We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Published Online:https://doi.org/10.4155/fmc.10.180

For every movement, heartbeat and thought, ion channels need to open and close. It is therefore not surprising that their malfunctioning leads to serious diseases. Currently, only approximately 10% of drugs, with a market value in excess of US$10 billion, act on ion channels. The systematic exploitation of this target class has started, enabled by novel assay technologies and fundamental advances of the structural and mechanistic understanding of channel function. The latter, which was rewarded with the Nobel Prize in 2003, has opened up an avenue for rational drug design. In this review we provide an overview of the current repertoire of screening technologies that has evolved to drive ion channel-targeted drug discovery towards new medicines of the future.

Papers of special note have been highlighted as: ▪▪ of considerable interest

Bibliography

  • Venter JC, Adams MD, Myers EW et al. The sequence of the human genome. Science291(5507),1304–1351 (2001).
  • Lehmann-Horn F, Rudel R, Dengler R, Lorkovic H, Haass A, Ricker K. Membrane defects in paramyotonia congenita with and without myotonia in a warm environment. Muscle Nerve4(5),396–406 (1981).
  • Schwartzkroin PA, Wyler AR. Mechanisms underlying epileptiform burst discharge. Ann. Neurol.7(2),95–107 (1980).
  • Riordan JR, Rommens JM, Kerem B et al. Identification of the cystic fibrosis gene: cloning and characterization of complementary DNA. Science245(4922),1066–1073 (1989).
  • Griggs RC, Nutt JG. Episodic ataxias as channelopathies. Ann. Neurol.37(3),285–287 (1995).
  • Ashcroft FM. From molecule to malady. Nature440(7083),440–447 (2006).
  • Imming P, Sinning C, Meyer A. Drugs, their targets and the nature and number of drug targets. Nat. Rev. Drug Discov.5(10),821–834 (2006).
  • Hamill OP, Marty A, Neher E, Sakmann B, Sigworth FJ. Improved patch-clamp techniques for high-resolution current recording from cells and cell-free membrane patches. Pflugers Arch.391(2),85–100 (1981).
  • Bouevitch O, Lewis A, Pinevsky I, Wuskell JP, Loew LM. Probing membrane potential with nonlinear optics. Biophys. J.65(2),672–679 (1993).
  • 10  Knisley SB, Blitchington TF, Hill BC et al. Optical measurements of transmembrane potential changes during electric field stimulation of ventricular cells. Circ. Res.72(2),255–270 (1993).
  • 11  Epps DE, Wolfe ML, Groppi V. Characterization of the steady-state and dynamic fluorescence properties of the potential-sensitive dye bis-(1,3-dibutylbarbituric acid)trimethine oxonol (Dibac4(3)) in model systems and cells. Chem. Phys. Lipids69(2),137–150 (1994).▪▪ Description of DiBAC4(3) as voltage sensor.
  • 12  Schroeder KS, Neagle BD. FLiPR: a new instrument for accurate high-throughput optical screening. J. Biomol. Screen.1,75–80 (1996).▪▪ First paper describing the FLiPR platform for ion channel screening.
  • 13  Holevinsky KO, Fan Z, Frame M, Makielski JC, Groppi V, Nelson DJ. ATP-sensitive K+ channel opener acts as a potent Cl- channel inhibitor in vascular smooth muscle cells. J. Membr. Biol.137(1),59–70 (1994).▪▪ First application of DiBAC4(3) for ion channel screening.
  • 14  Dorn A, Hermann F, Ebneth A et al. Evaluation of a high-throughput fluorescence assay method for HERG potassium channel inhibition. J. Biomol. Screen.10(4),339–347 (2005).
  • 15  Gopalakrishnan M, Molinari EJ, Shieh CC et al. Pharmacology of human sulphonylurea receptor SUR1 and inward rectifier K+ channel Kir6.2 combination expressed in HEK-293 cells. Br. J. Pharmacol.129(7),1323–1332 (2000).
  • 16  Terstappen GC, Pellacani A, Aldegheri L et al. The antidepressant fluoxetine blocks the human small conductance calcium-activated potassium channels SK1, SK2 and SK3. Neurosci. Lett.346(1–2),85–88 (2003).
  • 17  Whiteaker KL, Davis-Taber R, Scott VE, Gopalakrishnan M. Fluorescence-based functional assay for sarcolemmal ATP-sensitive potassium channel activation in cultured neonatal rat ventricular myocytes. J. Pharmacol. Toxicol. Methods46(1),45–50 (2001).
  • 18  Whiteaker KL, Gopalakrishnan SM, Groebe D et al. Validation of FLiPR membrane potential dye for high throughput screening of potassium channel modulators. J. Biomol. Screen.6(5),305–312 (2001).
  • 19  Vasilyev DV, Shan QJ, Lee YT et al. A novel high-throughput screening assay for HCN channel blocker using membrane potential-sensitive dye and FLiPR. J. Biomol. Screen.14(9),1119–1128 (2009).
  • 20  Benjamin ER, Pruthi F, Olanrewaju S et al. State-dependent compound inhibition of Nav1.2 sodium channels using the FLiPR Vm dye: on-target and off-target effects of diverse pharmacological agents. J. Biomol. Screen.11(1),29–39 (2006).
  • 21  Joesch C, Guevarra E, Parel SP et al. Use of FLiPR membrane potential dyes for validation of high-throughput screening with the FLiPR and microARCS technologies: identification of ion channel modulators acting on the GABAA receptor. J. Biomol. Screen.13(3),218–228 (2008).
  • 22  Slack M, Kirchhoff C, Moller C, Winkler D, Netzer R. Identification of novel Kv1.3 blockers using a fluorescent cell-based ion channel assay. J. Biomol. Screen.11(1),57–64 (2006).
  • 23  Gonzalez JE, Tsien RY. Voltage sensing by fluorescence resonance energy transfer in single cells. Biophys. J.69(4),1272–1280 (1995).▪▪ Introduction of fluorescence resonance energy transfer (FRET) for voltage sensing.
  • 24  Gonzalez JE, Tsien RY. Improved indicators of cell membrane potential that use fluorescence resonance energy transfer. Chem. Biol.4(4),269–277 (1997).
  • 25  Falconer M, Smith F, Surah-Narwal S et al. High-throughput screening for ion channel modulators. J. Biomol. Screen.7(5),460–465 (2002).▪▪ First paper describing the ViPR® platform for FRET assays.
  • 26  Felix JP, Williams BS, Priest BT et al. Functional assay of voltage-gated sodium channels using membrane potential-sensitive dyes. Assay Drug Dev. Technol.2(3),260–268 (2004).
  • 27  Liu CJ, Priest BT, Bugianesi RM et al. A high-capacity membrane potential FRET-based assay for NaV1.8 channels. Assay Drug Dev. Technol.4(1),37–48 (2006).
  • 28  Solly K, Cassaday J, Felix JP et al. Miniaturization and HTS of a FRET-based membrane potential assay for Kir channel inhibitors. Assay Drug Dev. Technol.6(2),225–234 (2008).
  • 29  Willow M, Kuenzel EA, Catterall WA. Inhibition of voltage-sensitive sodium channels in neuroblastoma cells and synaptosomes by the anticonvulsant drugs diphenylhydantoin and carbamazepine. Mol. Pharmacol.25(2),228–234 (1984).
  • 30  Rasmussen CA Jr, Sutko JL, Barry WH. Effects of ryanodine and caffeine on contractility, membrane voltage, and calcium exchange in cultured heart cells. Circ. Res.60(4),495–504 (1987).
  • 31  Rasmussen H, Takuwa Y, Park S. Protein kinase C in the regulation of smooth muscle contraction. FASEB J.1(3),177–185 (1987).
  • 32  Weir SW, Weston AH. The effects of BRL 34915 and nicorandil on electrical and mechanical activity and on 86Rb efflux in rat blood vessels. Br. J. Pharmacol.88(1),121–128 (1986).
  • 33  Terstappen GC. Functional analysis of native and recombinant ion channels using a high-capacity nonradioactive rubidium efflux assay. Anal. Biochem.272(2),149–155 (1999).▪▪ Introduction of atomic absorption spectroscopy for ion channel screening.
  • 34  Terstappen GC. Nonradioactive rubidium ion efflux assay and its applications in drug discovery and development. Assay Drug Dev. Technol.2(5),553–559 (2004).
  • 35  Terstappen GC. Nonradioactive rubidium efflux assay technology for screening of ion channels. In: Label-Free Assay Technologies for Drug Discovery. Cooper M, Mayer L (Eds). Wiley-VCH, Weinheim, Germany (2010) (In Press).
  • 36  Rezazadeh S, Hesketh JC, Fedida D. Rb+ flux through hERG channels affects the potency of channel blocking drugs: correlation with data obtained using a high-throughput Rb+ efflux assay. J. Biomol. Screen.9(7),588–597 (2004).
  • 37  Tang W, Kang J, Wu X et al. Development and evaluation of high throughput functional assay methods for HERG potassium channel. J. Biomol. Screen.6(5),325–331 (2001).
  • 38  Chaudhary KW, O’Neal JM, Mo ZL, Fermini B, Gallavan RH, Bahinski A. Evaluation of the rubidium efflux assay for preclinical identification of HERG blockade. Assay Drug Dev. Technol.4(1),73–82 (2006).
  • 39  Trivedi S, Dekermendjian K, Julien R et al. Cellular HTS assays for pharmacological characterization of Na(V)1.7 modulators. Assay Drug Dev. Technol.6(2),167–179 (2008).
  • 40  Gill S, Gill R, Xie Y, Wicks D, Liang D. Development and validation of HTS flux assay for endogenously expressed chloride channels in a CHO-K1 cell line. Assay Drug Dev. Technol.4(1),65–71 (2006).
  • 41  Kasner SE, Ganz MB. Regulation of intracellular potassium in mesangial cells: a fluorescence analysis using the dye, PBFI. Am. J. Physiol.262(3 Pt 2),F462–467 (1992).
  • 42  Minta A, Tsien RY. Fluorescent indicators for cytosolic sodium. J. Biol. Chem.264(32),19449–19457 (1989).
  • 43  Verkman AS, Takla R, Sefton B, Basbaum C, Widdicombe JH. Quantitative fluorescence measurement of chloride transport mechanisms in phospholipid vesicles. Biochemistry28(10),4240–4244 (1989).
  • 44  Grynkiewicz G, Poenie M, Tsien RY. A new generation of Ca2+ indicators with greatly improved fluorescence properties. J. Biol. Chem.260(6),3440–3450 (1985).▪▪ Description of Fura-2 as prototype of a new family of Ca2+ indicators.
  • 45  Mattheakis LC, Savchenko A. Assay technologies for screening ion channel targets. Curr. Opin. Drug Discov. Devel.4(1),124–134 (2001).
  • 46  Song Y, Buelow B, Perraud AL, Scharenberg AM. Development and validation of a cell-based high-throughput screening assay for TRPM2 channel modulators. J. Biomol. Screen.13(1),54–61 (2008).
  • 47  Dai G, Haedo RJ, Warren VA et al. A high-throughput assay for evaluating state dependence and subtype selectivity of Cav2 calcium channel inhibitors. Assay Drug Dev. Technol.6(2),195–212 (2008).
  • 48  Weaver CD, Harden D, Dworetzky SI, Robertson B, Knox RJ. A thallium-sensitive, fluorescence-based assay for detecting and characterizing potassium channel modulators in mammalian cells. J. Biomol. Screen.9(8),671–677 (2004).
  • 49  Titus SA, Beacham D, Shahane SA et al. A new homogeneous high-throughput screening assay for profiling compound activity on the human ether-a-go-go-related gene channel. Anal. Biochem.394(1),30–38 (2009).
  • 50  Kiss L, Bennett PB, Uebele VN et al. High throughput ion-channel pharmacology: planar-array-based voltage clamp. Assay Drug Dev. Technol.1(1,2),127–135 (2003).
  • 51  Schroeder K, Neagle B, Trezise DJ, Worley J. Ionworks HT: a new high-throughput electrophysiology measurement platform. J. Biomol. Screen.8(1),50–64 (2003).▪▪ First paper describing the IonWorks platform.
  • 52  Finkel A, Wittel A, Yang N, Handran S, Hughes J, Costantin J. Population patch clamp improves data consistency and success rates in the measurement of ionic currents. J. Biomol. Screen.11(5),488–496 (2006).▪▪ First paper describing population patch clamp and IonWorks Quattro platforms.
  • 53  Dunlop J, Bowlby M, Peri R, Vasilyev D, Arias R. High-throughput electrophysiology: an emerging paradigm for ion-channel screening and physiology. Nat. Rev. Drug Discov.7(4),358–368 (2008).▪▪ Describes recent advances in automated high-throughput electrophysiological instrumentation.
  • 54  Lu Q, An WF. Impact of novel screening technologies on ion channel drug discovery. Comb. Chem. High Throughput Screen.11(3),185–194 (2008).
  • 55  Bridgland-Taylor MH, Hargreaves AC, Easter A et al. Optimisation and validation of a medium-throughput electrophysiology-based hERG assay using IonWorks HT. J. Pharmacol. Toxicol. Methods54(2),189–199 (2006).
  • 56  Sorota S, Zhang XS, Margulis M, Tucker K, Priestley T. Characterization of a hERG screen using the IonWorks HT: comparison to a hERG rubidium efflux screen. Assay Drug Dev. Technol.3(1),47–57 (2005).
  • 57  John VH, Dale TJ, Hollands EC et al. Novel 384-well population patch clamp electrophysiology assays for Ca2+-activated K+ channels. J. Biomol. Screen.12(1),50–60 (2007).
  • 58  Jow F, Shen R, Chanda P et al. Validation of a medium-throughput electrophysiological assay for KCNQ2/3 channel enhancers using IonWorks HT. J. Biomol. Screen.12(8),1059–1067 (2007).
  • 59  Xie X, Van Deusen AL, Vitko I et al. Validation of high throughput screening assays against three subtypes of Cav3 T-type channels using molecular and pharmacologic approaches. Assay Drug Dev. Technol.5(2),191–203 (2007).
  • 60  Lee YT, Vasilyev DV, Shan QJ, Dunlop J, Mayer S, Bowlby MR. Novel pharmacological activity of loperamide and CP-339,818 on human HCN channels characterized with an automated electrophysiology assay. Eur. J. Pharmacol.581(1–2),97–104 (2008).
  • 61  Harmer AR, Abi-Gerges N, Easter A et al. Optimisation and validation of a medium-throughput electrophysiology-based hNav1.5 assay using IonWorks. J. Pharmacol. Toxicol. Methods57(1),30–41 (2008).
  • 62  Castle N, Printzenhoff D, Zellmer S, Antonio B, Wickenden A, Silvia C. Sodium channel inhibitor drug discovery using automated high throughput electrophysiology platforms. Comb. Chem. High Throughput Screen.12(1),107–122 (2009).
  • 63  Cox JJ, Reimann F, Nicholas AK et al. An SCN9A channelopathy causes congenital inability to experience pain. Nature444(7121),894–898 (2006).
  • 64  Goldberg YP, MacFarlane J, MacDonald ML et al. Loss-of-function mutations in the Nav1.7 gene underlie congenital indifference to pain in multiple human populations. Clin. Genet.71(4),311–319 (2007).
  • 65  Dunlop J, Bowlby M, Peri R et al. Ion channel screening. Comb. Chem. High Throughput Screen.11(7),514–522 (2008).
  • 66  Clare JJ, Chen MX, Downie DL, Trezise DJ, Powell AJ. Use of planar array electrophysiology for the development of robust ion channel cell lines. Comb. Chem. High Throughput Screen.12(1),96–106 (2009).
  • 67  Dubin AE, Nasser N, Rohrbacher J et al. Identifying modulators of hERG channel activity using the PatchXpress planar patch clamp. J. Biomol. Screen.10(2),168–181 (2005).
  • 68  Ghetti A, Guia A, Xu J. Automated voltage-clamp technique. Methods Mol. Biol.403,59–69 (2007).
  • 69  Asmild M, Oswald N, Krzywkowski KM et al. Upscaling and automation of electrophysiology: toward high throughput screening in ion channel drug discovery. Receptors Channels9(1),49–58 (2003).
  • 70  Beck EJ, Hutchinson TL, Qin N, Flores CM, Liu Y. Development and validation of a secondary screening assay for TRPM8 antagonists using QPatch HT. Assay Drug Dev. Technol.
  • 71  Mathes C, Friis S, Finley M, Liu Y. QPatch: the missing link between HTS and ion channel drug discovery. Comb. Chem. High Throughput Screen.12(1),78–95 (2009).
  • 72  McPate M, Lilley S, Gosling M, Friis S, Jacobsen RB, Tranter P. Evaluation of the QPatch HT and HTX systems as methods for ion channel screening. Biophys. J.98(3, Suppl. 1),340a (2010).
  • 73  Farre C, Stoelzle S, Haarmann C, George M, Bruggemann A, Fertig N. Automated ion channel screening: patch clamping made easy. Expert Opin. Ther. Targets11(4),557–565 (2007).
  • 74  Brueggemann A, George M, Klau M et al. Ion channel drug discovery and research: the automated nano-patch-clamp technology. Curr. Drug Discov. Technol.1(1),91–96 (2004).
  • 75  Bruggemann A, George M, Klau M et al. High quality ion channel analysis on a chip with the NPC technology. Assay Drug Dev. Technol.1(5),665–673 (2003).
  • 76  Farre C, Haythornthwaite A, Haarmann C et al. Port-a-patch and patchliner: high fidelity electrophysiology for secondary screening and safety pharmacology. Comb. Chem. High Throughput Screen.12(1),24–37 (2009).
  • 77  Balasubramanian B, Imredy JP, Kim D, Penniman J, Lagrutta A, Salata JJ. Optimization of Cav1.2 screening with an automated planar patch clamp platform. J. Pharmacol. Toxicol. Methods59(2),62–72 (2009).
  • 78  Guo L, Guthrie H. Automated electrophysiology in the preclinical evaluation of drugs for potential QT prolongation. J. Pharmacol. Toxicol. Methods52(1),123–135 (2005).
  • 79  Korsgaard MP, Strobaek D, Christophersen P. Automated planar electrode electrophysiology in drug discovery: examples of the use of QPatch in basic characterization and high content screening on Nav, K(Ca)2.3, and Kv11.1 channels. Comb. Chem. High Throughput Screen12(1),51–63 (2009).
  • 80  Kutchinsky J, Friis S, Asmild M et al. Characterization of potassium channel modulators with QPatch automated patch-clamp technology: system characteristics and performance. Assay Drug Dev. Technol.1(5),685–693 (2003).
  • 81  Tao H, Santa Ana D, Guia A et al. Automated tight seal electrophysiology for assessing the potential hERG liability of pharmaceutical compounds. Assay Drug Dev. Technol.2(5),497–506 (2004).
  • 82  Mo ZL, Faxel T, Yang YS, Gallavan R, Messing D, Bahinski A. Effect of compound plate composition on measurement of hERG current IC(50) using PatchXpress. J. Pharmacol. Toxicol. Methods60(1),39–44 (2009).
  • 83  Ratliff KS, Petrov A, Eiermann GJ et al. An automated electrophysiology serum shift assay for K(V) channels. Assay Drug Dev. Technol.6(2),243–253 (2008).
  • 84  Jones KA, Garbati N, Zhang H, Large CH. Automated patch clamping using the QPatch. Methods Mol. Biol.565,209–223 (2009).
  • 85  Schroder RL, Friis S, Sunesen M, Mathes C, Willumsen NJ. Automated patch-clamp technique: increased throughput in functional characterization and in pharmacological screening of small-conductance Ca2+ release-activated Ca2+ channels. J. Biomol. Screen.13(7),638–647 (2008).
  • 86  Dunlop J, Roncarati R, Jow B et al.In vitro screening strategies for nicotinic receptor ligands. Biochem. Pharmacol.74(8),1172–1181 (2007).
  • 87  Friis S, Mathes C, Sunesen M, Bowlby MR, Dunlop J. Characterization of compounds on nicotinic acetylcholine receptor α7 channels using higher throughput electrophysiology. J. Neurosci. Methods177(1),142–148 (2009).