Allosteric modulators of the hERG K+ channel: Radioligand binding assays reveal allosteric characteristics of dofetilide analogs

https://doi.org/10.1016/j.taap.2013.10.024Get rights and content

Highlights

  • Allosteric modulators on the hERG K+ channel were evaluated in binding assays.

  • LUF6200 was identified as a potent allosteric inhibitor.

  • Potassium ions were found to behave as allosteric enhancers.

  • Positive cooperativity and distinct allosteric sites for them were proposed.

Abstract

Drugs that block the cardiac K+ channel encoded by the human ether-à-go-go gene (hERG) have been associated with QT interval prolongation leading to proarrhythmia, and in some cases, sudden cardiac death. Because of special structural features of the hERG K+ channel, it has become a promiscuous target that interacts with pharmaceuticals of widely varying chemical structures and a reason for concern in the pharmaceutical industry. The structural diversity suggests that multiple binding sites are available on the channel with possible allosteric interactions between them. In the present study, three reference compounds and nine compounds of a previously disclosed series were evaluated for their allosteric effects on the binding of [3H]astemizole and [3H]dofetilide to the hERG K+ channel. LUF6200 was identified as an allosteric inhibitor in dissociation assays with both radioligands, yielding similar EC50 values in the low micromolar range. However, potassium ions increased the binding of the two radioligands in a concentration-dependent manner, and their EC50 values were not significantly different, indicating that potassium ions behaved as allosteric enhancers. Furthermore, addition of potassium ions resulted in a concentration-dependent leftward shift of the LUF6200 response curve, suggesting positive cooperativity and distinct allosteric sites for them. In conclusion, our investigations provide evidence for allosteric modulation of the hERG K+ channel, which is discussed in the light of findings on other ion channels.

Introduction

The hERG K+ channel encoded by the hERG gene (Keating and Sanguinetti, 2001) is responsible for the rapid delayed rectifier K+ current (IKr) that plays a critical role in the repolarization of cardiomyocytes during the cardiac action potential (Hoppe et al., 2001). It is made up of large intracellular N- and C-terminal domains (Ng et al., 2011, Schönherr and Heinemann, 1996) and four identical α-subunits, each of which is formed by six α-helical transmembrane domains and a looping “pore region” (Finlayson et al., 2004, Sanguinetti and Tristani-Firouzi, 2006). In humans, blockade of the hERG K+ channel by drugs can cause excessive lengthening of the action potential, which is reflected by a QT interval prolongation in the electrocardiogram (ECG) (Hancox et al., 2008, Vandenberg et al., 2001). The excessive action potential prolongation may combine to produce and sustain Torsade de Pointes (TdP), which can be self-limiting or degenerate into ventricular fibrillation rapidly leading to death (Hancox et al., 2008, Sanguinetti and Tristani-Firouzi, 2006). Therefore, it has become a routine practice in the pharmaceutical industry to test compounds for their hERG liability during early preclinical safety assessments according to the FDA guidelines (Sanguinetti and Mitcheson, 2005). In recent years, most attention has been paid to assess the affinity for the hERG K+ channel of potential drug candidates in order to avoid and discard modest-to-high affinity compounds during the lead finding and optimization process (Gintant, 2011). However, allosteric modulation of the hERG K+ channel as an alternative way of interaction has not been studied in any details until now.

With regard to G protein-coupled receptors (GPCRs), successful drugs that mediate their effects through the allosteric modulation of target activity have already reached the market, also in view of their potential greater selectivity, potency and/or safety profile when compared to orthosteric ligands (IJzerman et al., 2001, May et al., 2007). Radioligand binding assays, in particular kinetic radioligand dissociation assays, have been widely utilized to quantify the allosteric effects of GPCR ligands (Christopoulos, 2002). As for ion channels, allosteric modulators have been reported for ligand-gated ion channels in particular. For example, GW791343 might be applied to treat inflammatory disorders and pain due to its allosteric inhibition of the P2X7 receptor (Michel et al., 2008a, Michel et al., 2008b). A negative allosteric modulator of the nicotinic acetylcholine receptor, UCI-30002, had been reported to have significant benefits as a strategy for treating nicotine addiction because of its high subtype-selectivity (Yoshimura et al., 2007). It has also become increasingly apparent that new potent and selective allosteric modulators of the GABA-A receptor and 5-HT3 receptor may replace conventional antagonists and agonists for these targets (Sancar and Czajkowski, 2011, Trattnig et al., 2012). Allosteric modulation of voltage-gated ion channels such as several types of calcium, sodium and potassium channels and their possible clinical applications has been investigated as well, but to a lesser extent. As an example, a novel quinazolinone ligand (TTA-Q4) showed a positive allosteric interaction with the T-Type calcium channel since it enhanced radioligand binding, increased affinity in a saturable manner and slowed dissociation (Uebele et al., 2009).

Radioligand binding assays as a means of studying the hERG K+ channel have been developed over the years. Two radioligands, [3H]astemizole and [3H]dofetilide, have mostly been used (Chadwick et al., 1993, Chiu et al., 2004, Finlayson et al., 2001). The purpose of the present study was to identify and investigate allosteric modulators of the hERG K+ channel using these two radioligands. The anti-depressant fluvoxamine (Mitcheson, 2003) and the channel opener PD118057 (Perry et al., 2009, Zhou et al., 2005) (Fig. 1), reported to have binding sites different from conventional hERG blockers, were selected as representative reference compounds. We identified a number of dofetilide analogs (Fig. 1) displaying allosteric modulation in a pilot experiment and these were further investigated as were the allosteric effects of potassium ions in the same binding assays. Our findings suggest potential applications for such allosteric modulators, which might provide novel solutions for drug cardiotoxicity due to blockade of the hERG K+ channel.

Section snippets

Chemicals and reagents

Astemizole, terfenadine, fluvoxamine and PD118057 were purchased from Sigma Aldrich (Zwijndrecht, The Netherlands). Dofetilide and all the LUF compounds were synthesized in our own laboratory, as published previously (Shagufta et al., 2009). [3H]Astemizole (specific activity 78.9 Ci mmol 1) and [3H]dofetilide (specific activity 70.0 Ci mmol 1) were purchased from PerkinElmer (Groningen, The Netherlands). Bovine serum albumin (BSA, fraction V) was purchased from Sigma (St. Louis, MO, USA). G418 was

Optimization of assay conditions for [3H]astemizole and [3H]dofetilide kinetic study

Assay conditions were optimized according to a general radioligand binding protocol in our laboratory (Heitman et al., 2008a). Firstly, displacement assays for [3H]astemizole and [3H]dofetilide were performed at five different amounts of hERG/HEK293 membranes (10, 15, 20, 30 and 50 μg). A suitable window of specific [3H]astemizole binding was obtained using 30 μg of membrane protein, whereas 20 μg of protein was required for [3H]dofetilide binding to achieve a similar window. These membrane

Discussion

In the present study, we evaluated the allosteric effects of a series of compounds with diverse chemical structures on the hERG K+ channel using both [3H]astemizole and [3H]dofetilide binding assays, and also addressed the influence of potassium ions on the binding of two radioligands. To the best of our knowledge, this study is the first time to utilize radioligand binding assays for the characterization of allosteric modulators for the hERG K+ channel. From these initial results, one potent

Conflict of interest statement

None.

Acknowledgments

We are grateful to Annelien Zweemer and Anouk van der Gracht for helpful discussions. This study was financially supported by the Dutch Top Institute Pharma, project number D2-201, and a grant to Z.Y. by the Chinese Scholarship Council.

References (51)

  • M.T. Keating et al.

    Molecular and cellular mechanisms review of cardiac arrhythmias

    Cell

    (2001)
  • X. Liu et al.

    Characterization of A-935142, a hERG enhancer, in the presence and absence of standard hERG blockers

    Life Sci.

    (2012)
  • J.T. Milnes et al.

    Preferential closed channel blockade of HERG potassium currents by chemically synthesised BeKm-1 scorpion toxin

    FEBS Lett.

    (2003)
  • F. Sancar et al.

    Allosteric modulators induce distinct movements at the GABA-binding site interface of the GABA-A receptor

    Neuropharmacology

    (2011)
  • M.C. Sanguinetti et al.

    Predicting drug-hERG channel interactions that cause acquired long QT syndrome

    Trends Pharmacol. Sci.

    (2005)
  • P. Smith et al.

    Measurement of protein using bicinchoninic acid

    Anal. Biochem.

    (1985)
  • S.M. Trattnig et al.

    Discovery of a novel allosteric modulator of 5-HT3 receptors: inhibition and potentiation of Cys-loop receptor signaling through a conserved transmembrane intersubunit site

    J. Biol. Chem.

    (2012)
  • J.I. Vandenberg et al.

    HERG K+ channels: friend and foe

    Trends Pharmacol. Sci.

    (2001)
  • H.M. Vargas et al.

    Scientific review and recommendations on preclinical cardiovascular safety evaluation of biologics

    J. Pharmacol. Toxicol. Methods

    (2008)
  • B. Zünkler et al.

    Allosteric interactions between terfenadine and chlorobutanol on hERG channels

    Toxicol. Lett.

    (2009)
  • A. Almilaji et al.

    AMP-activated protein kinase regulates hERG potassium channel

    Pflugers Arch. Eur. J. Physiol

    (2013)
  • C.C. Chadwick et al.

    Identification of a specific radioligand for the cardiac rapidly activating delayed rectifier K+ channel

    Circ. Res.

    (1993)
  • F. Chen et al.

    Characterization of an allosteric citalopram-binding site at the serotonin transporter

    J. Neurochem.

    (2005)
  • A. Christopoulos

    Allosteric binding sites on cell-surface receptors: novel targets for drug discovery

    Nat. Rev. Drug Discov.

    (2002)
  • H.J. Duff et al.

    High-and low-affinity sites for [3H] dofetilide binding to guinea pig myocytes

    Circ. Res.

    (1995)
  • Cited by (22)

    • The utility of hERG channel inhibition data in the derivation of occupational exposure limits

      2022, Regulatory Toxicology and Pharmacology
      Citation Excerpt :

      As expected, cardiac drugs have been shown to inhibit hERG channels and induce QT prolongation based on their pharmacology (Munawar et al., 2018; Paul et al., 2001; Sanguinetti and Tristani-Firouzi, 2006). However, hERG channel blockade may also occur through allosteric interactions at multiple binding sites (Thomas et al., 2002; Yu et al., 2014). While potent hERG inhibitors may bind internally at the S6 domain, less potent hERG inhibitors are suggested to bind externally (Mitcheson, 2003; Perry et al., 2004; Ridley et al., 2004).

    • Synthesis and biological evaluation of negative allosteric modulators of the K<inf>v</inf>11.1(hERG) channel

      2015, European Journal of Medicinal Chemistry
      Citation Excerpt :

      Likewise, NS3623 (1-[4-bromo-2-(2H-tetrazol-5-yl)phenyl]-3-[3-(trifluoro-methyl)phenyl]urea) produced a reversal of E-4031 (N-[4-[[1-[2-(6-Methyl-2-pyridinyl)ethyl]-4-piperidinyl]carbonyl]phenyl]methanesulfonamide dihydrochloride)-induced QT prolongation in both anaesthetized and conscious guinea pigs [16] In this sense, the so-called activators might be negative allosteric modulators of the action of Kv11.1 blockers, and so normalize the pharmacologically induced action potential prolongation. Indeed, the allosteric terminology at the Kv11.1 channel has been introduced in our previous study and a recent review [4,17]. Classic methods for screening allosteric modulators have been based on techniques used to identify Kv11.1 blockers, such as voltage-dependent fluorescence experiments and laborious patch clamp assays.

    • Computational investigations of hERG channel blockers: New insights and current predictive models

      2015, Advanced Drug Delivery Reviews
      Citation Excerpt :

      Similarly Yu et al. [96] demonstrated that LUF6200 is an allosteric inhibitor. The binding sites of several peptide blockers like saxitoxin and BeKm-1 are located in the extracellular parts of the hERG K+ channel [97,98]. Multiple binding sites for these diverse compounds on the hERG K+ channel imply a plausible allosteric modulation among them.

    • Sensitivity of pharmacokinetic-pharmacodynamic analysis for detecting small magnitudes of QTc prolongation in preclinical safety testing

      2015, Journal of Pharmacological and Toxicological Methods
      Citation Excerpt :

      Despite these model misspecifications, estimated concentration–effect relationships allowed still good precision of typical ΔQTc predictions, with smaller variations than from ANCOVA derived predictions. For the future it would be interesting to evaluate how the inclusion of in vitro PKPD data (e.g. ion-channel EC50 estimates (Jonker et al., 2005; Mirams et al., 2011; Watson et al., 2011)) and pharmacological concepts (e.g. binding and dissociation constants (Gabrielsson, Green, & Van der Graaf, 2010; Yu, Klaasse, Heitman, & Ijzerman, 2014)) could provide even more confidence in predicted effects and/or give a better understanding of underlying pharmacological mechanisms. Currently however, not all ion-channels that affect the QT interval (Redfern et al., 2003) are regularly assessed, making fully mechanistic modeling strategies (O'Hara, Virág, Varró, & Rudy, 2011; Ten Tusscher, Noble, Noble, & Panfilov, 2004) difficult.

    View all citing articles on Scopus
    View full text