761
Views
40
CrossRef citations to date
0
Altmetric
Review

An update on the advancing high-throughput screening techniques for patch clamp-based ion channel screens: implications for drug discovery

, , , , , , , , , , & show all
Pages 269-277 | Received 28 Sep 2017, Accepted 12 Jan 2018, Published online: 17 Jan 2018

References

  • Dunlop J, Bowlby M, Peri R, et al. High-throughput electrophysiology: an emerging paradigm for ion-channel screening and physiology. Nat Rev Drug Discov. 2008;7:358–368.
  • Bell DC, Dallas ML. Using automated patch clamp electrophysiology platforms in pain-related ion channel research: insights from industry and academia. Br J Pharmacol. 2017. Epub ahead of print. doi: 10.1111/bph.13916
  • Obergrussberger A, Stölzle-Feix S, Becker N, et al. Novel screening techniques for ion channel targeting drugs. Channels. 2015;9:367–375.
  • 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. 2009;12:24–37.
  • Stoelzle S, Obergrussberger A, Brüggemann A, et al. State-of-the-art automated patch clamp devices: heat activation, action potentials, and high throughput in ion channel screening. Front Pharmacol. 2011;2:76.
  • Haythornthwaite A, Stoelzle S, Hasler A, et al. Characterizing human ion channels in induced pluripotent stem cell-derived neurons. J Biomol Screen. 2012;17:1264–1272.
  • Milligan CJ, Li J, Sukumar P, et al. Robotic multiwell planar patch-clamp for native and primary mammalian cells. Nat Protoc. 2009;4:244–255.
  • Obergrussberger A, Haarmann C, Rinke I, et al. Automated patch clamp analysis of nAChα7 and NaV 1.7 channels. Curr Protoc Pharmacol. 2014;65:11.13.1–11.13.48.
  • Mathes C, Friis S, Finley M, et al. QPatch: the missing link between HTS and ion channel drug discovery. Comb Chem High Throughput Screen. 2009;12:78–95.
  • Friis S, Mathes C, Sunesen M, et al. Characterization of compounds on nicotinic acetylcholine receptor alpha7 channels using higher throughput electrophysiology. J Neurosci Methods. 2009;177:142–148.
  • Obergrussberger A, Brüggemann A, Goetze TA, et al. Automated patch clamp meets high-throughput screening: 384 cells recorded in parallel on a planar patch clamp module. J Lab Autom. 2016;21:779–793.
  • Li T, Lu G, Chiang EY, et al. High-throughput electrophysiological assays for voltage gated ion channels using SyncroPatch 768PE. PLoS One. 2017;12:e0180154.
  • Potet F, Vanoye CG, George AL. Use-dependent block of human cardiac sodium channels by GS967. Mol Pharmacol. 2016;90:52–60.
  • Sauter DRP, Sørensen CE, Rapedius M, et al. pH-sensitive K+ channel TREK-1 is a novel target in pancreatic cancer. Biochim Biophys Acta. 2016;1862:1994–2003.
  • Haraguchi Y, Ohtsuki A, Oka T, et al. Electrophysiological analysis of mammalian cells expressing hERG using automated 384-well-patch-clamp. BMC Pharmacol Toxicol. 2015;16:39.
  • Calhoun JD, Vanoye CG, Kok F, et al. Characterization of a KCNB1 variant associated with autism, intellectual disability, and epilepsy. Neurol Genet. 2017;3:e198.
  • Chambers C, Witton I, Adams C, et al. High-throughput screening of NaV1.7 modulators using a giga-seal automated patch clamp instrument. Assay Drug Dev Technol. 2016;14:93–108.
  • Comley J. Automated patch clamping trends [Internet]. Drug Discov. From Technol. Networks. 2016; Available from: https://www.technologynetworks.com/drug-discovery/articles/automated-patch-clamping-trends-184254 [Last accessed 17 January 2018].
  • Select Biosciences Market Reports: Automated Patch Clamping Trends 2015. Published by HTSTec Ltd. 2015. Available at: http://selectbiosciences.com/MarketReportsID.aspx?reportID=93. [Last accessed 17 January 2018].
  • Golden AP, Li N, Chen Q, et al. IonFlux: a microfluidic patch clamp system evaluated with human Ether-à-go-go related gene channel physiology and pharmacology. Assay Drug Dev Technol. 2011;9:608–619.
  • Kauthale RR, Dadarkar SS, Husain R, et al. Assessment of temperature-induced hERG channel blockade variation by drugs. J Appl Toxicol. 2015;35:799–805.
  • Milligan CJ, Möller C. Automated planar patch-clamp. In: Gamper N, editor. Ion channels: methods and protocols (methods in molecular biology). Springer Science+Business Media, LLC. 2013. p. 171–187.
  • Sitzia F, Brown JT, Randall AD, et al. Voltage- and temperature-dependent allosteric modulation of α7 nicotinic receptors by PNU120596. Front Pharmacol. 2011;2:81.
  • Papakosta M, Dalle C, Haythornthwaite A, et al. The chimeric approach reveals that differences in the TRPV1 pore domain determine species-specific sensitivity to block of heat activation. J Biol Chem. 2011;286:39663–39672.
  • Garami A, Shimansky YP, Pakai E, et al. Contributions of different modes of TRPV1 activation to TRPV1 antagonist-induced hyperthermia. J Neurosci. 2010;30:1435–1440.
  • Zhou Z, Gong Q, Ye B, et al. Properties of HERG channels stably expressed in HEK 293 cells studied at physiological temperature. Biophys J. 1998;74:230–241.
  • Brüggemann A, Stoelzle S, George M, et al. Microchip technology for automated and parallel patch-clamp recording. Small. 2006;2:840–846.
  • Farre C, Stoelzle S, Haarmann C, et al. Automated ion channel screening: patch clamping made easy. Expert Opin Ther Targets. 2007;11:557–565.
  • Brinkwirth N, Friis S, Goetze T, et al. Investigation of the ion channels TMEM16A and TRPC5 and their modulation by intracellular calcium. Biophys J. 2017;112:413a.
  • Sager PT, Gintant G, Turner JR, et al. Rechanneling the cardiac proarrhythmia safety paradigm: a meeting report from the Cardiac Safety Research Consortium. Am Heart J. 2014;167:292–300.
  • Cavero I, Holzgrefe H. Comprehensive in vitro Proarrhythmia Assay, a novel in vitro/in silico paradigm to detect ventricular proarrhythmic liability: a visionary 21st century initiative. Expert Opin Drug Safe. 2014;13:745–758.
  • Gintant G, Sager PT, Stockbridge N. Evolution of strategies to improve preclinical cardiac safety testing. Nat Rev Drug Discov. 2016;7:457-471.
  • Rajamohan D, Kalra S, Hoang MD, et al. Automated electrophysiological and pharmacological evaluation of human pluripotent stem cell-derived cardiomyocytes. Stem Cells Dev. 2016;25:439–452.
  • Jonsson MKB, Vos MA, Mirams GR, et al. Application of human stem cell-derived cardiomyocytes in safety pharmacology requires caution beyond hERG. J Mol Cell Cardiol. 2012;52:998–1008.
  • Peters F, Gennerich A, Czesnik D, et al. Low frequency voltage clamp: recording of voltage transients at constant average command voltage. J Neurosci Methods. 2000;99:129–135.
  • Nerbonne J, Kass R. Molecular physiology of cardiac repolarization. Physiol Rev. 2005;85:1205–1253.
  • Zhang S, Zhou Z, Gong Q, et al. Mechanism of block and identification of the verapamil binding domain to HERG potassium channels. Circ Res. 1999;84:989–998.
  • Doss MX, Di Diego JM, Goodrow RJ, et al. Maximum diastolic potential of human induced pluripotent stem cell-derived cardiomyocytes depends critically on IKr. PLoS One. 2012;7:e40288.
  • Vaidyanathan R, Markandeya YS, Kamp TJ, et al. I K1 -enhanced human-induced pluripotent stem cell-derived cardiomyocytes: an improved cardiomyocyte model to investigate inherited arrhythmia syndromes. Am J Physiol Hear Circ Physiol. 2016;310:H1611–H1621.
  • Wilders R. Dynamic clamp: a powerful tool in cardiac electrophysiology. J Physiol. 2006;576:349–359.
  • Bett GC, Kaplan AD, Lis A, et al. Electronic “expression” of the inward rectifier in cardiocytes derived from human induced pluripotent stem cells. Hear Rhythm. 2013;10:1903–1910.
  • Meijer van Putten RM, Mengarelli I, Guan K, et al. Ion channelopathies in human induced pluripotent stem cell derived cardiomyocytes: a dynamic clamp study with virtual IK1. Front Physiol. 2015;6:1–17.
  • Goversen B, Becker N, Stoelzle-Feix S, et al. Hybrid models for safety pharmacology on an automated patch clamp platform: joining iPSC-derived cardiomyocytes and simulations of IK1 ion channels in real-time. Front Pharmacol. 2018. Provisionally accepted article: doi: 10.3389/fphys.2017.01094.
  • Zhang F, Wang L-P, Boyden ES, et al. Channelrhodopsin-2 and optical control of excitable cells. Nat Methods. 2006;3:785–792.
  • Xpress4U - Getting the ideal transfection method. NCardia, Germany, Belgium, Netherlands, USA. 2017. Available at: http://ncardia.com/news-events-innovations/xpress.4u.html. [Last accessed 17 January 2018].
  • Polosukhina A, Litt J, Tochitsky I, et al. Photochemical restoration of visual responses in blind mice. Neuron. 2012;75:271–282.
  • Tochitsky I, Polosukhina A, Degtyar VE, et al. Restoring visual function to blind mice with a photoswitch that exploits electrophysiological remodeling of retinal ganglion cells. Neuron. 2014;81:800–813.
  • Laprell L, Tochitsky I, Kaur K, et al. Photopharmacological control of bipolar cells restores visual function in blind mice. J Clin Invest. 2017;127:2598–2611.
  • Frank JA, Moroni M, Moshourab R, et al. Photoswitchable fatty acids enable optical control of TRPV1. Nat Commun. 2015;6:7118.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.