Associate editor: H. Bönisch
Human ether-a-go-go-related (HERG) gene and ATP-sensitive potassium channels as targets for adverse drug effects

https://doi.org/10.1016/j.pharmthera.2006.03.002Get rights and content

Abstract

Torsades de pointes (TdP) arrhythmia is a potentially fatal form of ventricular arrhythmia that occurs under conditions where cardiac repolarization is delayed (as indicated by prolonged QT intervals from electrocardiographic recordings). A likely mechanism for QT interval prolongation and TdP arrhythmias is blockade of the rapid component of the cardiac delayed rectifier K+ current (IKr), which is encoded by human ether-a-go-go-related gene (HERG). Over 100 non-cardiovascular drugs have the potential to induce QT interval prolongations in the electrocardiogram (ECG) or TdP arrhythmias. The binding site of most HERG channel blockers is located inside the central cavity of the channel. An evaluation of possible effects on HERG channels during the development of novel drugs is recommended by international guidelines. During cardiac ischaemia activation of ATP-sensitive K+ (KATP) channels contributes to action potential (AP) shortening which is either cardiotoxic by inducing re-entrant ventricular arrhythmias or cardioprotective by inducing energy-sparing effects or ischaemic preconditioning (IPC). KATP channels are formed by an inward-rectifier K+ channel (Kir6.0) and a sulfonylurea receptor (SUR) subunit: Kir6.2 and SUR2A in cardiac myocytes, Kir6.2 and SUR1 in pancreatic β-cells. Sulfonylureas and glinides stimulate insulin secretion via blockade of the pancreatic β-cell KATP channel. Clinical studies about cardiotoxic effects of sulfonylureas are contradictory. Sulfonylureas and glinides differ in their selectivity for pancreatic over cardiovascular KATP channels, being either selective (tolbutamide, glibenclamide) or non-selective (repaglinide). The possibility exists that non-selective KATP channel inhibitors might have cardiovascular side effects. Blockers of the pore-forming Kir6.2 subunit are insulin secretagogues and might have cardioprotective or cardiotoxic effects during cardiac ischaemia.

Introduction

Ion channels are membrane-spanning protein complexes that allow ions to travel across the lipid bilayer and take part in fundamental cellular processes of almost all organisms. K+ channels are a large family that show K+ ion selectivity and play a central role in the electrical activity of excitable cells. Since cloning and identification of the first K+ channel gene (Shaker) from Drosophila in 1987 (Papazian et al., 1987), more than 80 genes encoding human K+ channels have been cloned. Three main classes of K+ channels can be identified on the basis of the transmembrane topology of the pore-forming α-subunits: (1) 6 transmembrane helix (6TM) voltage-gated K+ channels (Kv), which are activated by membrane depolarization; (2) 2 transmembrane helix (2TM) inward rectifier K+ channels (Kir), which are activated by intracellular factors such as G-proteins, nucleotides or polyamines; (3) 4 transmembrane helix (4TM) 2-pore K+ channels. In the case of both Kv and Kir channels, the K+ channel has a tetrameric structure formed by the assembly of 4 α-subunits (reviews in Tamargo et al., 2004, Recanatini et al., 2005). Co-expression of K+ channel β-subunits with α-subunits regulates cell surface expression, gating kinetics and drug-sensitivity of K+ channels. K+ channel β-subunits represent a diverse molecular group, which includes cytoplasmic proteins (Kvβ1–3, K+ channel-interacting proteins (KChIP) and K+ channel-associated proteins (KChAP)) which interact with the intracellular domains of Kv channels, single transmembrane spanning domains, such as minK and minK-related proteins (MiRP) encoded by the KCNE gene family, and ATP-binding cassette (ABC) transport-related proteins, such as the sulfonylurea receptor (SUR) for the inward rectifiers Kir6.0 (review in Pourrier et al., 2003). With regard to the structural characteristics of the pore-forming region of the K+ channels, several evidences have been provided by crystallographic studies carried out on bacterial K+ channels first by the recent Nobel Laureate R. MacKinnon (Doyle et al., 1998).

Action potentials (AP) of the cardiac ventricular myocyte can last for several hundred milliseconds (about 200–400 ms; Fig. 1). This prolonged depolarization (plateau) phase is essential for normal excitation-contraction coupling and renders the cell relatively refractory to premature excitation. Cardiac APs are long because the many types of K+ channels that repolarize the membrane either activate slowly, inactivate rapidly, or conduct less current at positive membrane potentials. The principal K+ currents participating in the repolarization of the AP in ventricular myocytes are (Fig. 1 top left): a transient outward K+ current (Ito), the delayed rectifier K+ current (IK) and the inward rectifier K+ current (IK1). Each of these currents has relatively different importance at different stages of the AP. Ito contributes to the initial repolarization phase of the AP—the “notch”. IK is the major time-dependent outward current that participates in mediating repolarization of the cardiac AP and, therefore, plays an important role in determining the duration of the AP (Noble & Tsien, 1969). The delayed rectifier K+ current IK in ventricular myocytes consists of a rapid component (cardiac rapidly activating delayed rectifier K+ current, IKr) and a slow component (cardiac slowly activating delayed rectifier K+ current, IKs). These 2 components have first been described by Sanguinetti and Jurkiewicz (1990) and have also been detected in human ventricular cells (Li et al., 1996). IK1 determines the resting membrane potential and also contributes to the terminal repolarization phase. During ischaemia, the ATP-sensitive K+ (KATP) current (IKATP) becomes important and contributes to AP shortening in ischaemic myocardium (reviews in Rees & Curtis, 1996, Sanguinetti & Keating, 1997).

This review describes the main repolarizing K+ channels in the heart under physiological conditions (IKr/human ether-a-go-go-related gene (HERG) channels) and under ischaemic conditions (KATP channels) as targets for cardiotoxic effects of drugs which act as inhibitors of the open probabilities of these channels. Furthermore, this review also describes the pore-forming Kir6.2 subunit of the KATP channel as a target for both cardiac and hypoglycemic effects of drugs, since KATP channels also play a major role in the regulation of insulin secretion from pancreatic β-cells and pancreatic β-cells express the same pore-forming Kir6.2 subunit of the KATP channel as cardiac myocytes. HERG and KATP channels serve as examples for drug targets involved in mechanisms underlying cardiotoxic effects and (in case of KATP channels) also in mechanisms leading to alterations of blood glucose homeostasis.

Section snippets

Torsades de pointes cardiac arrhythmias

The opening of K+ channels terminates the AP of cardiac myocytes (Fig. 1). The duration of the QT interval is a measure of the time required for depolarization and repolarization of the cardiac ventricles. Dysfunction of cardiac K+ channels in congenital long QT syndrome (LQTS) produces a prolonged AP. Similarly, drugs that block cardiac K+ channels can prolong AP. Prolonged repolarization becomes evident as a longer QT interval in the electrocardiogram (ECG). Inhibition of IKr currents leads

ATP-sensitive K+ channels in cardiac myocytes and in pancreatic β-cells

The KATP channel is widely distributed in many tissues and cell types, including heart, pancreatic β-cells, specific regions of the brain (including hypothalamus, hippocampus, substantia nigra, neocortex), skeletal and smooth muscles. In the brain KATP channels contribute to glucose sensing, seizure protection and ischaemic protection. In skeletal muscle KATP channels play roles in glucose uptake, K+ efflux, fatigue and recovery. In vascular smooth muscle KATP channels regulate muscle tone (for

Conclusions

Within the last 15 years the torsadogenic potential of drugs has been the most frequent reason (besides drug-induced hepatotoxicity) for withdrawal of drugs from the market or for severely restriction of their approved use. HERG channels have been identified as targets for these cardiotoxic drugs, and the last decade has seen a marked progress in the understanding of both the molecular interactions between drugs and the HERG channel and in the mechanisms of drug-induced TdP cardiac arrhythmias.

Acknowledgments

I wish to thank Mrs. A. Wessel for preparing the figures and Dr. K. Spicher for helpful comments.

References (235)

  • M.E. Curran et al.

    A molecular basis for cardiac arrhythmia: HERG mutations cause long QT syndrome

    Cell

    (1995)
  • D. Fernandez et al.

    Physicochemical features of the hERG channel drug binding site

    J Biol Chem

    (2004)
  • A. Fujita et al.

    Molecular aspects of ATP-sensitive K+ channels in the cardiovascular system and K+ channel openers

    Pharmacol Ther

    (2000)
  • S. Haider et al.

    Focus on Kir6.2: A key component of the ATP-sensitive potassium channel

    J Mol Cell Cardiol

    (2005)
  • P.J. Hanley et al.

    KATP channels and preconditioning: A re-examination of the role of mitochondrial KATP channels and an overview of alternative mechanisms

    J Mol Cell Cardiol

    (2005)
  • N. Inagaki et al.

    A family of sulfonylurea receptors determines the pharmacological properties of ATP-sensitive K+ channels

    Neuron

    (1996)
  • G.C. Kane et al.

    Cardiac KATP channels in health and disease

    J Mol Cell Cardiol

    (2005)
  • M.T. Keating et al.

    Molecular and cellular mechanisms of cardiac arrhythmias

    Cell

    (2001)
  • J. Kikuta et al.

    Carvedilol blocks cardiac KATP and KG but not IK1 channels by acting at the bundle-crossing regions

    Eur J Pharmacol

    (2006)
  • L. Aguilar-Bryan et al.

    Molecular biology of adenosine triphosphate-sensitive potassium channels

    Endocrinol Rev

    (1999)
  • L. Aguilar-Bryan et al.

    Cloning of the β cell high-affinity sulfonylurea receptor: A regulator of insulin secretion

    Science

    (1995)
  • L. Aguilar-Bryan et al.

    Toward understanding the assembly and structure of KATP channels

    Physiol Rev

    (1998)
  • C.L. Anderson et al.

    Most LQT2 mutations reduce Kv11.1 (hERG) current by a class 2 (trafficking-deficient) mechanism

    Circulation

    (2006)
  • J.F. Antcliff et al.

    Functional analysis of a structural model of the ATP-binding site of the KATP channel Kir6.2 subunit

    EMBO J

    (2005)
  • C. Antzelevitch et al.

    Clinical relevance of cardiac arrhythmias generated by afterdepolarizations. Role of M cells in the generation of U waves, triggered activity and torsade de pointes

    J Am Coll Cardiol

    (1989)
  • C. Antzelevitch et al.

    Heterogeneity within the ventricular wall. Electrophysiology and pharmacology of epicardial, endocardial, and M cells

    Circ Res

    (1991)
  • C. Antzelevitch et al.

    Brugada syndrome: A decade of progress

    Circ Res

    (2002)
  • C. Antzelevitch et al.

    Electrophysiological effects of ranolazine, a novel antianginal agent with antiarrhythmic properties

    Circulation

    (2004)
  • C. Antzelevitch et al.

    Brugada syndrome: Report of the second consensus conference

    Circulation

    (2005)
  • F.M. Ashcroft

    ATP-sensitive potassium channelopathies: Focus on insulin secretion

    J Clin Invest

    (2005)
  • F.M. Ashcroft et al.

    Electrophysiology of the pancreatic β-cell

    Prog Biophys Mol Biol

    (1991)
  • F.M. Ashcroft et al.

    Glucose induces closure of single potassium channels in isolated rat pancreatic beta-cells

    Nature

    (1984)
  • A.P. Babenko et al.

    Reconstituted human cardiac KATP channels: Functional identity with the native channels from the sarcolemma of human ventricular cells

    Circ Res

    (1998)
  • T. Baukrowitz et al.

    KATP channels gated by intracellular nucleotides and phospholipids

    Eur J Biochem

    (2000)
  • L. Bianchi et al.

    Herg encodes a K+ current highly conserved in tumors of different histogenesis: A selective advantage for cancer cells?

    Cancer Res

    (1998)
  • C. Bollensdorff et al.

    Amiloride derivatives are potent blockers of KATP channels

    Naunyn-Schmiedeberg's Arch Pharmacol

    (2001)
  • R. Brugada et al.

    Sudden death associated with short-QT syndrome linked to mutations in HERG

    Circulation

    (2004)
  • R. Brugada et al.

    Short QT syndrome

    CMAJ

    (2005)
  • J. Bryan et al.

    Toward linking structure with function in ATP-sensitive K+ channels

    Diabetes

    (2004)
  • J. Bryan et al.

    Insulin secretagogues, sulfonylurea receptors and KATP channels

    Curr Pharm Des

    (2005)
  • O. Casis et al.

    Mechanism of action of a novel human ether-a-go-go-related gene channel activator

    Mol Pharmacol

    (2006)
  • I. Cavero et al.

    ICH S7B draft guideline on the non-clinical strategy for testing delayed cardiac repolarisation risk of drugs: A critical analysis

    Expert Opin Drug Saf

    (2005)
  • I. Cavero et al.

    The use of electrocardiograms in clinical trials: A public discussion of the proposed ICH E14 regulatory guidance

    Expert Opin Drug Saf

    (2005)
  • J. Chen et al.

    Position of aromatic residues in the S6 domain, not inactivation, dictates cisapride sensitivity of HERG and eag potassium channels

    Proc Natl Acad Sci U S A

    (2002)
  • S. Claaßen et al.

    Comparison of the effects of metoclopramide and domperidone on HERG channels

    Pharmacology

    (2005)
  • C.E. Clancy et al.

    K+ channel structure-activity relationships and mechanisms of drug-induced QT prolongation

    Annu Rev Pharmacol Toxicol

    (2003)
  • J.P. Clement IV et al.

    Association and stoichiometry of KATP channel subunits

    Neuron

    (1997)
  • W.C. Cole et al.

    ATP-regulated K+ channels protect the myocardium against ischemia/reperfusion damage

    Circ Res

    (1991)
  • Committee for Proprietary Medicinal Products (CPMP/986/96; 1997). Points to consider: The assesssment of the potential...
  • D.L. Cook et al.

    Intracellular ATP directly blocks K+ channels in pancreatic B-cells

    Nature

    (1984)
  • Cited by (0)

    View full text