S.N. The inhibitory actions by lacosamide, a functionalized amino acid, on voltage-gated Na+ currents. Neuroscience 2015, 287, 125–136. [Google Scholar] [CrossRef]
  31. Rogawski, M.A.; Tofighy, A.; White, H.S.; Matagne, A.; Wolff, C. Current understanding of the mechanism of action of the antiepileptic drug lacosamide. Epilepsy Res. 2015, 110, 189–205. [Google Scholar] [CrossRef] [PubMed]
  32. Carona, A.; Bicker, J.; Silva, R.; Fonseca, C.; Falcão, A.; Fortuna, A. Pharmacology of lacosamide: From its molecular mechanisms and pharmacokinetics to future therapeutic applications. Life Sci. 2021, 275, 119342. [Google Scholar] [CrossRef] [PubMed]
  33. Serôdio, P.; Vega-Saenz de Miera, E.; Rudy, B. Cloning of a novel component of A-type K+ channels operating at subthreshold potentials with unique expression in heart and brain. J. Neurophysiol. 1996, 75, 2174–2179. [Google Scholar] [CrossRef] [PubMed]
  34. Carrasquillo, Y.; Burkhalter, A.; Nerbonne, J.M. A-type K+ channels encoded by Kv4.2, Kv4.3 and Kv1.4 differentially regulate intrinsic excitability of cortical pyramidal neurons. J. Physiol. 2012, 590, 3877–3890. [Google Scholar] [CrossRef] [Green Version]
  35. Simasko, S.M. Evidence for a delayed rectifier-like potassium current in the clonal rat pituitary cell line GH3. Am. J. Physiol. 1991, 261, E66–E75. [Google Scholar] [CrossRef]
  36. Wu, S.N.; Chen, I.J.; Lo, Y.C.; Yu, H.S. The characteristics in the inhibitory effects of capsaicin on voltage-dependent K(+) currents in rat atrial myocytes. Environ. Toxicol. Pharmacol. 1996, 2, 39–47. [Google Scholar] [CrossRef]
  37. Kehl, S.J. A Model of the Block of Voltage-Gated Potassium Kv4.2 Ionic Currents by 4-Aminopyridine. J. Pharmacol. Exp. Ther. 2017, 363, 184–195. [Google Scholar] [CrossRef]
  38. Castle, N.A. Differential inhibition of potassium currents in rat ventricular myocytes by capsaicin. Cardiovasc. Res. 1992, 26, 1137–1144. [Google Scholar] [CrossRef]
  39. Chen, C.; Heyward, P.; Zhang, J.; Wu, D.; Clarke, I.J. Voltage-dependent potassium currents in ovine somatotrophs and their function in growth hormone secretion. Neuroendocrinology 1994, 59, 1–9. [Google Scholar] [CrossRef]
  40. Luther, J.A.; Halmos, K.C.; Tasker, J.G. A slow transient potassium current expressed in a subset of neurosecretory neurons of the hypothalamic paraventricular nucleus. J. Neurophysiol. 2000, 84, 1814–1825. [Google Scholar] [CrossRef]
  41. Van Goor, F.; Zivadinovic, D.; Stojilkovic, S.S. Differential expression of ionic channels in rat anterior pituitary cells. Mol. Endocrinol. 2001, 15, 1222–1236. [Google Scholar] [CrossRef]
  42. Huang, C.W.; Huang, C.C.; Liu, Y.C.; Wu, S.N. Inhibitory effect of lamotrigine on A-type potassium current in hippocampal neuron-derived H19-7 cells. Epilepsia 2004, 45, 729–736. [Google Scholar] [CrossRef]
  43. Andrásfalvy, B.K.; Makara, J.K.; Johnston, D.; Magee, J.C. Altered synaptic and non-synaptic properties of CA1 pyramidal neurons in Kv4.2 knockout mice. J. Physiol. 2008, 586, 3881–3892. [Google Scholar] [CrossRef]
  44. Khaliq, Z.M.; Bean, B.P. Dynamic, nonlinear feedback regulation of slow pacemaking by A-type potassium current in ventral tegmental area neurons. J. Neurosci. 2008, 28, 10905–10917. [Google Scholar] [CrossRef] [Green Version]
  45. Fransén, E.; Tigerholm, J. Role of A-type potassium currents in excitability, network synchronicity, and epilepsy. Hippocampus 2010, 20, 877–887. [Google Scholar] [CrossRef] [Green Version]
  46. Stojilkovic, S.S.; Tabak, J.; Bertram, R. Ion channels and signaling in the pituitary gland. Endocr. Rev. 2010, 31, 845–915. [Google Scholar] [CrossRef] [Green Version]
  47. Ping, Y.; Waro, G.; Licursi, A.; Smith, S.; Vo-Ba, D.A.; Tsunoda, S. Shal/K(v)4 channels are required for maintaining excitability during repetitive firing and normal locomotion in Drosophila. PLoS ONE 2011, 6, e16043. [Google Scholar] [CrossRef] [Green Version]
  48. Mendonça, P.R.F.; Kyle, V.; Yeo, S.H.; Colledge, W.H.; Robinson, H.P.C. Kv4.2 channel activity controls intrinsic firing dynamics of arcuate kisspeptin neurons. J. Physiol. 2018, 596, 885–899. [Google Scholar] [CrossRef]
  49. Zemel, B.M.; Ritter, D.M.; Covarrubias, M.; Muqeem, T. A-Type K(V) Channels in Dorsal Root Ganglion Neurons: Diversity, Function, and Dysfunction. Front. Mol. Neurosci. 2018, 11, 253. [Google Scholar] [CrossRef] [Green Version]
  50. Ni, H.; Zhang, H.; Grandi, E.; Narayan, S.M.; Giles, W.R. Transient outward K(+) current can strongly modulate action potential duration and initiate alternans in the human atrium. Am. J. Physiol. Heart Circ. Physiol. 2019, 316, H527–H542. [Google Scholar] [CrossRef]
  51. Kim, K.R.; Lee, S.Y.; Yoon, S.H.; Kim, Y.; Jeong, H.J.; Lee, S.; Suh, Y.H.; Kang, J.S.; Cho, H.; Lee, S.H.; et al. Kv4.1, a Key Ion Channel For Low Frequency Firing of Dentate Granule Cells, Is Crucial for Pattern Separation. J. Neurosci. 2020, 40, 2200–2214. [Google Scholar] [CrossRef] [PubMed]
  52. Fletcher, P.A.; Sherman, A.; Stojilkovic, S.S. Common and diverse elements of ion channels and receptors underlying electrical activity in endocrine pituitary cells. Mol. Cell Endocrinol. 2018, 463, 23–36. [Google Scholar] [CrossRef]
  53. Errington, A.C.; Coyne, L.; Stöhr, T.; Selve, N.; Lees, G. Seeking a mechanism of action for the novel anticonvulsant lacosamide. Neuropharmacology 2006, 50, 1016–1029. [Google Scholar] [CrossRef] [PubMed]
  54. Evans, N.J.; Bayliss, A.L.; Reale, V.; Evans, P.D. Characterisation of Signalling by the Endogenous GPER1 (GPR30) Receptor in an Embryonic Mouse Hippocampal Cell Line (mHippoE-18). PLoS ONE 2016, 11, e0152138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Sun, W.; Smith, D.; Fu, Y.; Cheng, J.X.; Bryn, S.; Borgens, R.; Shi, R. Novel potassium channel blocker, 4-AP-3-MeOH, inhibits fast potassium channels and restores axonal conduction in injured guinea pig spinal cord white matter. J. Neurophysiol. 2010, 103, 469–478. [Google Scholar] [CrossRef] [PubMed]
  56. Wu, S.N.; Yang, W.H.; Yeh, C.C.; Huang, H.C. The inhibition by di(2-ethylhexyl)-phthalate of erg-mediated K⁺ current in pituitary tumor (GH₃) cells. Arch Toxicol. 2012, 86, 713–723. [Google Scholar] [CrossRef] [PubMed]
  57. Sheets, P.L.; Heers, C.; Stoehr, T.; Cummins, T.R. Differential block of sensory neuronal voltage-gated sodium channels by lacosamide [(2R)-2-(acetylamino)-N-benzyl-3-methoxypropanamide], lidocaine, and carbamazepine. J. Pharmacol. Exp. Ther. 2008, 326, 89–99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. May, T.W.; Brandt, C.; Helmer, R.; Bien, C.G.; Cawello, W. Comparison of lacosamide concentrations in cerebrospinal fluid and serum in patients with epilepsy. Epilepsia 2015, 56, 1134–1140. [Google Scholar] [CrossRef]
  59. Männikkö, R.; Elinder, F.; Larsson, H.P. Voltage-sensing mechanism is conserved among ion channels gated by opposite voltages. Nature 2002, 419, 837–841. [Google Scholar] [CrossRef]
  60. Panagopoulos, D.J.; Karabarbounis, A.; Yakymenko, I.; Chrousos, G.P. Human-made electromagnetic fields: Ion forced-oscillation and voltage-gated ion channel dysfunction, oxidative stress and DNA damage (Review). Int. J. Oncol. 2021, 59, 92. [Google Scholar] [CrossRef]
  61. Villalba-Galea, C.A.; Chiem, A.T. Hysteretic Behavior in Voltage-Gated Channels. Front. Pharm. 2020, 11, 579596. [Google Scholar] [CrossRef]
  62. Hsiao, H.T.; Lu, G.L.; Liu, Y.C.; Wu, S.N. Effective Perturbations of the Amplitude, Gating, and Hysteresis of I(K(DR)) Caused by PT-2385, an HIF-2α Inhibitor. Membranes 2021, 11, 636. [Google Scholar] [CrossRef]
  63. Chang, W.T.; Liu, P.Y.; Gao, Z.H.; Lee, S.W.; Lee, W.K.; Wu, S.N. Evidence for the Effectiveness of Remdesivir (GS-5734), a Nucleoside-Analog Antiviral Drug in the Inhibition of I (K(M)) or I (K(DR)) and in the Stimulation of I (MEP). Front. Pharm. 2020, 11, 1091. [Google Scholar] [CrossRef]
  64. Hattori, S.; Murakami, F.; Song, W.J. Quantitative relationship between Kv4.2 mRNA and A-type K+ current in rat striatal cholinergic interneurons during development. J. Neurophysiol. 2003, 90, 175–183. [Google Scholar] [CrossRef] [Green Version]
  65. Zhu, X.R.; Wulf, A.; Schwarz, M.; Isbrandt, D.; Pongs, O. Characterization of human Kv4.2 mediating a rapidly-inactivating transient voltage-sensitive K+ current. Recept. Channels 1999, 6, 387–400. [Google Scholar]
  66. Delaunois, A.; Colomar, A.; Depelchin, B.O.; Cornet, M. Cardiac safety of lacosamide: The non-clinical perspective. Acta Neurol. Scand. 2015, 132, 337–345. [Google Scholar] [CrossRef]
  67. Cho, H.Y.; Chuang, T.H.; Wu, S.N. Effective Perturbations on the Amplitude and Hysteresis of Erg-Mediated Potassium Current Caused by 1-Octylnonyl 8-[(2-hydroxyethyl)[6-oxo-6(undecyloxy)hexyl]amino]-octanoate (SM-102), a Cationic Lipid. Biomedicines 2021, 9, 1367. [Google Scholar] [CrossRef]
Figure 1. Effects of LCS on the magnitude of IK(A) in GH3 pituitary tumor cells. These experiments were made in cells that were bathed in Ca2+−free, Tyrode’s solution containing 1 μM tetrodotoxin (TTX) and 0.5 mM CdCl2, and we backfilled the recording electrode by using a K+−containing (145 mM) solution. (A) Representative IK(A) traces obtained in the control (black color) and during cell exposure to 10 μM LCS (brown color) or 100 μM LCS (red color). The uppermost part denotes the voltage−clamp protocol used. (B) Current traces showing an expanded record from those in (A); their trajectories were fitted by a two exponential (smooth gray line). Data points (indicated in open circles) were taken with or without the addition of LCS (10 or 100 μM). (C) Kinetic estimate in LCS−mediated block of IK(A) in GH3 cells (mean ± SEM; n = 8 for each point). The reciprocal of the slow component in the inactivation time constant (1/τinact(S)) of IK(A) derived from the exponential fit of the IK(A) trajectory was collated and then linearly plotted against the LCS concentration (gray straight line). Forward (on, k+1*) or backward (off, k−1) rate constant for the binding scheme, derived from the slope and the ordinate axis of the interpolated line, was 0.0213 s−1μM−1 or 1.112 s−1, respectively; thereafter, the KD value (k−1/k+1* = 52.3 μM) was yielded. (D) Concentration-dependent relationship of LCS effect on peak (red open circles) or sustained (black filled squares) IK(A) activated by 1−sec membrane depolarization (mean ± SEM; n = 8 for each point). Current amplitude was measured at the beginning or end-pulse of each depolarizing step, from −80 to −30 mV, with a duration of 1 s. The sigmoidal curve (black or red line) represents the best fit to the Hill equation (detailed in Materials and Methods). The statistical analyses were conducted with ANOVA−2 for repeated measures, (p (factor 1, groups among data taken at different LCS concentrations) < 0.05, p (factor 2, groups between the sustained and peak IK(A)) < 0.05, p (interaction) < 0.05, followed by post-hoc Fisher’s least−significance difference test, p < 0.05).
Figure 1. Effects of LCS on the magnitude of IK(A) in GH3 pituitary tumor cells. These experiments were made in cells that were bathed in Ca2+−free, Tyrode’s solution containing 1 μM tetrodotoxin (TTX) and 0.5 mM CdCl2, and we backfilled the recording electrode by using a K+−containing (145 mM) solution. (A) Representative IK(A) traces obtained in the control (black color) and during cell exposure to 10 μM LCS (brown color) or 100 μM LCS (red color). The uppermost part denotes the voltage−clamp protocol used. (B) Current traces showing an expanded record from those in (A); their trajectories were fitted by a two exponential (smooth gray line). Data points (indicated in open circles) were taken with or without the addition of LCS (10 or 100 μM). (C) Kinetic estimate in LCS−mediated block of IK(A) in GH3 cells (mean ± SEM; n = 8 for each point). The reciprocal of the slow component in the inactivation time constant (1/τinact(S)) of IK(A) derived from the exponential fit of the IK(A) trajectory was collated and then linearly plotted against the LCS concentration (gray straight line). Forward (on, k+1*) or backward (off, k−1) rate constant for the binding scheme, derived from the slope and the ordinate axis of the interpolated line, was 0.0213 s−1μM−1 or 1.112 s−1, respectively; thereafter, the KD value (k−1/k+1* = 52.3 μM) was yielded. (D) Concentration-dependent relationship of LCS effect on peak (red open circles) or sustained (black filled squares) IK(A) activated by 1−sec membrane depolarization (mean ± SEM; n = 8 for each point). Current amplitude was measured at the beginning or end-pulse of each depolarizing step, from −80 to −30 mV, with a duration of 1 s. The sigmoidal curve (black or red line) represents the best fit to the Hill equation (detailed in Materials and Methods). The statistical analyses were conducted with ANOVA−2 for repeated measures, (p (factor 1, groups among data taken at different LCS concentrations) < 0.05, p (factor 2, groups between the sustained and peak IK(A)) < 0.05, p (interaction) < 0.05, followed by post-hoc Fisher’s least−significance difference test, p < 0.05).
Ijms 23 01171 g001
Figure 2. Effect of LCS, tetraethylammonium chloride (TEA), 4-aminopyridine-3-methanol (4-AP-3-MeOH), 4-aminopyridine (4-AP), or capsaicin (Cap) on the peak amplitude of IK(A) identified in GH3 cells. In these experiments, we bathed GH3 cells in Ca2+-free, Tyrode’s solution, and the recording electrode was filled with K+-enriched (145 mM) solution. Current amplitude was measured at the start of the depolarizing pulse, from −80 to −30 mV. Each bar represents the mean ± SEM (n = 8). Data analysis was performed by ANOVA-1 (p < 0.05). * Significantly different from control (p < 0.05).
Figure 2. Effect of LCS, tetraethylammonium chloride (TEA), 4-aminopyridine-3-methanol (4-AP-3-MeOH), 4-aminopyridine (4-AP), or capsaicin (Cap) on the peak amplitude of IK(A) identified in GH3 cells. In these experiments, we bathed GH3 cells in Ca2+-free, Tyrode’s solution, and the recording electrode was filled with K+-enriched (145 mM) solution. Current amplitude was measured at the start of the depolarizing pulse, from −80 to −30 mV. Each bar represents the mean ± SEM (n = 8). Data analysis was performed by ANOVA-1 (p < 0.05). * Significantly different from control (p < 0.05).
Ijms 23 01171 g002
Figure 3. Effect of LCS on the current versus voltage (I−V) relationship of IK(A) identified in GH3 cells. (A) Representative current traces obtained in the control period (i.e., LCS was not present, top panel) and during the exposure to 100 μM LCS (bottom panel). The uppermost part shows the voltage−clamp protocol applied. In (B,C), mean I−V relationships of peak or sustained IK(A) achieved in the absence (filled symbols) and presence (open symbols) of 100 μM LCS are respectively illustrated (mean ± SEM; n = 7 for each point). The amplitude of peak or sustained IK(A) was respectively measured at the beginning− or end−pulse of step depolarization from −80 to −30 mV with a duration of 1 s. The statistical analyses were made by ANOVA−2 for repeated measures, (p (factor 1, groups among data taken at different level of membrane potentials) < 0.05, p (factor 2, groups between the absence and presence of LCS) < 0.05, p (interaction) < 0.05, followed by post−hoc Fisher’s least−significance difference test, p < 0.05). Of notice, no change in the overall I−V relationship of peak or sustained IK(A) was detected in the presence of LCS, despite its reduction in the amplitude of peak or sustained IK(A).
Figure 3. Effect of LCS on the current versus voltage (I−V) relationship of IK(A) identified in GH3 cells. (A) Representative current traces obtained in the control period (i.e., LCS was not present, top panel) and during the exposure to 100 μM LCS (bottom panel). The uppermost part shows the voltage−clamp protocol applied. In (B,C), mean I−V relationships of peak or sustained IK(A) achieved in the absence (filled symbols) and presence (open symbols) of 100 μM LCS are respectively illustrated (mean ± SEM; n = 7 for each point). The amplitude of peak or sustained IK(A) was respectively measured at the beginning− or end−pulse of step depolarization from −80 to −30 mV with a duration of 1 s. The statistical analyses were made by ANOVA−2 for repeated measures, (p (factor 1, groups among data taken at different level of membrane potentials) < 0.05, p (factor 2, groups between the absence and presence of LCS) < 0.05, p (interaction) < 0.05, followed by post−hoc Fisher’s least−significance difference test, p < 0.05). Of notice, no change in the overall I−V relationship of peak or sustained IK(A) was detected in the presence of LCS, despite its reduction in the amplitude of peak or sustained IK(A).
Ijms 23 01171 g003
Figure 4. Effect of LCS on the quasi−steady−state inactivation curve of IK(A) in GH3 cells. In this set of experiments, the conditioning voltage pulses with a duration of 1 s to a series of command voltage steps, ranging from −100 to −10 mV in 10−mV increments, were delivered to the examined cell from a holding potential of −80 mV. Following each conditioning pulse, a test pulse to −30 mV with a duration of 1 s was applied to evoke IK(A). (A) Representative current traces obtained in the absence (a) and presence (b) of 100 μM LCS. The voltage−clamp protocol is illustrated in the uppermost part. (B) Steady-state inactivation curve of IK(A) achieved in the absence (■) and presence (○) of 100 μM LCS (mean ± SEM; n = 7 for each point). The smooth curves were well fitted by the modified Boltzmann equation, as defined in Materials and Methods. The statistical analyses were made by ANOVA−2 for repeated measures, (p (factor 1, groups among data taken at the conditioning voltage levels) < 0.05, p (factor 2, groups between the absence and presence of LCS) < 0.05, p (interaction) < 0.05, followed by post−hoc Fisher’s least−significance difference test, p < 0.05). Notice that the exposure to LCS shifts the midpoint of the inactivation curve along the voltage axis toward less hyperpolarized voltage (i.e., to the right); however, it is devoid of modifications in the gating charge of the curve in its presence.
Figure 4. Effect of LCS on the quasi−steady−state inactivation curve of IK(A) in GH3 cells. In this set of experiments, the conditioning voltage pulses with a duration of 1 s to a series of command voltage steps, ranging from −100 to −10 mV in 10−mV increments, were delivered to the examined cell from a holding potential of −80 mV. Following each conditioning pulse, a test pulse to −30 mV with a duration of 1 s was applied to evoke IK(A). (A) Representative current traces obtained in the absence (a) and presence (b) of 100 μM LCS. The voltage−clamp protocol is illustrated in the uppermost part. (B) Steady-state inactivation curve of IK(A) achieved in the absence (■) and presence (○) of 100 μM LCS (mean ± SEM; n = 7 for each point). The smooth curves were well fitted by the modified Boltzmann equation, as defined in Materials and Methods. The statistical analyses were made by ANOVA−2 for repeated measures, (p (factor 1, groups among data taken at the conditioning voltage levels) < 0.05, p (factor 2, groups between the absence and presence of LCS) < 0.05, p (interaction) < 0.05, followed by post−hoc Fisher’s least−significance difference test, p < 0.05). Notice that the exposure to LCS shifts the midpoint of the inactivation curve along the voltage axis toward less hyperpolarized voltage (i.e., to the right); however, it is devoid of modifications in the gating charge of the curve in its presence.
Ijms 23 01171 g004
Figure 5. LCS−induced prolongation in the recovery of the IK(A) block in GH3 cells. Cells were bathed in Ca2+−free, Tyrode’s solution, and we filled up the electrode with K+−containing solution. During the recordings, we applied another set of two-pulse voltage−clamp protocol to the examined cells. (A) Representative current traces, demonstrating current recovery from the block by use of two−step voltage protocol with varying inter−pulse intervals (as indicated in the uppermost part). a: control; b: in the presence of 100 μM LCS. The uppermost part shows the voltage−clamp protocol applied. (B) Time course of recovery from IK(A) inactivation taken in the control period (■) and during exposure to 100 μM LCS (○) (mean ± SEM; n = 8 for each point). The smooth line, with or without the addition of LCS, was well fitted by single exponential function. The statistical analyses were made by ANOVA−2 for repeated measures, (p (factor 1, groups among data taken at different inter-pulse interval) < 0.05, p (factor 2, groups between the absence and presence of LCS) < 0.05, p (interaction) < 0.05, followed by post−hoc Fisher’s least−significance difference test, p < 0.05).
Figure 5. LCS−induced prolongation in the recovery of the IK(A) block in GH3 cells. Cells were bathed in Ca2+−free, Tyrode’s solution, and we filled up the electrode with K+−containing solution. During the recordings, we applied another set of two-pulse voltage−clamp protocol to the examined cells. (A) Representative current traces, demonstrating current recovery from the block by use of two−step voltage protocol with varying inter−pulse intervals (as indicated in the uppermost part). a: control; b: in the presence of 100 μM LCS. The uppermost part shows the voltage−clamp protocol applied. (B) Time course of recovery from IK(A) inactivation taken in the control period (■) and during exposure to 100 μM LCS (○) (mean ± SEM; n = 8 for each point). The smooth line, with or without the addition of LCS, was well fitted by single exponential function. The statistical analyses were made by ANOVA−2 for repeated measures, (p (factor 1, groups among data taken at different inter-pulse interval) < 0.05, p (factor 2, groups between the absence and presence of LCS) < 0.05, p (interaction) < 0.05, followed by post−hoc Fisher’s least−significance difference test, p < 0.05).
Ijms 23 01171 g005
Figure 6. Inhibitory effect of LCS on the voltage-dependent hysteresis (Vhys) of IK(A) activated by inverted isosceles−triangular ramp pulse in GH3 cells. In this set of whole−cell current recordings, the potential applied to the examined cell was maintained at −80 mV, and we then imposed an inverted isosceles−triangular ramp pulse with the duration of 600 or 800 ms (i.e., ramp speed of ±150 or ±112.5 mV/s) to activate IK(A) in response to descending (from +10 to −80 mV) and ascending (from −80 to +10 mV) ramp voltage−clamp commands. (A) Representative current traces obtained in the control period (A1) and during cell exposure to 100 μM LCS (A2). The uppermost part shows the voltage−clamp protocol applied. The black and blue colors in (A1) or red and pink colors in (A2) indicate the current traces obtained with the ramp duration of 600 and 800 ms, respectively. (B) Representative instantaneous I−V relationships of IK(A) in response to inverted isosceles−triangular ramp pulse with a duration of 600 ms (black color in (B1) or red color in (B2)) or 800 ms (blue color in (B1) or pink color in (B2)). In (B1) and (B2), the instantaneous I−V relationships of IK(A) are illustrated in the absence and presence of 100 μM LCS, respectively. The dashed arrow in (B1) and (B2) shows the direction of IK(A) trajectories in which time passes during current elicitation by the inverted isosceles−triangular ramp pulse with a duration of 600 or 800 ms. (C) Summary bar graph demonstrating effects of LCS (30 or 100 μM) on the hysteretic area (Δarea) of IK(A) activated by isosceles−triangular ramp pulse (mean ± SEM; n = 8 for each bar). The hysteretic area indicates the one under the curve activated during the descending and ascending ends of the triangular ramp pulse. Red or green bars respectively indicate the hysteretic areas taken in the duration of 600 or 800 ms. Data analysis was made by ANOVA−1 (p < 0.05). Of note, there was an evident occurrence of voltage−dependent hysteresis (Vhys) for IK(A) activated by triangular ramp pulse, and the presence of LCS was able to attenuate the hysteretic area of the current. * Significantly different from controls (p < 0.05).
Figure 6. Inhibitory effect of LCS on the voltage-dependent hysteresis (Vhys) of IK(A) activated by inverted isosceles−triangular ramp pulse in GH3 cells. In this set of whole−cell current recordings, the potential applied to the examined cell was maintained at −80 mV, and we then imposed an inverted isosceles−triangular ramp pulse with the duration of 600 or 800 ms (i.e., ramp speed of ±150 or ±112.5 mV/s) to activate IK(A) in response to descending (from +10 to −80 mV) and ascending (from −80 to +10 mV) ramp voltage−clamp commands. (A) Representative current traces obtained in the control period (A1) and during cell exposure to 100 μM LCS (A2). The uppermost part shows the voltage−clamp protocol applied. The black and blue colors in (A1) or red and pink colors in (A2) indicate the current traces obtained with the ramp duration of 600 and 800 ms, respectively. (B) Representative instantaneous I−V relationships of IK(A) in response to inverted isosceles−triangular ramp pulse with a duration of 600 ms (black color in (B1) or red color in (B2)) or 800 ms (blue color in (B1) or pink color in (B2)). In (B1) and (B2), the instantaneous I−V relationships of IK(A) are illustrated in the absence and presence of 100 μM LCS, respectively. The dashed arrow in (B1) and (B2) shows the direction of IK(A) trajectories in which time passes during current elicitation by the inverted isosceles−triangular ramp pulse with a duration of 600 or 800 ms. (C) Summary bar graph demonstrating effects of LCS (30 or 100 μM) on the hysteretic area (Δarea) of IK(A) activated by isosceles−triangular ramp pulse (mean ± SEM; n = 8 for each bar). The hysteretic area indicates the one under the curve activated during the descending and ascending ends of the triangular ramp pulse. Red or green bars respectively indicate the hysteretic areas taken in the duration of 600 or 800 ms. Data analysis was made by ANOVA−1 (p < 0.05). Of note, there was an evident occurrence of voltage−dependent hysteresis (Vhys) for IK(A) activated by triangular ramp pulse, and the presence of LCS was able to attenuate the hysteretic area of the current. * Significantly different from controls (p < 0.05).
Ijms 23 01171 g006
Figure 7. Effect of LCS on IK(A) identified in mouse mHippoE−14 hippocampal neurons. Cells were bathed in Ca2+−free, Tyrode’s solution, and the recording electrode was filled with K+−enriched (145 mM) solution. (A) Representative current traces obtained in the absence (black color) and presence of 30 μM LCS (green color) or 100 μM LCS (red color). The inset denotes the voltage−clamp protocol given. The bottom panel shows an expanded record from the dashed box in the top panel. The data points (open circles) in each current trajectory were reduced by 50 for better illustration. The current trajectory was fitted with the goodness of fits by a two−exponential function (indicated by gray smooth line). In (B) or (C), the summary bar graph depicts the LCS effect on the sustained IK(A) amplitude or τinact(S) value of IK(A) inactivation, respectively (mean ± SEM; n = 8). Current amplitude was activated by membrane depolarization from −80 to −30 mV with a duration of 1 s, and each current trajectory was fitted by a two−exponential function. Statistical analyses in (B,C) were made by ANOVA−1 (p < 0.05). * Significantly different from controls (p < 0.05).
Figure 7. Effect of LCS on IK(A) identified in mouse mHippoE−14 hippocampal neurons. Cells were bathed in Ca2+−free, Tyrode’s solution, and the recording electrode was filled with K+−enriched (145 mM) solution. (A) Representative current traces obtained in the absence (black color) and presence of 30 μM LCS (green color) or 100 μM LCS (red color). The inset denotes the voltage−clamp protocol given. The bottom panel shows an expanded record from the dashed box in the top panel. The data points (open circles) in each current trajectory were reduced by 50 for better illustration. The current trajectory was fitted with the goodness of fits by a two−exponential function (indicated by gray smooth line). In (B) or (C), the summary bar graph depicts the LCS effect on the sustained IK(A) amplitude or τinact(S) value of IK(A) inactivation, respectively (mean ± SEM; n = 8). Current amplitude was activated by membrane depolarization from −80 to −30 mV with a duration of 1 s, and each current trajectory was fitted by a two−exponential function. Statistical analyses in (B,C) were made by ANOVA−1 (p < 0.05). * Significantly different from controls (p < 0.05).
Ijms 23 01171 g007
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cho, H.-Y.; Chuang, T.-H.; Wu, S.-N. Evidence for Inhibitory Perturbations on the Amplitude, Gating, and Hysteresis of A-Type Potassium Current, Produced by Lacosamide, a Functionalized Amino Acid with Anticonvulsant Properties. Int. J. Mol. Sci. 2022, 23, 1171. https://doi.org/10.3390/ijms23031171

AMA Style

Cho H-Y, Chuang T-H, Wu S-N. Evidence for Inhibitory Perturbations on the Amplitude, Gating, and Hysteresis of A-Type Potassium Current, Produced by Lacosamide, a Functionalized Amino Acid with Anticonvulsant Properties. International Journal of Molecular Sciences. 2022; 23(3):1171. https://doi.org/10.3390/ijms23031171

Chicago/Turabian Style

Cho, Hsin-Yen, Tzu-Hsien Chuang, and Sheng-Nan Wu. 2022. "Evidence for Inhibitory Perturbations on the Amplitude, Gating, and Hysteresis of A-Type Potassium Current, Produced by Lacosamide, a Functionalized Amino Acid with Anticonvulsant Properties" International Journal of Molecular Sciences 23, no. 3: 1171. https://doi.org/10.3390/ijms23031171

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