Appraisal of state-of-the art
Troubleshooting problems with in vitro screening of drugs for QT interval prolongation using HERG K+ channels expressed in mammalian cell lines and Xenopus oocytes

https://doi.org/10.1016/S1056-8719(03)00041-8Get rights and content

Abstract

The majority of drugs associated with QT interval prolongation share an ability to inhibit ionic currents passed by HERG potassium channels. One method of screening new chemical entities (NCEs) for QT prolonging potential is therefore to use heterologous systems expressing HERG channels. Such systems are also of value in the understanding of the function, kinetics, sorting, pharmacological sensitivities, and important molecular determinants of the HERG potassium channel. The methods for incorporating the HERG potassium channel into cells and measuring the consequent current are a mixture of techniques that are standard (for heterologous expression of most ion channels) and individualised to HERG. This review presents a selection of the most commonly used methods for examining heterologous HERG currents, as well as introducing some of the technical problems that may be encountered and their solutions. In mammalian cell lines, problems such as fragile membranes, high leak currents, inability to form a gigaseal, diminished HERG current, endogenous transient outward current, altered kinetics, and even occasional run down can interfere with measurements. In Xenopus oocytes, endogenous chloride currents, insufficient superfusate flow, diminished HERG current and HERG current ‘run up’ may create difficulties.

Introduction

HERG is a potassium (K+) channel α subunit that comprises the conducting pore mediating the current in ventricular myocytes known as the ‘rapid’ delayed rectifier current (IKr), and pharmacological blockade of this current or mutation of the channel can lead to long QT syndrome (LQTS), a disorder of the cardiac rhythm in which the sufferers, who can be otherwise healthy, are subject to a risk of fainting or sudden arrhythmogenic death Tseng, 2001, Witchel & Hancox, 2000. LQTS derives its name from its defining clinical feature: on the electrocardiogram the duration of time between the Q wave and the completion of the T wave is prolonged. When the HERG protein was successfully expressed heterologously, the characteristics of the observed potassium ion currents fit relatively well with what was known of IKr in ventricular myocytes Sanguinetti et al., 1995, Trudeau et al., 1995. Subsequently, a large number of mutations were found to occur in HERG in families suffering from LQTS Sanguinetti et al., 1996, Splawski et al., 2000. In addition to mutations, pharmacological inhibition of this channel was shown to be associated with LQTS and could also lead to a risk of sudden death (Sanguinetti et al., 1995). Because the HERG channel has been associated mechanistically with the risk of sudden death for pharmaceutical agents from a wide variety of drug classes, it has attracted a lot of attention in the pharmaceutical industry, among clinical guidelines, and particularly with government regulators Committee for Proprietary Medicinal Products, 1997, Crumb & Cavero, 1999, Food and Drug Administration USA, 2002, Haverkamp et al., 2000, Redfern et al., 2003. Ultimately, this has resulted in the withdrawal or reclassification of drugs that had already been approved for the USA and European markets (e.g., Committee on Safety of Medicines, 2001).

Much effort has been made to exploit the heterologous expression of HERG channels to evaluate in vitro the risk of potential drug-induced repolarisation anomalies. Although there have been a variety of efforts to incorporate high-throughput strategies into drug screening processes for HERG blockade, using either fluorescence (Netzer, Ebneth, Bischoff, & Pongs, 2001) or radioligand binding (Finlayson, Turnbull, January, Sharkey, & Kelly, 2001) based assays, the vast majority of the work has involved electrophysiological measurements of currents from single cells, and as of this date, the electrophysiological approach remains the “gold standard” for testing HERG channel blockade. These electrophysiological recordings are typically in the voltage-clamp mode, and include whole cell patch clamp, perforated patch clamp, two (sharp) electrode voltage clamp, and single-channel recordings.

In electrophysiological recordings in the voltage-clamp mode, in addition to conductance changes derived from actions of pharmacological agents, changes in channel state can be inferred from the current elicited by step depolarisations. For the purposes of interpreting current recordings, it can be helpful to think of K+ channels as a membrane spanning K+ selective pore with two doors or gates (the activation and inactivation gates) that open and close in a voltage- and time-dependent manner. For K+ ions to pass through the pore, generating a current flow across the membrane, both gates must be simultaneously open (see Fig. 1). Closure of either gate results in zero current flow. The gates are controlled by the membrane potential; upon depolarisation (i.e. the membrane potential changing to a more positive voltage), the activation gate opens (a process called activation), and upon repolarisation (i.e. the membrane potential changing to a more negative voltage approaching EK, which is approximately −80 mV in physiological salt solutions), the activation gate closes (deactivation). By contrast, the inactivation gate closes with depolarisation (a process called inactivation), and opens with repolarisation (recovery from inactivation). In cell membranes with large numbers of ion channels, the speed of the gates' response to voltage changes (the kinetics of the channel) is what controls the time course and amplitude of the current. If the movement of the two gates were simultaneous, instantaneous, and occurred in all channels, there would never be any current flowing through the channel, because one of the gates would always be closed. However, typically, in A-type inactivating K+ channels, the activation gate responds to voltage changes faster than the inactivation gate, which results in a transient current upon membrane depolarisation. For the HERG channel, the inactivation gate responds to voltage changes faster than the activation gate, hence, it displays a current that is radically different from A-type K+ channels.

The voltage protocols used to identify HERG are designed to display the idiosyncratic currents associated with HERG, which are described as “resurgent” (Robertson, 2000). When the membrane is at depolarised potentials, much of the HERG channel population is in an inactivated state in which current does not flow through the channel; in the process of the membrane returning to a repolarised state, the HERG channels recover from inactivation and the channels linger in a highly stable open state before closing. The result of this transition is that the channels can transiently conduct more current as the membrane potential moves toward EK (see Fig. 2A), a process that would otherwise be thought to reduce K+ current magnitude, due to the reduced electromotive driving force for outward K+ ion movement.

The cells that are used for these electrophysiological measurements can be those that naturally have HERG current in them (‘native’ IKr currents, presumably mediated by HERG-encoded channels), such as isolated ventricular cardiomyocytes (Jurkiewicz & Sanguinetti, 1993), atrial cell lines (e.g., mouse AT-1 cells; Busch et al., 1998), or neuroblastoma cell lines (e.g., SH-SY5Y cells; Arcangeli et al., 1998), but the majority of studies employ cells that have the HERG channel introduced into them using molecular biological methods, a process known as heterologous expression. One advantage of heterologous expression is that one can choose what cell type to add the HERG channel current to, and such investigations invariably choose cell types that are much less electrically active than the cells that have native HERG. This means that the cells that have heterologous HERG are virtually without other contaminating currents that will be evident during the electrophysiological examination, while cells with native HERG often have other current types that must be excluded pharmacologically, ionically, or by other electrophysiological means. This is of great value in the mechanistic investigation of drug–channel interactions, and is one reason why heterologous HERG expression is increasingly favoured over IKr from cardiomyocytes during drug screening. Also, native IKr is typically of comparatively small amplitude, and signal-to-noise levels are typically better in the heterologous systems used because heterologous systems allow for high levels of the channel gene to be expressed. The most important advantage that heterologous systems offer over cells with native HERG is that mutated versions of the HERG channel may be compared with the unmutated (wild type) version of the channel in the heterologous systems. The examination of the mutated channels allows for the examination of the molecular determinants of channel blockade, as well as for the investigation of the role of functional domains within the channel in state-dependent blockade (e.g., Milnes et al., 2003, Mitcheson et al., 2000).

While a complete discussion of the methods for heterologous expression and electrophysiology would be too long and outside the scope of a review such as this, good reviews can be found for the use of Xenopus oocytes (Wagner, Friedrich, Setiawan, Lang, & Broer, 2000), patch clamp (Cahalan & Neher, 1992), transfection (Schenborn, 2000), and mammalian cell line care (Pollard, 1997). This review will focus specifically on issues relating to the experimentation on the pharmacology of HERG, concentrating on the options that are used most commonly, and will discuss approaches for troubleshooting the problems that our laboratories have observed in this endeavour.

Section snippets

Putative β subunits: hMiRP1 and minK

Although HERG encodes a K+ channel subunit that passes current with many similarities to native IKr, questions have arisen as to whether or not heterologous expression of HERG accurately recapitulates the native current, or whether this requires coexpression with the cDNA for an auxiliary subunit. The first endogenous channel subunit found to complex with HERG was minK (McDonald et al., 1997). This short integral membrane protein had previously been identified as forming a complex with KvLQT1

Mammalian cell lines

The advantages of using mammalian cell lines compared to Xenopus oocytes as a heterologous expression system for electrophysiological investigation of HERG are based around the concept that measurements in mammalian cell lines would be more representative of what would actually occur in the physiological case in human patients. The advantages include being able to make measurements at 37 °C and the consequently more rapid kinetics for HERG current (IHERG) compared to those at ambient

Attaining gigaseal and trypsinisation

Attaining gigaseal in heterologous or transfected cell lines is highly influenced by the state of the membrane. Empirically, we have found that the inability to achieve a gigaseal usually means that the membranes have not been trypsinised recently enough. We have found that it is possible to briefly trypsinise dishes of cells that will not form gigaseals, and thus make them more amenable to patch clamp; such a trypsin treatment does not appear to alter the profile of whole cell IHERG in our

Xenopus oocytes

Xenopus oocytes are an important heterologous system for measuring HERG currents, in part because the HERG current in this system is comparatively large and well defined. In addition, the Xenopus oocyte system provides an opportunity for gathering much larger quantities of data from individual cells than the whole-cell patch-clamp system because recordings are more stable due to the absence of significant run down of HERG current when expressed in oocytes, and to the recording set-up being more

Conclusions

The issue of pharmacological HERG blockade continues to increase in importance within the spheres of drug development and pharmaceutical regulation. The state of the field now requires an ability to acquire medium to high throughput HERG data; currently, this is being approached with medium- to high-throughput automated multi-well voltage-clamp systems (although some efforts have been made with fluorescent indicators of resting membrane potential as well as with radiolabeled drug binding

Acknowledgements

We gratefully acknowledge all our collaborators, neighbours, and friends of the Cardiovascular Research Labs over the years for sharing their protocols, anecdotes, and ideas with us on this topic. We acknowledge support of our work by the British Heart Foundation, Wellcome Trust, the Medical Research Council and Pfizer Global Research and Development. We also thank Stuart Murdoch and Stuart David for the description of accumulation. If your lab has any experience on troubleshooting HERG that

References (71)

  • Z. Zhou et al.

    Properties of HERG channels stably expressed in HEK 293 cells studied at physiological temperature

    Biophysical Journal

    (1998)
  • G.W. Abbott et al.

    Disease-associated mutations in KCNE potassium channel subunits (MiRPs) reveal promiscuous disruption of multiple currents and conservation of mechanism

    FASEB Journal

    (2002)
  • A. Arcangeli et al.

    HERG- and IRK-like inward rectifier currents are sequentially expressed during neuronal development of neural crest cells and their derivatives

    European Journal of Neuroscience

    (1997)
  • A. Arcangeli et al.

    Modulation of HERG current and herg gene expression during retinoic acid treatment of human neuroblastoma cells: Potentiating effects of BDNF

    Journal of Neurobiology

    (1999)
  • J. Barhanin et al.

    KvLQT1 and Isk (minK) proteins associate to form the IKs cardiac potassium channel

    Nature

    (1996)
  • E.M. Blumenthal et al.

    The minK potassium channel exists in functional and nonfunctional forms when expressed in the plasma membrane of Xenopus oocytes

    Journal of Neuroscience

    (1994)
  • A.E. Busch et al.

    Blockade of HERG channels by the class III antiarrhythmic azimilide: Mode of action

    British Journal of Pharmacology

    (1998)
  • C. Chouabe et al.

    HERG and KvLQT1/IsK, the cardiac K+ channels involved in long QT syndromes, are targets for calcium channel blockers

    Molecular Pharmacology

    (1998)
  • Committee for Proprietary Medicinal Products (1997). Points to consider: The assessment for the potential for QT...
  • Committee on Safety of Medicines

    QT interval prolongation with antipsychotics

    Current Problems in Pharmacovigilance

    (2001)
  • P. Daleau et al.

    Erythromycin blocks the rapid component of the delayed rectifier potassium current and lengthens repolarization of guinea pig ventricular myocytes

    Circulation

    (1995)
  • L. Faravelli et al.

    A HERG-like K+ channel in rat F-11 DRG cell line: Pharmacological identification and biophysical characterization

    Journal of Physiology (London)

    (1996)
  • Food and Drug Administration [FDA] USA (2002). Safety pharmacology studies for assessing the potential for delayed...
  • G.B. Gurrola et al.

    A toxin to nervous, cardiac, and endocrine ERG K+ channels isolated from Centruroides noxius scorpion venom

    FASEB Journal

    (1999)
  • J.C. Hancox et al.

    Time course and voltage dependence of expressed HERG current compared with native ‘rapid’ delayed rectifier K current during the cardiac ventricular action potential

    Pflügers Archiv

    (1998)
  • S.F. Hausdorff et al.

    Functional characterization of a minimal K+ channel expressed from a synthetic gene

    Biochemistry

    (1991)
  • W. Haverkamp et al.

    The potential for QT prolongation and pro-arrhythmia by non-anti-arrhythmic drugs: Clinical and regulatory implications. Report on a Policy Conference of the European Society of Cardiology

    Cardiovascular Research

    (2000)
  • Y. Jiang et al.

    X-ray structure of a voltage-dependent K+ channel

    Nature

    (2003)
  • N.K. Jurkiewicz et al.

    Rate-dependent prolongation of cardiac action potentials by a methanesulfonanilide class III antiarrhythmic agent. Specific block of rapidly activating delayed rectifier K+ current by dofetilide

    Circulation Research

    (1993)
  • M.E. Jurman et al.

    Visual identification of individual transfected cells for electrophysiology using antibody-coated beads

    BioTechniques

    (1994)
  • K. Kamiya et al.

    Open channel block of HERG K+ channels by vesnarinone

    Molecular Pharmacology

    (2001)
  • F. Lesage et al.

    Are Xenopus oocytes unique in displaying functional IsK channel heterologous expression?

    Receptors and Channels

    (1993)
  • A.J. Levi et al.

    A method for making rapid changes of superfusate whilst maintaining temperature at 37 degrees C

    Pflügers Archiv

    (1996)
  • J.D. Lewis et al.

    The role of the cap structure in RNA processing and nuclear export

    European Journal of Biochemistry

    (1997)
  • R. Mazhari et al.

    Molecular interactions between two long-QT syndrome gene products, HERG and KCNE2, rationalized by in vitro and in silico analysis

    Circulation Research

    (2001)
  • Cited by (117)

    • Tricyclic hydrocarbon fluorene attenuates ventricular ionic currents and pressure development in the navaga cod

      2023, Comparative Biochemistry and Physiology Part - C: Toxicology and Pharmacology
    • hERG liability classification models using machine learning techniques

      2019, Computational Toxicology
      Citation Excerpt :

      Traditional and gold-standard patch clamp electrophysiology studies [19–21], radio-ligand binding assays [22], cell-based fluorescence assays [23] and Rubidium flux assays [24] are commonly used assay methods (in-vitro) to assess the hERG liability of molecules. However, these experimental assays are often time consuming, costly and laborious [25,26]. Many automated high throughput assay methods have been developed by miniaturizing the hERG assay [20,27–31], but the results from these have shown significant levels of variability, which affects the reliability and applicability of these assessments [32].

    • Interactions between amiodarone and the hERG potassium channel pore determined with mutagenesis and in silico docking

      2016, Biochemical Pharmacology
      Citation Excerpt :

      Previous experiments using Xenopus oocytes yielded an amiodarone IC50 value for IhERG of 9.8 μM [10] whilst in mammalian expression systems IhERG IC50 values between ∼26 and 300 nM were reported [12–14,16,17]. Amiodarone is highly lipophilic and for such agents the use of Xenopus oocytes can markedly underestimate blocking potency due to drug accumulation in the yolk sac [39,40]. Amiodarone has also been shown to produce greater IhERG block at physiological (37 °C) than at ambient (23 °C) temperature (IC50 of 0.30 μM versus 0.56 μM, respectively) [17].

    • In silico assessment of kinetics and state dependent binding properties of drugs causing acquired LQTS

      2016, Progress in Biophysics and Molecular Biology
      Citation Excerpt :

      Furthermore, the kinetics of drug binding and unbinding to these two states can vary significantly (Hill et al., 2014). As a consequence, if the relative occupancies of open and inactivated states evoked by two stimulation protocols are different then the apparent IC50 from these protocols will be different (Milnes et al., 2010; Witchel et al., 2002; Yao et al., 2005). Furthermore, even if two drugs had the same apparent affinities for the open and inactivated states but they had different kinetics of binding and unbinding then they could also have different apparent overall affinities depending on the protocol used to measure channel block.

    View all citing articles on Scopus
    View full text