• Nem Talált Eredményt

L-364,373 (R-L3) enantiomers have adverse modulating effects on IKs in mammalian ventricular myocytes

N/A
N/A
Protected

Academic year: 2022

Ossza meg "L-364,373 (R-L3) enantiomers have adverse modulating effects on IKs in mammalian ventricular myocytes"

Copied!
23
0
0

Teljes szövegt

(1)

L-364,373 (R-L3) enantiomers have adverse modulating effects on IKs in mammalian ventricular myocytes

Claudia Coricia, Zsófia Kohajdac, Attila Kristófc, András Horvátha,c, László Virága,c, Tamás Széla, Norbert Nagyc, Zsolt Szakonyib, Ferenc Fülöpb, Danina M. Munteand, András Varróa,c, Norbert Josta,c.

aDepartment of Pharmacology & Pharmacotherapy, Faculty of Medicine, University of Szeged

bDepartment of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Szeged,

cDivision of Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged, Hungary

dDepartment of Pathopysiology, "Victor Babes" University of Medicine and Pharmacy, Timisora, Romania

Correspondence:

Norbert Jost, PhD

Division of Cardiovascular Pharmacology, Hungarian Academy of Sciences and Department of Pharmacology & Pharmacotherapy, Faculty of Medicine

University of Szeged.

Dóm tér 12, P.O. Box 427, H-6701 Szeged, Hungary Tel.: (36-62) 546885, Fax: (36-62) 545680

E-mail: jost.norbert@med.u-szeged.hu

Running title: RL-3 enantiomers adverse modulate IKs

(2)

Abstract

Activators of slow delayed rectifier K+ current (IKs) have been suggested as promising tools to suppress ventricular arrhythmias due to prolongation of repolarization. Recently, L-364,373 (R-L3), was nominated to activate IKs in myocytes from several species, however, in some studies failed to activate IKs. One later study suggested the adverse modulating effects of the R-L3 enantiomers as possible explanation for this discrepancy. Therefore, we have analysed the effect of the RL-3 enantiomers on IKs in ventricular mammalian myocytes, by applying standard microelectrode and whole-cell patch clamp techniques at 37 C. We synthesised two substances, ZS_1270B (right) and ZS_1271B (left), the two enantiomers of R-L3. In rabbit myocytes ZS_1270B enhanced IKs tail current by about 30%, while ZS_1271B reduced IKs

tails by 45%. In guinea pig right ventricular preparations ZS_1270B shortened APD90 by 12%, while ZS_1271B, conversely lengthened it approximately 15%. We concluded that R- L3 enantiomers indeed have adverse modulating effects on IKs in the same concentration range, which may explain why the racemic drug R-L3 previously failed to activate IKs.

ZS_1270B is a potent IKs activator: therefore, this substance is adequate to test whether IKs

activators are indeed ideal tools to suppress ventricular arrhythmias originating from prolongation of action potentials.

Keywords: L-364,373, slow delayed rectifier potassium current, IKs activator, IKs blocker, enantiomers, antiarrhythmic drugs

(3)

Introduction

Action potential repolarization is an important phenomenon in the heart where it controls action potential duration (APD) and thus affects refractoriness and conduction of electrical impulses throughout the heart. Several pharmacological agents intended to abate cardiac arrhythmias specifically target mechanisms that regulate APD. In the ventricles, APD, in large part, determines dynamic changes in QT duration on a beat-by-beat basis. The rapid component of the delayed rectifier potassium current (IKr) has been identified in several mammalian species, including humans, and pharmacological agents that selectively block IKr

(eg, E-4031, sotalol, and dofetilide) markedly increase APD, QT duration, and ventricular refractoriness (for details see comprehensive review by Nattel, 2008). As such, high doses of these IKr blockers are associated with induction of torsade de pointes arrhythmias (Hondeghem and Snyders, 1990; Hohnloser and Woosley, 1994).

The role of the slow delayed rectifier potassium current (IKs) in human ventricular muscle action potential repolarization, on the other hand, has been debated. As with IKr, IKs

has been identified in several mammalian species, including humans (Salata et al, 1996;

Varró et al, 2000; Virág et al, 2001) and mutations in KCNQ1 and KCNE1, the α and β- subunits of the IKs potassium channel, are associated with specific form of the inherited long- QT syndrome, LQT1 (Roden et al, 1996). However, we have previously demonstrated that complete pharmacological block of IKs by either chromanol 293B or L-735,821 has little effect on APD in isolated dog and rabbit ventricular muscle (Jost et al, 2005) over a wide range of physiological pacing frequencies. These findings led us to speculate that IKs normally plays little role in ventricular muscle action potential repolarization, but when APD is abnormally long, IKs likely provides an important safety mechanism that, when removed, increases arrhythmic risk. Therefore, IKs is the key player of the repolarization reserve (Jost et al, 2005).

Same concept raised another hypothesis. The high proarrhythmic risk associated with these conditions could be markedly diminished by activation of the slow delayed rectifier K+ current (IKs), providing thus a promising strategy to increase the repolarization reserve without significantly lengthening action potential duration, i.e. to prevent development of early afterdepolarizations without shortening the refractory period. Testing the hypothesis was long time hampered by lacking of selective IKs agonists. The recently synthesized

(4)

benzodiazepine derivative compound L-364,373 (R-L3) has been reported to activate IKs at micromolar concentrations in ventricular myocytes of guinea pig (Salata et al. 1998) and rabbit (Xu et al. 2002), as well as in KCNQ1 channels expressed in Xenopus oocytes (Seebohm et al. 2003).

However, in another study we reported RL-3 to fail to activate IKs current in dog ventricular myocytes (Magyar et al, 2006). In this study we have shown that 1 µM R-L3 did not affect at all IKs current amplitude, while at higher concentration as 3 µM, the compound even blocked the current. One later study suggested a possible explanation for this discrepancy: the two enantiomers of the racemic R-L3 have different activities, namely that the d enantiomer activates, while the l enantiomer potently blocks IKs (Nissen et al, 2009).

This mixed activating and blocking effect might be a plausible answer why the racemic substance failed to activate IKs in dogs previously.

Therefore, the aim of the present study was to synthesise the two enantiomers of R-L3 and to analyse the effect of these compounds on IKs current in isolated ventricular rabbit myocytes, by applying the whole-cell patch clamp and standard microelectrode techniques at 37 C.

Methods

Experiments were carried out in ventricular myocytes isolated from rabbit and guinea pig hearts. All experiments were conducted in compliance with the Guide for the Care and Use of Laboratory Animals (USA NIH publication No 85-23, revised 1985). The protocols were approved by the review board of the Department of Animal Health and Food Control of the Ministry of Agriculture and Rural Development, Hungary (XII./01031/000/2008 and XIII./1211/2012).

Action potential measurements

Guinea pigs (300-500 grams ) of either sex were used. The animals were sacrificed by cervical dislocation after receiving 400 IU/kg heparin intravenously. The chest was opened and the heart was quickly removed and immediately rinsed in oxygenated Locke's solution containing (in mM): NaCl, 120; KCl, 4; CaCl2, 2; MgCl2, 1; NaHCO3, 22; and glucose, 11.

The pH of this solution was 7.35 to 7.45 when gassed with 95% O2 and 5% CO2 at 37 °C.

(5)

Papillary muscles were obtained from the right ventricle of the hearts. The preparations were placed in a tissue bath and allowed to equilibrate for at least 1 h while superfused with oxygenated Locke's solution (flow rate 4–5 ml min−1) warmed to 37 °C.

Initially, each preparation was stimulated at a basic cycle length of 1000 ms (frequency = 1 Hz), using 2 ms long rectangular constant voltage pulses isolated from ground and delivered via bipolar platinum electrodes in contact with the preparation using an EMG 4767 type stimulator (Medicor Ltd, Budapest, H-1147, Hungary). One hour or more was allowed for each preparation to equilibrate while continuously superfused with Tyrode's solution warmed to 37 °C. Transmembrane potentials were recorded using a conventional glass microelectrode filled with 3 M KCl with a tip resistance of 5-20 Mohms connected to an high impedance electrometer (Bio-Logic VF102, CLAIX, F-38640, France) referenced to ground. The first derivative of transmembrane potential (Vmax) was electronically obtained derived using a Bio-Logic DV-140 (Claix, F-38640, France) differentiator designed and calibrated to have a linear response over the range of 10 to 1000 V/s. Amplifier outputs were digitized using an ADA 3300 analog-to-digital converter (Real Time Devices Inc, State College, PA 16804, USA) with a maximum sampling frequency rate of 50 kHz connected to an IBM compatible personal computer. Data was stored and analyzed on a personal computer (PC) while also monitored on a dual beam memory oscilloscope (Tektronix 2230, Beaverton, OR 97077, USA). Resting membrane potential (RP), action potential amplitude (APA), and action potential durations (APD), measured at 50% and 90% repolarization (APD50 and APD90), were automatically measured with software developed in our laboratory (APES, Hugo Sachs Elektronik, March-Hugstetten, D-79229, Germany). Stimulation pulses were also controlled by PC software providing constant current pulses with programmed timing and amplitudes to the preparation via an EMG 47671 type signal isolator (Medicor Ltd, Budapest, H-1147, Hungary).

In each experiment, baseline action potential characteristics were first obtained during superfusion with normal 37 ºC Tyrode’s solution during while continuously pacing at a basic cycle length of 1000 ms. Recordings were continuously monitored to confirm one-to-one activation throughout the procedure.

After baseline measurements were obtained, each preparation was superfused with Tyrode’s solution containing a single test drug diluted to the proper concentration for 40 to 60 minutes before measurements were repeated at 3 min intervals in the continued presence of the test drug until less than a 5% change occurred in action potential characteristics between subsequent samples. When microelectrode impalement was lost, reimpalement was attempted.

(6)

If action potential characteristics recorded with the new impalement deviated by more than 5% from the preceding ones, the experiment was terminated, and results were excluded from evaluation.

Voltage clamp measurements.

Preparation of rabbit ventricular myocytes

Untreated New-Zealand white rabbits (body weights 1.5-2 kg) of either sex were used for the study. Single ventricular myocytes were obtained by enzymatic dissociation. The animals were sacrificed by cervical dislocation after receiving 400 IU/kg heparin intravenously. The chest was opened and the heart was quickly removed and placed into cold (4°C) solution with the following composition (mM): NaCl 135, KCl 4.7, KH2PO4 1.2, MgSO4 1.2, HEPES 10, NaHCO3 4.4, Glucose 10, CaCl2 1.8, (pH 7.2). The heart was mounted on a modified, 60 cm high Langendorff column and perfused with oxygenated and prewarmed (37°C) solution mentioned above. After washing out of blood (3-5 min) it was perfused with nominally Ca-free solution until the heart ceased (approx. 3-4 minutes). The digestion was performed by perfusion with the same solution supplemented with 0.33 mg/ml (90 U/ml) Collagenase (Type I, SIGMA Chemical, St. Louis, MO, USA,) and 0.02 mg/ml Pronase E (SIGMA) with 0.1% Albumin using a perfusion pump (flow rate approx. 15 ml/min). In the 15th minute of the enzyme perfusion the calcium concentration was elevated by 200 µM. After 30-35 minute the heart was removed from the canula and was placed into enzyme free solution containing 1.8 mM CaCl2 and 1% Albumin and equilibrated at 37°C for 10 minutes. Then the tissue was cut into small fragments. After gentle agitation, the cells were separated from the chunks by filtering through nylon mesh. Sedimentation was used for harvesting cells; as soon as most myocytes reached the bottom of the vessel the supernatant was removed and replaced by HEPES buffered Tyrode’s solution. This solution contained (mM): NaCl 144, NaH2PO4 0.33, KCl 4.0, CaCl2 1.8, MgCl2 0.53, Glucose 5.5, and HEPES 5.0 at pH of 7.4 containing 1.8 mM CaCl2. This procedure was repeated two times. The cells were stored at room temperature in the Tyrode solution.

Experimental techniques

One drop of cell suspension was placed within a transparent recording chamber

(7)

mounted on the stage of an inverted microscope (Olympus IX51, Tokyo, Japan), and individual myocytes were allowed to settle and adhere to the chamber bottom for at least 5 minutes before superfusion was initiated. Only rod shape cells with clear striations were used.

Cell capacitance (114.23±8.12 pF, n=20 for rabbit cell) was measured by applying a 10 mV hyperpolarising pulse from –10 mV. The holding potential was -90 mV. The capacity was measured by integration of the capacitive transient divided by the amplitude of the voltage step (10 mV). HEPES buffered Tyrode's solution served as the normal superfusate. Patch- clamp micropipettes were fabricated from glass capillaries (Clark, UK) using a using a computer controlled horizontal puller (Sutter, Sutter Co, Novato, CA, USA). These electrodes had resistances between 1.5 and 2.5 Mohms when filled with pipette solution containing (mM): KCl 140, MgCl2 4, K2ATP 5, HEPES 10, EGTA 1; pH adjusted to 7.2 by KOH. 1 µM nisoldipine (gift from the Bayer AG, Leverkusen, Germany) was placed in the external solution to eliminate inward Ca2+ current (ICa). When measuring IKs, sodium current (INa), was inactivated by applying a short 20 ms prepulse to -40 mV, which also largely inactivated transient outward current (Ito). The rapid IKr and slow IKs components of the delayed rectifier potassium current were separated by using 1 µM dofetilide (Sequoia Research Products, Ltd, UK), a selective IKr blocker.

Membrane currents were recorded with an Axopatch 200B amplifier (Molecular Devices-Axon Instruments, Union City, CA, USA) using the whole-cell configuration of the patch-clamp technique. After establishing a high (1-10 Gohm) resistance seal by gentle suction, the cell membrane beneath the tip of the electrode was disrupted by suction or by application of 1.5 V electrical pulses for 1-5 ms. The series resistance was typically 4-8 Mohm before compensation (50 - 80%, depending on the voltage protocols). Experiments where the series resistance was high, or substantially increased during measurement, were terminated and the results were excluded from analyses. Membrane currents were digitized using a 333 kHz analog-to-digital converter (Digidata 1320 and 1440, Molecular Devices- Axon Instruments) under software control (pClamp 8.0 and pClmap10, Axon Instruments).

Analyses were performed using Axon (pClamp 8.0 and pClamp 10) softwares after low-pass filtering at 1 kHz. All patch-clamp data were collected at 37 °C.

Statistical analysis

Results were compared using Student's t-tests for paired and unpaired data. When p<0.05, results were considered significant. Numerical data are expressed as mean ± SEM.

(8)

Results

Chemistry. Synthesis of L-364,373 (RL-3) and its optical enantiomers.

1H and 13C NMR spectra of L-364,373 (4, ZS_1270B) and it’s (S) enantiomer 7 (ZS_1271B) were recorded on a Bruker Avance DRX 400 spectrometer (400 MHz, δ=0 (TMS)), in CDCl3. Chemical shifts are expressed in ppm (δ) relative to TMS as internal reference. Melting points were determined on a Kofler apparatus and are uncorrected. MS measurements were made on a Finnigan MAT 95S double focusing mass spectrometer.

Typical source conditions were: temperature 443 K, electron energy 70eV, ionization current 700 µA and acceleration voltage 4 kV. The enantiomeric purity of 4 and 7 was determined by means of chiral HPLC and was found ee > 99% (Jasco instrument, Chiracel OD-H column, 4.6x250 mm, n-hexane/IPA = 90/10 as eluent, 0.5 mL/min flow rate, detection at 250 nm, tR1

= 63.6 min for 7 and tR2 = 75.4 min for 4). Optical rotations were obtained with a Perkin- Elmer 341 polarimeter.

(R)- and (S)-5-(2-Fluorophenyl)-1,3-dihydro-3-(1H-indol-3-ylmethyl)-2H-1,4- benzodiazepin-2-one (3 and 6) were prepared by literature method starting from 2-amino-2'- fluorobenzophenone and D- and L-tryptophan acid chloride hydrochloride (Figure 1), and all the physical and analytical data were similar to those in literature (Evans et al. 1987).

(R)- and (S)-5-(2-Fluorophenyl)-1,3-dihydro-3-(1H-indol-3-ylmethyl)-l-methyl-2H- 1,4-benzodiazepin-2-one) 4 (ZS_1270B) and 7 (ZS_1271B) were prepared by modified literature method of Evans et al. (Figure 1, Evans et al. 1987) to avoid the formation of the lactim ether side-product. To a stirred solution of compound 3 or 6 (324 mg, 0.85 mM) in dry acetone (20 mL), cesium carbonate (1.68 g, 5.1 mM) and methyl iodide (106 µL, 1.70 mM) was added. The reaction mixture was stirred for 10 h at room temperature (monitored by means of TLC). The inorganic material was filtered off and the filtrate was evaporated to dryness. The residue was chromatographed on silica gel (250-400 mesh) eluted with 25 % (v/v) ethyl acetate in n-hexane. Evaporation of the product fraction gave 4 (ZS_1270B) and 7 (ZS_1271B) as a white solid (yield 89 %), which was recrystallized from CH2Cl2/n-hexane.

The 1H NMR measuring showed the presence of 0.8 equivalent of CH2Cl2 in the resulted

(9)

compound (in the literature, product 4 was reported containing 0.6 equivalent of CH2Cl2).

After drying the crystals in vacuo at 80 °C for 8 h, the solvent was successfully eliminated.

Compound 4 (ZS_1270B): mp: 114-116 °C (Lit.: 105-113 °C), [α]D20

= +31.0 (c = 0.26, MeOH); 1H NMR (CDC13) δ 3.47 (3 H, s, NCH3), 3.66 (1 H, dd, J1 = 17 Hz, J2 = 10 Hz, CH2), 3.73-3.87 (2 H, m, CH2b + C3H), 6.93-7.61 (m, 13 H, Ar), 8.03 (br s, 1 H, indole NH).

13C NMR (CDC13) δ 28.2, 36.0, 65.1, 111.4, 113.9, 114.1, 116.6, 119.7, 122.2, 122.4, 124.6, 128.0, 128.3, 128.5, 129.3, 129.5, 130.7, 131.7, 131.9, 132.4, 136.6, 143.2, 160.2, 165.9, 170.8. MS (70eV), m/z 397 (M+, 21), 396 (1), 339 (7), 279 (6), 268 (4), 239 (8), 211 (18), 167 (12), 149 (30), 130 (100). C25H20FN3O: calcd. 397.15904; found 397.15928 (HRMS).

Compound 7 (ZS_1271B): [α]D20

= -31.0 (c = 0.26, MeOH); all the spectroscopic data and the mp were similar to those for the 3R enantiomer 4. C25H20FN3O: calcd. 397.15904; found 397.15939 (HRMS).

The investigation of the effect of R-L3 enantiomers on IKs current

We have decided to use rabbit ventricular myocytes for the study of effect of RL-3 enantiomers on IKs because of relatively large and easy measure IKs currents have been described particularly in this species (Lengyel et al, 2001).

The left panels of Figure 2 show typical IKs current traces recorded in rabbit ventricular myocyte. IKs was examined in isolated rabbit ventricular myocytes using test pulses of 5000 ms (IKs) in duration to between -10 mV and +50 mV from the holding potential of -80 mV. The pulse frequency was 0.05 Hz (IKr) or 0.1 Hz (IKs). The decaying tail current at -40 mV after the test pulse was assessed as IKs (Figure 2A). The amplitudes of the IKs tail currents have been determined as the difference between the peak tail current and the holding current level at -40 mV. When measured IKs, dofetilide (1 µM) was added to the nutrient solution to completely block IKr. The bottom panel of the same Figure 2A show that 1 µM ZS_1270B largely increased both outward depolarizing and deactivating tail currents. The corresponding right panel of Figure 2 shows the current-voltage relationships of IKs, before and after the superfusion with ZS_1270B measured on the average of n=6 cells. Figure 2B shows that at 40 mV, IKs tail current amplitude increased by about 26% in a statistically significant manner from 45.9±4.97 pA to 66.1±4.54 pA, after drug superfusion (n=6,

*p<0.05).

The left panels of Figure 3 show the effect of the left enantiomer ZS_1271B on the IKs

(10)

current recorded in rabbit ventricular myocytes (Figure 3A). The original recordings and the corresponding current-voltage relationships of IKs (Figure 3B) before and after the superfusion with ZS_1271B (1 µM) measured on the average of n=7 cells clearly show that the left enantiomer is a potent inhibitor of the IKs current. Figure 3B shows that at 40 mV, IKs tail current amplitude decreased in a statistically significant manner to about 47 % from 97.1±9.7 pA to 45.86±11.6 pA, after drug superfusion (n=7, *p<0.05).

Effect of selective IKs blockade and activation on the APD

These previous results demonstrated that the two R-L3 enantiomers indeed adversely modulates the IKs current, and are suitable to test the effect of both selective blockade and activation of the IKs current on the ventricular action potential repolarization.

For these experiments we investigate the effect of the two R-L3 enantiomers in guinea pig ventricular papillary muscle preparations. Guinea pig is the species known to have significant larger IKs current than in myocytes originated from other species including human (Jost et al, 2004). It is known that IKs is relatively small current in most mammalians, and it was shown that blockade of IKs current did not lengthen action potential duration in normal condition (without sympathetic stimulation) in most species including rabbits (Varró et al, 2000; Lengyel at el, 2001; Jost et al, 2005).

In a previous study we have reported that the only largely investigated species where IKs is large enough so that even without sympathetic stimulation selective IKs blockade lengthened APD in noticeable manner is guinea pig (Jost et al, 2004). Therefore, we opted for these experiments for guinea pig ventricular myocytes since in this species is expected to noticeably measure both lengthening and shortening of the ventricular APD.

As Figure 4 shows in guinea pig right ventricular preparations the activator right enantiomer ZS_1270B (1 µM) shortened APD90 to about 12.2±0.9 % (n=4), *p<0.05), while the IKs blocker left enantiomer ZS_1271B (1 µM) lengthened it to about 14.2±1.3 % (n=4,

*p<0.05) in the average of 4-4 cells. Table 1 summarizes all action potential data recorded in guinea pig ventricular preparations.

Discussion

(11)

The major finding of this study is that the racemic benzodiazepine derivative compound L-364,373 indeed as suggested by a previous study (Nissen et al, 2009) has two optical enantiomers that might have adverse modulating effect on the slow delayed rectifier potassium current (IKs). The right enantiomer proved to be a successful activator of IKs

current, while the left enantiomer is a potent blocker of IKs current. Moreover it seems that the two enantiomers have these adverse modulating effects at close concentrations. We have synthesised two substances, ZS_1270B (right) and ZS 1271B (left) the two enantiomers of R- L3 (Figure 1). We have shown that the right enantiomer ZS_1270B at 1 µM increased to about 26 % (Figure 2), while the left enantiomer ZS_1271B applied on the same concentration of 1 µM blocked to about 47% (Figure 3) the IKs current. In some previous studies other showed that L-364,373 (R-L3) in guinea pig and rabbit myocytes, in concentration dependent manner (0.1–1 µM) markedly increased IKs associated with a due to causing a leftward shift in its activation curve (Salata et al, 1998; Xu et al, 2002).

In 2006 in another study in dog ventricular myocytes we investigated the effect of the racemic compound R-L3, and we have shown that up to 3 µM concentration failed to activate the IKs current. Moreover at 3 µM the R-L3 compound even slightly decreased the outward (activating) and deactivating (tail) currents also (Magyar et al, 2006). At that time we could not explain the reason for this discrepancy. We just speculated whether this might be a species dependent different blocking property of the compound (Magyar et al, 2006).

In a latter study performed by a Danish group, it was reported that the pharmacologically induced Long QT type 2 can be rescued by activating the IKs current with the application of the benzodiazepine R-L3 (Nissen et al, 2009). In the paper based on an observation by Salata et al (1998) they hypothesized that the left enantiomer of the R-L3 (S- L3) might be a potent IKs blocker, thereby even at small impurities in the racemic R-L3 used could therefore cause significant reductions in the APD shortening effect of optically pure R- L3 (Nissen et al, 2009).

This observation was the working hypothesis to synthesize the two enantiomers. The results of the investigation clearly showed that indeed the right enantiomer is a successful activator, while the left enantiomer is a potent blocker of the IKs, therefore they are optimal compound to test the IKs modulator effect on the cardiac repolarization. We have applied the right enantiomer on the action potential repolarization in guinea pig papillary muscle and we showed that1 µM ZS_1270B shortened APD90, while conversely the left enantiomer ZS_1271B applied on the same concentration of 1 µM significantly lengthened the guinea pig repolarization (Figure 4).

(12)

IKs current is composed by co-assembling expression of the KvLQT1 and MinK proteins that can associate to form functional cardiac IKs channels (Barhanin et al, 1996, Sanguinetti et al, 1996). It has been known for many years that the cardiac current IKs is upregulated following sympathetic stimulation (Volders et al. 2003). This upregulation of the KCNQ1/KCNE1 current is mediated by β-adrenergic receptor activation, leading to an increased level of cAMP and thereby PKA stimulation, which interacts with the IKs complex through an A-kinase anchoring protein (AKAP) called yotiao (Chen et al, 2005). It would an important question to discuss at which pathways activates the ZS_1270B compound the IKs

current, ie. whether the IKs activating properties of the synthetized compounds is or not mediated via sympathetic stimulation. We have checked the chemical structures of the isoprenaline and other beta adrenergic agonist compounds, and may say the catecholamine but at least beta-phenylethylamine structure is an important criteria for beta-adrenergic stimulation on adrenerg receptors. If we looked the structure of the IKs activator enantiomer ZS_1270B, we found that none of the three critical pharmacophoric groups (Easson-Stedman hypothesis, 1933) could be found in the structure of ZS_1270B, therefore, in principle we can concluded that the activating effect of this compound is not mediated via beta-adrenergic stimulation pathways. Obviously it would be worthwhile in later studies to investigate the origin of the agonist effects of ZS_1270B compound.

KCNQ1 potassium channels are expressed in several other tissues than heart throughout the body and regulate key physiological functions. Potassium channels (KCNQ1 channels) are crucial for both keeping a proper membrane potential, which is necessary for transepithelial transport, and for participating in potassium absorption or secretion (Lohrmann et al, 1995). Epithelial KCNQ1 channels in combination with different β-subunits have been described in both apical and basolateral membranes, however the determinants for the subcellular localization of KCNQ1 in epithelial cells are largely unknown (Jespersen et al, 2004).

KCNQ1 channels are involved in the secretory process also. Increased KCNQ1 channel activity during secretion is primarily obtained by cAMP regulation of the channels, which consequently involves the cAMP activated intracellular Ca2+ mediated secretory processes (Lohrmann et al, 1995. One may speculate that it might be useful to investigate the effects of activation of epithelial KCNQ1 channels via an another pathway than the cAMP mediated one.

This observation that a racemic compound have two enantiomers with complete adverse modulating effect on a current, receptor, etc, (ie. one is an activator, while the other is

(13)

a blocker, moreover with a more or less similar activity), is quite unique and unusual, however not unknown at all. For example, the stereoisomers of the new dihydropyridine derivative 202-791 [isopropyl 4-(2,1,3-benzoxadiazol-4-yl)-1,4-dihydro-2,6-dimethyl-5-nitro- 3 -pyridinecarboxylate] were synthesized separately and tested on isolated rabbit aortic rings for effects on depolarization-induced contraction and depolarization-stimulated uptake of 45Ca2+. The result in that investigation showed the right enantiomer inhibited contraction, while by contrast, the left enantiomer of 202-791 compound shifted the concentration- response curve for depolarization-induced contraction in an almost parallel fashion to the left, thus enhancing contraction (Hof et al, 1985). Similar observation were done in some other studies also however, as we mentioned before, this is not a usual behaviour of the stereoisomers of the racemic compounds (Gonzales et al, 1983; Zittoun et al, 1991; Caillet et al, 2012).

Conclusions

These results indicate that the two enantiomers of R-L3 indeed have adverse modulating effects on IKs in the same concentration range, which may explain why the racemic drug R-L3 failed to activate IKs in previous studies. ZS_1270B is a potent activator of IKs, therefore, this substance is adequate to test whether IKs activators are indeed ideal tools to suppress ventricular arrhythmias originating from prolongation of action potentials.

Acknowledgements

This work was supported by grants from OTKA (CNK-77855, K-82079 and NK- 104331), National Office for Research and Technology (NKFP_07_01-RYT07_AF and REG- DA-09-2-2009-0115), National Development Agency and co-financed by the European Regional Fund (TÁMOP-4.2.1/B-09/1/KONV-2010-0005 and TÁMOP-4.2.2/B-10/1-2010- 0012), EU-FP7 (ICT-2008-224381), HU-RO Cross-Border Cooperation Programmes (HURO/0901/137-HU-RO_TRANSMED and HURO/0802/011_AF-HURO_CARDIOPOL) and the Hungarian Academy of Sciences. Dr. Zs. Szakonyi acknowledges the receipt of a János Bolyai Fellowship.

References:

(14)

Barhanin J, Lesage F, Guillemare E, Fink M, Lazdunski M, Romey G.. K(V)LQT1 and IsK (minK) proteins associate to form the I(Ks) cardiac potassium current. Nature. 1996;384:78- 80.

Caillet C, Chauvelot-Moachon L, Montastruc JL, Bagheri H; French Association of Regional Pharmacovigilance Centers (2012). Safety profile of enantiomers vs. racemic mixtures: it's the same? Br J Clin Pharmacol., 74, 886-889.

Chen L, Kurokawa J, Kass RS (2005). Phosphorylation of the A-kinase-anchoring protein Yotiao contributes to protein kinase A regulation of a heart potassium channel. J Biol Chem.

280(36), 31347-31352.

Easson LH, Stedman E (1933). CLXX. Studies on the relationship between chemical constitution and physiological action. V. Molecular dissymmetry and physiological activity.

Biochem. J. 27, 1257-1266.

Evans BE, Rittle KE, Bock MG, DiPardo RM, Freidinger RM, Whitter WL, Gould NP, Lundell GF, Homnick CF, Veber DF, Anderson PS, Chang RSL, Lotti VJ, Cerino DJ, Chen TJ, King PJ, Kunkel KA, Springer JP, Hirshfieldt J (1987). Design of nonpeptidal ligands for a peptide receptor: cholecystokinin antagonists. J Med Chem 30, 1229-1239.

González E, Ledesma de Paolo MI, Celener D, Celener FP, Rosembeck G, Panzitta MT, Bustos Fernández L (1993). The effect of different lactic acid isomers in the colon of rats.

Acta Gastroenterol Latinoam, 23, 203-210.

Hof RP, Rüegg UT, Hof A, Vogel A (1985). Stereoselectivity at the calcium channel:

opposite action of the enantiomers of a 1,4-dihydropyridine. J Cardiovasc Pharmacol., 7, 689- 693.

Hondeghem LM, Snyders DJ (1990). Class III antiarrhythmic agents have a lot of potential but a long way to go. Reduced effectiveness and dangers of reverse use dependence.

Circulation, 81, 686-690.

(15)

Hohnloser SH, Woosley RL (1994). Sotalol. New Engl J Med, 331, 31-38.

Jespersen T, Rasmussen HB, Grunnet M, Jensen HS, Angelo K, Dupuis DS, Vogel LK, Jorgensen NK, Klaerke DA, and Olesen SP (2004). Basolateral localisation of KCNQ1 potassium channels in MDCK cells: molecular identification of an N-terminal targeting motif.

J Cell Sci 117, 4517–4526.

Jost N, Virág L., Baláti B, Lengyel C, Németh M, Bitay M., Bogáts G., Varró A., Papp JGy.

A késıi egyenirányító káliumáram gyors (IKr) és lassú komponensének (IKs) összehasonlító vizsgálata egésszséges emberi, kutya, nyúl és tengerimalac kamrai szívizomsejteken.

Cardiol. Hung., 34, Suppl. E, E75-E84, 2004.

Jost N, L Virág, M Bitay, J Takács, Cs Lengyel, P Biliczki, ZA Nagy, G Bogáts, DA Lathrop, JGy Papp, A Varró (2005). Restricting excessive cardiac action potential and QT prolongation: a vital role for IKs in human ventricular muscle. Circulation, 112, 1392-1399.

Lengyel Cs, Iost N, Virág L, Varró A, Lathrop DA, Papp JGy (2001). Pharmacological block of the slow component of the outward delayed rectifier current (IKs) fails to lengthen rabbit ventricular muscle QTc and action potential duration. Br J Pharmacol, 132, 101-110.

Lohrmann E, Burhoff I, Nitschke RB, Lang HJ, Mania D, Englert HC, Hropot M, Warth R, Rohm W, Bleich M, and Greger R (1995). A new class of inhibitors of cAMP-mediated Cl secretion in rabbit colon, acting by the reduction of cAMP-activated K+ conductance. Pflügers Arch 429: 517–530.

Magyar J, Horváth B, Bányász T, Szentandrássy N, Birinyi P, Varró A, Szakonyi Z, Fülöp F, Nánási PP (2006). L-364,373 fails to activate the slow delayed rectifier K(+) current in canine ventricular cardiomyocytes. Naunyn Schmiedebergs Arch Pharmacol, 373, 85–90.

Nattel S (2008). Delayed-rectifier potassium currents and the control of cardiac repolarization: Noble and Tsien 40 years after. J Physiol., 586, 5849-5852.

Nissen JD, Diness JG, Diness TG, Hansen RS, Grunnet M, Jespersen T (2009).

Pharmacologically induced long QT type 2 can be rescued by activation of IKs with

(16)

benzodiazepine R-L3 in isolated guinea pig cardiomyocytes. J Cardiovasc Pharmacol., 54, 169-777.

Roden DM, Lazzara R, Rosen M, Schwartz PJ, Towbin J, Vincent GM (1996), for the SADS Foundation Task Force on LQTS. Multiple mechanisms in the long QT syndrome: current knowledge, gaps, and future directions. Circulation, 94, 1996 –2012.

Sanguinetti MC, Curran ME, Zou A, Shen J, Spector PS, Atkinson DL, Keating MT (1996).

Coassembly of K(V)LQT1 and minK (IsK) proteins to form cardiac I(Ks) potassium channel.

Nature 384(6604):80-83.

Salata JJ, Jurkiewicz NK, Jow B, Folander K, Guinosso PJ, Raynor B, Swanson R, Fermini B (1996). IK of rabbit ventricle is composed of two currents: evidence for IKs. Am J Physiol, 271, H2477-H2489.

Salata JJ, Jurkiewicz NK, Wang J, Evans BE, Orme HT, Sanguinetti MC (1998). A novel benzodiazepine that activates cardiac slow delayed rectifier K+ currents. Mol Pharmacol 54:220–230.

Seebohm G, Pusch M, Chen J, Sanguinetti MC (2003). Pharmacological activation of normal and arrhythmia-associated mutant KCNQ1 potassium channels. Circ Res 93, 941–947.

Varró A, B. Baláti, N. Iost, J. Takács, L. Virág, D. A. Lathrop, C. Lengyel, L. Tálosi, J. Gy.

Papp (2000). The role of the delayed rectifier component IKs in dog ventricular muscle and Purkinje fibre repolarization. Journal of Physiology (London), 523, 67-81.

Virág L, N Iost, M Opincariu, J Szolnoky, J Szécsi, G Bogáts, P Szenohradszky, A Varró, JGy Papp (2001). The slow component of the delayed rectifier potassium current in undiseased human ventricular myocytes. Cardiovasc. Res., 49, 790-797.

Volders PG, Stengl M, van Opstal JM, Gerlach U, Spatjens RL, Beekman JD, Sipido KR, Vos MA (2003). Probing the contribution of IKs to canine ventricular repolarization: key role for beta-adrenergic receptor stimulation. Circulation 107, 2753-2760.

(17)

Xu X, Salata JJ, Wang J, Wu Y, Yan GX, Liu T, Marinchak RA, Kowey PR (2002).

Increasing IKs corrects abnormal repolarization in rabbit models of acquired LQT2 and ventricular hypertrophy. Am J Physiol 283, H664–H670.

Zittoun J, Marquet J, Pilorget JJ, Tonetti C, De Gialluly E (1991). Comparative effect of 6S, 6R and 6RS leucovorin on methotrexate rescue and on modulation of 5-fluorouracil. Br J Cancer, 63, 885-888.

(18)

Figure legends.

Figure 1. Synthesis of L-364,373 (4, ZS_1270B) and it's enantiomer (7, ZS_1271B). 1. THF, 2 h, 0 °C, 2. MeOH/H2O, NaOH, 2 days (Evans et al. 1987), 3. 6 equiv. Cs2CO3, 2 equiv.

MeI, dry acetone, rt, 10 h, 89%.

Figure 2. Panel A. Effect of ZS_1270B on the slow delayed rectifier K+ current (IKs). IKs

current traces recorded from rabbit ventricular myocyte under control conditions (top) and in the presence of 1 µM ZS_1270B (bottom). Experiments were performed in the presence of the selective IKr blocker dofetilide (1 µM), added to the bath solution. Panel B. Corresponding IKs- tail current-voltage relationship before and after application of 1 µM ZS_1270B. The inset in the top of the figure shows the voltage protocol applied during measurements. Values represent mean ± SEM, n=7, *p<0.05.

Figure 3. Panel A. Effect of ZS_1271B on the slow delayed rectifier K+ current (IKs). IKs current traces recorded from rabbit ventricular myocyte under control conditions (top) and in the presence of 1 µM ZS_1271B (bottom). Experiments were performed in the presence of the selective IKr blocker dofetilide (1 µM), added to the bath solution. Panel B. Corresponding IKs- tail current-voltage relationship before and after application of 1 µM ZS_1271B. The inset in the top of the figure shows the voltage protocol applied during measurements. Values represent mean ± SEM, n=7, *p<0.05.

Figure 4. Guinea pig ventricular papillary muscle action potential recordings in the absence of any sympathetic agonist before and after 40 minutes of superfusion with the RL-3 enantiomers 1 µM ZS_1270B (left panel) and 1 µM ZS_1271B (right panel), respectively.

Stimulation frequency was 1 Hz.

(19)

Table 1. The electrophysiological effects of the RL-3 enantiomers ZS_1271B (l µM, top panel) and 1 µM ZS_1270B (1 µM, bottom panel), respectively, in guinea pig right papillary muscle at recorded at the basic cycle length of 1000 ms. Drug effect was measured after 40 min of exposure. Values are given Mean±SEM, * p < 0.05 vs control.

Parameters RP (mV) APA (mV) APD50 (ms) APD90 (ms) Vmax (V/s)

Control (n=4)

-84.2 ± 0.6 106.2 ± 2.2 149.0 ± 6.2 182.1 ± 10.1 238.0 ± 9.2

1 µM ZS_1271B (n=4)

-84.3 ± 0.5 107.7 ± 2.4 159.2 ± 6.2* 209.2 ± 8.0* 233.9 ± 9.4

Parameters RP (mV) APA (mV) APD50 (ms) APD90 (ms) Vmax (V/s)

Control (n=4)

-85.2 ± 0.3 109.4 ± 0.3 149.4 ± 5.1 184.7 ± 8.1 231.0 ± 6.2

1 µM ZS_1270B (n=4)

-84.6 ± 1.6 108.6 ± 1.7 141.0 ± 5.4* 165.7 ± 11.3* 229.9 ± 8.4

(20)

Figure 1. Synthesis of L-364,373 (4, ZS_1270B) and it's enantiomer (7, ZS_1271B). 1. THF, 2 h, 0 °C, 2.

MeOH/H2O, NaOH, 2 days (Evans et al. 1987), 3. 6 equiv. Cs2CO3, 2 equiv. MeI, dry acetone, rt, 10 h, 89%.

81x60mm (300 x 300 DPI)

(21)

Figure 2. Panel A. Effect of ZS_1270B on the slow delayed rectifier K+ current (IKs). IKs current traces recorded from rabbit ventricular myocyte under control conditions (top) and in the presence of 1 µM ZS_1270B (bottom). Experiments were performed in the presence of the selective IKr blocker dofetilide (1

µM), added to the bath solution. Panel B. Corresponding IKs-tail current-voltage relationship before and after application of 1 µM ZS_1270B. The inset in the top of the figure shows the voltage protocol applied

during measurements. Values represent mean ± SEM, n=7, *p<0.05.

81x60mm (300 x 300 DPI)

(22)

Figure 3. Panel A. Effect of ZS_1271B on the slow delayed rectifier K+ current (IKs). IKs current traces recorded from rabbit ventricular myocyte under control conditions (top) and in the presence of 1 µM ZS_1271B (bottom). Experiments were performed in the presence of the selective IKr blocker dofetilide (1

µM), added to the bath solution. Panel B. Corresponding IKs-tail current-voltage relationship before and after application of 1 µM ZS_1271B. The inset in the top of the figure shows the voltage protocol applied

during measurements. Values represent mean ± SEM, n=7, *p<0.05.

81x60mm (300 x 300 DPI)

(23)

Figure 4. Guinea pig ventricular papillary muscle action potential recordings in the absence of any sympathetic agonist before and after 40 minutes of superfusion with the RL-3 enantiomers 1 µM ZS_1270B

(left panel) and 1 µM ZS_1271B (right panel), respectively. Stimulation frequency was 1 Hz.

81x60mm (300 x 300 DPI)

Ábra

Table 1. The electrophysiological effects of the RL-3 enantiomers ZS_1271B (l µM, top panel) and 1 µM ZS_1270B (1  µM, bottom panel), respectively, in guinea pig right papillary muscle at recorded at the basic cycle length of 1000 ms
Figure 1. Synthesis of L-364,373 (4, ZS_1270B) and it's enantiomer (7, ZS_1271B). 1. THF, 2 h, 0 °C, 2
Figure 2. Panel A. Effect of ZS_1270B on the slow delayed rectifier K+ current (IKs). IKs current traces  recorded from rabbit ventricular myocyte under control conditions (top) and in the presence of 1 µM  ZS_1270B (bottom)
Figure 3. Panel A. Effect of ZS_1271B on the slow delayed rectifier K+ current (IKs). IKs current traces  recorded from rabbit ventricular myocyte under control conditions (top) and in the presence of 1 µM  ZS_1271B (bottom)
+2

Hivatkozások

KAPCSOLÓDÓ DOKUMENTUMOK

Based on the DC-link voltage, grid currents and grid voltages, the rectifier model calculates the output current of the rectifier (I RECT ) in the n th step.. Meanwhile the inverter

Contribution of ion currents to beat-to-beat variability of action potential duration in canine ventricular

We also showed that in freshly isolated ventricular myocytes, peak I Ca,L density and Cav1.2α (primary subunit of the L-type Ca 2+ channel protein) channel protein were 25–30%

Key words: Phosphodiesterase-3 inhibitors, Transient outward potassium current, Pharmacology, Ventricular fibrillation, Sudden Cardiac Death, Electrophysiology, Early

Contribution of ion currents to beat-to-beat variability of action potential duration in canine ventricular myocytes.. Norbert Szentandrássy &amp; Kornél Kistamás &amp; Bence

In the present study, [Ca 2+ ] i -dependent contribution of I K1 to repolarization was directly evaluated in isolated canine ventricular myocytes and papillary muscle

to determine the potential role of speckle myocardial imaging (SMI) for identifying left ventricular dysfunction in patients with isolated left ventricular non-compaction

Nowadays climate change has become a reality. As a result, the focus has been on avoiding, mitigating and adjusting to expected adverse effects. In this study, we focus on