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

ABSTRACT Introduction: Automated patch clamp (APC) devices have become commonplace in many industrial and academic labs. Their ease-of-use and flexibility have ensured that users can perform routine screening experiments and complex kinetic experiments on the same device without the need for months of training and experience. APC devices are being developed to increase throughput and flexibility. Areas covered: Experimental options such as temperature control, internal solution exchange and current clamp have been available on some APC devices for some time, and are being introduced on other devices. A comprehensive review of the literature pertaining to these features for the Patchliner, QPatch and Qube and data for these features for the SyncroPatch 384/768PE, is given. In addition, novel features such as dynamic clamp on the Patchliner and light stimulation of action potentials using channelrhodosin-2 is discussed. Expert opinion: APC devices will continue to play an important role in drug discovery. The instruments will be continually developed to meet the needs of HTS laboratories and for basic research. The use of stem cells and recordings in current clamp mode will increase, as will the development of complex add-ons such as dynamic clamp and optical stimulation on high throughput devices.

[1]  Icilio Cavero,et al.  Comprehensive in vitro Proarrhythmia Assay, a novel in vitro/in silico paradigm to detect ventricular proarrhythmic liability: a visionary 21st century initiative , 2014, Expert opinion on drug safety.

[2]  Tianbo Li,et al.  High-throughput electrophysiological assays for voltage gated ion channels using SyncroPatch 768PE , 2017, PloS one.

[3]  Andrea Brüggemann,et al.  pH-sensitive K(+) channel TREK-1 is a novel target in pancreatic cancer. , 2016, Biochimica et biophysica acta.

[4]  Y. Shimansky,et al.  Contributions of Different Modes of TRPV1 Activation to TRPV1 Antagonist-Induced Hyperthermia , 2010, The Journal of Neuroscience.

[5]  Gary R. Mirams,et al.  Application of human stem cell-derived cardiomyocytes in safety pharmacology requires caution beyond hERG. , 2012, Journal of molecular and cellular cardiology.

[6]  Carlos G. Vanoye,et al.  Characterization of a KCNB1 variant associated with autism, intellectual disability, and epilepsy , 2017, Neurology: Genetics.

[7]  C. Grimm,et al.  The Chimeric Approach Reveals That Differences in the TRPV1 Pore Domain Determine Species-specific Sensitivity to Block of Heat Activation* , 2011, The Journal of Biological Chemistry.

[8]  N. Fertig,et al.  Automated Patch Clamp Analysis of nAChα7 and NaV1.7 Channels , 2014, Current protocols in pharmacology.

[9]  Clemens Möller,et al.  Automated planar patch-clamp. , 2013, Methods in molecular biology.

[10]  Ralf Kettenhofen,et al.  State-of-the-Art Automated Patch Clamp Devices: Heat Activation, Action Potentials, and High Throughput in Ion Channel Screening , 2011, Front. Pharmacol..

[11]  Sonja Stoelzle,et al.  Microchip technology for automated and parallel patch-clamp recording. , 2006, Small.

[12]  N. Fertig,et al.  Investigation of the Ion Channels TMEM16A and TRPC5 and their Modulation by Intracellular Calcium , 2017 .

[13]  D. Schild,et al.  Low frequency voltage clamp: recording of voltage transients at constant average command voltage , 2000, Journal of Neuroscience Methods.

[14]  G. Gintant,et al.  Evolution of strategies to improve preclinical cardiac safety testing , 2016, Nature Reviews Drug Discovery.

[15]  Andrea Brüggemann,et al.  Novel screening techniques for ion channel targeting drugs , 2015, Channels.

[16]  C. January,et al.  Properties of HERG channels stably expressed in HEK 293 cells studied at physiological temperature. , 1998, Biophysical journal.

[17]  Michael George,et al.  Automated Patch Clamp Meets High-Throughput Screening , 2016, Journal of laboratory automation.

[18]  Rahul R Kauthale,et al.  Assessment of temperature‐induced hERG channel blockade variation by drugs , 2017, Journal of applied toxicology : JAT.

[19]  A. George,et al.  Use-Dependent Block of Human Cardiac Sodium Channels by GS967 , 2016, Molecular Pharmacology.

[20]  Ronald Wilders,et al.  Dynamic clamp: a powerful tool in cardiac electrophysiology , 2006, The Journal of physiology.

[21]  Damian C Bell,et al.  Using automated patch clamp electrophysiology platforms in pain‐related ion channel research: insights from industry and academia , 2018, British journal of pharmacology.

[22]  C. January,et al.  IK1-enhanced human-induced pluripotent stem cell-derived cardiomyocytes: an improved cardiomyocyte model to investigate inherited arrhythmia syndromes. , 2016, American journal of physiology. Heart and circulatory physiology.

[23]  Michael George,et al.  Characterizing Human Ion Channels in Induced Pluripotent Stem Cell–Derived Neurons , 2012, Journal of biomolecular screening.

[24]  Jon T. Brown,et al.  Voltage- and Temperature-Dependent Allosteric Modulation of α7 Nicotinic Receptors by PNU120596 , 2011, Front. Pharmacol..

[25]  Qinlian Zhou,et al.  Electronic "expression" of the inward rectifier in cardiocytes derived from human-induced pluripotent stem cells. , 2013, Heart rhythm.

[26]  Yuji Haraguchi,et al.  Electrophysiological analysis of mammalian cells expressing hERG using automated 384-well-patch-clamp , 2015, BMC Pharmacology and Toxicology.

[27]  Chris Chambers,et al.  High-Throughput Screening of Na(V)1.7 Modulators Using a Giga-Seal Automated Patch Clamp Instrument. , 2016, Assay and drug development technologies.

[28]  Michael George,et al.  Automated ion channel screening: patch clamping made easy , 2007, Expert opinion on therapeutic targets.

[29]  Michael Xavier Doss,et al.  Maximum Diastolic Potential of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes Depends Critically on IKr , 2012, PloS one.

[30]  Cristian Ionescu-Zanetti,et al.  IonFlux: a microfluidic patch clamp system evaluated with human Ether-à-go-go related gene channel physiology and pharmacology. , 2011, Assay and drug development technologies.

[31]  Gary Gintant,et al.  Rechanneling the cardiac proarrhythmia safety paradigm: a meeting report from the Cardiac Safety Research Consortium. , 2014, American heart journal.

[32]  Feng Zhang,et al.  Channelrhodopsin-2 and optical control of excitable cells , 2006, Nature Methods.

[33]  Dirk Trauner,et al.  Restoring Visual Function to Blind Mice with a Photoswitch that Exploits Electrophysiological Remodeling of Retinal Ganglion Cells , 2014, Neuron.

[34]  Martin Biel,et al.  Photopharmacological control of bipolar cells restores visual function in blind mice , 2017, The Journal of clinical investigation.

[35]  J. Nerbonne,et al.  Molecular physiology of cardiac repolarization. , 2005, Physiological reviews.

[36]  C. January,et al.  Mechanism of block and identification of the verapamil binding domain to HERG potassium channels. , 1999, Circulation research.

[37]  D. Trauner,et al.  Photoswitchable fatty acids enable optical control of TRPV1 , 2015, Nature Communications.

[38]  Dirk Trauner,et al.  Photochemical Restoration of Visual Responses in Blind Mice , 2012, Neuron.

[39]  Mark L Dallas,et al.  Robotic multiwell planar patch-clamp for native and primary mammalian cells , 2009, Nature Protocols.

[40]  A. V. van Ginneken,et al.  Ion channelopathies in human induced pluripotent stem cell derived cardiomyocytes: a dynamic clamp study with virtual IK1 , 2015, Front. Physiol..

[41]  R. Peri,et al.  High-throughput electrophysiology: an emerging paradigm for ion-channel screening and physiology , 2008, Nature Reviews Drug Discovery.

[42]  Yi Liu,et al.  QPatch: the missing link between HTS and ion channel drug discovery. , 2009, Combinatorial chemistry & high throughput screening.

[43]  S. Friis,et al.  Characterization of compounds on nicotinic acetylcholine receptor alpha7 channels using higher throughput electrophysiology , 2009, Journal of Neuroscience Methods.

[44]  Michael George,et al.  Port-a-patch and patchliner: high fidelity electrophysiology for secondary screening and safety pharmacology. , 2009, Combinatorial chemistry & high throughput screening.

[45]  Xuebin B. Yang,et al.  Automated Electrophysiological and Pharmacological Evaluation of Human Pluripotent Stem Cell-Derived Cardiomyocytes , 2016, Stem cells and development.

[46]  Nadine Becker,et al.  A Hybrid Model for Safety Pharmacology on an Automated Patch Clamp Platform: Using Dynamic Clamp to Join iPSC-Derived Cardiomyocytes and Simulations of Ik1 Ion Channels in Real-Time , 2018, Front. Physiol..