1
|
Thoma G, Miltz W, Srinivas H, Penno CA, Kiffe M, Gajewska M, Klein K, Evans A, Beerli C, Röhn TA. Structure-Guided Elaboration of a Fragment-Like Hit into an Orally Efficacious Leukotriene A4 Hydrolase Inhibitor. J Med Chem 2024; 67:5093-5108. [PMID: 38476002 DOI: 10.1021/acs.jmedchem.4c00290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
Leukotriene A4 hydrolase (LTA4H) is the final and rate-limiting enzyme in the biosynthesis of pro-inflammatory leukotriene B4 (LTB4). Preclinical studies have provided strong evidence that LTA4H is an attractive drug target for the treatment of chronic inflammatory diseases. Here, we describe the transformation of compound 2, a fragment-like hit, into the potent inhibitor of LTA4H 3. Our strategy involved two key steps. First, we aimed to increase the polarity of fragment 2 to improve its drug-likeness, particularly its solubility, while preserving both its promising potency and low molecular weight. Second, we utilized structural information and incorporated a basic amino function, which allowed for the formation of an essential hydrogen bond with Q136 of LTA4H and consequently enhanced the potency. Compound 3 exhibited exceptional selectivity and showed oral efficacy in a KRN passive serum-induced arthritis model in mice. The anticipated human dose to achieve 90% target engagement at the trough concentration was determined to be 40 mg administered once daily.
Collapse
Affiliation(s)
- Gebhard Thoma
- Global Discovery Chemistry, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Wolfgang Miltz
- Global Discovery Chemistry, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Honnappa Srinivas
- Discovery Sciences, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Carlos A Penno
- Discovery Sciences, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Michael Kiffe
- PK Sciences, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Monika Gajewska
- PK Sciences, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Kai Klein
- PK Sciences, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Amanda Evans
- Immunology Disease Area, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Christian Beerli
- Immunology Disease Area, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Till A Röhn
- Immunology Disease Area, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| |
Collapse
|
2
|
Thoma G, Markert C, Lueoend R, Miltz W, Spanka C, Bollbuck B, Wolf RM, Srinivas H, Penno CA, Kiffe M, Gajewska M, Bednarczyk D, Wieczorek G, Evans A, Beerli C, Röhn TA. Discovery of Amino Alcohols as Highly Potent, Selective, and Orally Efficacious Inhibitors of Leukotriene A4 Hydrolase. J Med Chem 2023; 66:16410-16425. [PMID: 38015154 DOI: 10.1021/acs.jmedchem.3c01866] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
The discovery of chiral amino alcohols derived from our previously disclosed clinical LTA4H inhibitor LYS006 is described. In a biochemical assay, their optical antipodes showed similar potencies, which could be rationalized by the cocrystal structures of these compounds bound to LTA4H. Despite comparable stabilities in liver microsomes, they showed distinct in vivo PK properties. Selective O-phosphorylation of the (R)-enantiomers in blood led to clearance values above the hepatic blood flow, whereas the (S)-enantiomers were unaffected and exhibited satisfactory metabolic stabilities in vivo. Introduction of two pyrazole rings led to compound (S)-2 with a more balanced distribution of polarity across the molecule, exhibiting high selectivity and excellent potency in vitro and in vivo. Furthermore, compound (S)-2 showed favorable profiles in 16-week IND-enabling toxicology studies in dogs and rats. Based on allometric scaling and potency in whole blood, compound (S)-2 has the potential for a low oral efficacious dose administered once daily.
Collapse
Affiliation(s)
- Gebhard Thoma
- Global Discovery Chemistry, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Christian Markert
- Global Discovery Chemistry, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Rainer Lueoend
- Global Discovery Chemistry, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Wolfgang Miltz
- Global Discovery Chemistry, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Carsten Spanka
- Global Discovery Chemistry, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Birgit Bollbuck
- Global Discovery Chemistry, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Romain M Wolf
- Global Discovery Chemistry, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Honnappa Srinivas
- Chemical Biology & Therapeutics, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Carlos A Penno
- Chemical Biology & Therapeutics, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Michael Kiffe
- PK Sciences, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Monika Gajewska
- PK Sciences, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Dallas Bednarczyk
- Discovery & Translational Lab, Biomedical Research, Novartis Pharma AG, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Grazyna Wieczorek
- Immunology Disease Area, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Amanda Evans
- Immunology Disease Area, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Christian Beerli
- Immunology Disease Area, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Till A Röhn
- Immunology Disease Area, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| |
Collapse
|
3
|
Ma JG, Vandenberg JI, Ng CA. Development of automated patch clamp assays to overcome the burden of variants of uncertain significance in inheritable arrhythmia syndromes. Front Physiol 2023; 14:1294741. [PMID: 38089476 PMCID: PMC10712320 DOI: 10.3389/fphys.2023.1294741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/13/2023] [Indexed: 10/16/2024] Open
Abstract
Advances in next-generation sequencing have been exceptionally valuable for identifying variants in medically actionable genes. However, for most missense variants there is insufficient evidence to permit definitive classification of variants as benign or pathogenic. To overcome the deluge of Variants of Uncertain Significance, there is an urgent need for high throughput functional assays to assist with the classification of variants. Advances in parallel planar patch clamp technologies has enabled the development of automated high throughput platforms capable of increasing throughput 10- to 100-fold compared to manual patch clamp methods. Automated patch clamp electrophysiology is poised to revolutionize the field of functional genomics for inheritable cardiac ion channelopathies. In this review, we outline i) the evolution of patch clamping, ii) the development of high-throughput automated patch clamp assays to assess cardiac ion channel variants, iii) clinical application of these assays and iv) where the field is heading.
Collapse
Affiliation(s)
- Joanne G. Ma
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Jamie I. Vandenberg
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Chai-Ann Ng
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
4
|
Jahanfar F, Sadofsky L, Morice A, D’Amico M. Nebivolol as a Potent TRPM8 Channel Blocker: A Drug-Screening Approach through Automated Patch Clamping and Ligand-Based Virtual Screening. MEMBRANES 2022; 12:954. [PMID: 36295712 PMCID: PMC9609861 DOI: 10.3390/membranes12100954] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 09/19/2022] [Accepted: 09/24/2022] [Indexed: 06/16/2023]
Abstract
Transient Receptor Potential Melastatin 8 (TRPM8) from the melastatin TRP channel subfamily is a non-selective Ca2+-permeable ion channel with multimodal gating which can be activated by low temperatures and cooling compounds, such as menthol and icilin. Different conditions such as neuropathic pain, cancer, overactive bladder syndrome, migraine, and chronic cough have been linked to the TRPM8 mode of action. Despite the several potent natural and synthetic inhibitors of TRPM8 that have been identified, none of them have been approved for clinical use. The aim of this study was to discover novel blocking TRPM8 agents using automated patch clamp electrophysiology combined with a ligand-based virtual screening based on the SwissSimilarity platform. Among the compounds we have tested, nebivolol and carvedilol exhibited the greatest inhibitory effect, with an IC50 of 0.97 ± 0.15 µM and 9.1 ± 0.6 µM, respectively. This study therefore provides possible candidates for future drug repurposing and suggests promising lead compounds for further optimization as inhibitors of the TRPM8 ion channel.
Collapse
Affiliation(s)
- Farhad Jahanfar
- Di.V.A.L. Toscana S.r.l., Via Madonna del Piano 6, 50019 Sesto Fiorentino, Italy
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Laura Sadofsky
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull HU6 7RX, UK
| | - Alyn Morice
- Respiratory Research Group, Hull York Medical School, Castle Hill Hospital, Cottingham HU16 5JQ, UK
| | - Massimo D’Amico
- Di.V.A.L. Toscana S.r.l., Via Madonna del Piano 6, 50019 Sesto Fiorentino, Italy
| |
Collapse
|
5
|
Translating the measurement of hERG kinetics and drug block for CiPA to a high throughput platform. J Pharmacol Toxicol Methods 2022; 117:107192. [PMID: 35750310 DOI: 10.1016/j.vascn.2022.107192] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/13/2022] [Accepted: 06/16/2022] [Indexed: 11/23/2022]
Abstract
The Comprehensive in vitro Proarrhythmic Assay (CiPA) has promoted use of in silico models of drug effects on cardiac repolarization to improve proarrhythmic risk prediction. These models contain a pharmacodynamic component describing drug binding to hERG channels that required in vitro data for kinetics of block, in addition to potency, to constrain them. To date, development and validation has been undertaken using data from manual patch-clamp. The application of this approach at scale requires the development of a high-throughput, automated patch-clamp (APC) implementation. Here, we present a comprehensive analysis of the implementation of the Milnes, or CiPA dynamic protocol, on an APC platform, including quality control and data analysis. Kinetics and potency of block were assessed for bepridil, cisapride, terfenadine and verapamil with data retention/QC pass rate of 21.8% overall, or as high as 50.4% when only appropriate sweep lengths were considered for drugs with faster kinetics. The variability in IC50 and kinetics between manual and APC was comparable to that seen between sites/platforms in previous APC studies of potency. Whilst the experimental success is less than observed in screens of potency alone, it is still significantly greater than manual patch. With the modifications to protocol design, including sweep length, number of repetitions, and leak correction recommended in this study, this protocol can be applied on APC to acquire data comparable to manual patch clamp.
Collapse
|
6
|
Koda Y, Sato S, Yamamoto H, Niwa H, Watanabe H, Watanabe C, Sato T, Nakamura K, Tanaka A, Shirouzu M, Honma T, Fukami T, Koyama H, Umehara T. Design and Synthesis of Tranylcypromine-Derived LSD1 Inhibitors with Improved hERG and Microsomal Stability Profiles. ACS Med Chem Lett 2022; 13:848-854. [PMID: 35586426 PMCID: PMC9109268 DOI: 10.1021/acsmedchemlett.2c00120] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 04/21/2022] [Indexed: 12/26/2022] Open
Abstract
Lysine-specific demethylase 1 (LSD1/KDM1A) is a promising therapeutic target for the treatment of cancers. Several derivatives of tranylcypromine (trans-2-phenylcyclopropylamine) have been developed as LSD1 inhibitors. One such derivative is S2157; however, this compound has a high hERG channel inhibitory activity and a low microsomal stability, making it unsuitable as a drug candidate. Here, using an in silico hERG inhibition prediction model, we designed, synthesized, and evaluated a novel series of S2157 derivatives characterized by modifications of the benzyloxy and piperazine groups. Among the synthesized derivatives, a compound possessing 2-fluoropyridine and 2,8-diaza-spiro[4.5]decane groups (compound 10) showed the most desirable activities, and its eutomer, S1427, was isolated by the optical resolution of 10. In addition to potent LSD1 inhibitory activity, S1427 exhibited desirable hERG channel inhibition and microsomal stability profiles.
Collapse
Affiliation(s)
- Yasuko Koda
- Drug Discovery Chemistry Platform Unit, Drug Discovery Seed Compounds Exploratory Unit, Chemical Genomics Research Group, RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Shin Sato
- Laboratory for Epigenetics Drug Discovery, RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi, Yokohama 230-0045, Japan
- Drug Discovery Structural Biology Platform Unit, RIKEN Center for Biosystems Dynamic Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Hirofumi Yamamoto
- Drug Discovery Chemistry Platform Unit, Drug Discovery Seed Compounds Exploratory Unit, Chemical Genomics Research Group, RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Hideaki Niwa
- Laboratory for Epigenetics Drug Discovery, RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi, Yokohama 230-0045, Japan
- Drug Discovery Structural Biology Platform Unit, RIKEN Center for Biosystems Dynamic Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Hisami Watanabe
- Laboratory for Epigenetics Drug Discovery, RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi, Yokohama 230-0045, Japan
- Drug Discovery Structural Biology Platform Unit, RIKEN Center for Biosystems Dynamic Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Chiduru Watanabe
- Drug Discovery Computational Chemistry Platform Unit, RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi, Yokohama 230-0045, Japan
| | - Tomohiro Sato
- Drug Discovery Computational Chemistry Platform Unit, RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi, Yokohama 230-0045, Japan
| | - Kana Nakamura
- Drug Discovery Structural Biology Platform Unit, RIKEN Center for Biosystems Dynamic Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Akiko Tanaka
- Drug Discovery Structural Biology Platform Unit, RIKEN Center for Biosystems Dynamic Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Mikako Shirouzu
- Drug Discovery Structural Biology Platform Unit, RIKEN Center for Biosystems Dynamic Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Teruki Honma
- Drug Discovery Computational Chemistry Platform Unit, RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi, Yokohama 230-0045, Japan
| | - Takehiro Fukami
- RIKEN Program for Drug Discovery and Medical Technology Platforms, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Hiroo Koyama
- Drug Discovery Chemistry Platform Unit, Drug Discovery Seed Compounds Exploratory Unit, Chemical Genomics Research Group, RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Takashi Umehara
- Laboratory for Epigenetics Drug Discovery, RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi, Yokohama 230-0045, Japan
- Drug Discovery Structural Biology Platform Unit, RIKEN Center for Biosystems Dynamic Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| |
Collapse
|
7
|
Yehia A, Wei H. Studying Nicotinic Acetylcholine Receptors Using the IonFlux™ Microfluidic-Based Automated Patch-Clamp System with Continuous Perfusion and Fast Solution Exchange. ACTA ACUST UNITED AC 2021; 88:e73. [PMID: 32068960 DOI: 10.1002/cpph.73] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Automated patch-clamp (APC) systems have become indispensable tools of drug-discovery programs by allowing high-throughput electrophysiology-based screening of ion channel compounds. The recent development and introduction of microfluidics-based APC systems have made it possible to study the interactions of ligand-gated ion channels with pharmacological reagents, such as agonists, antagonists, or positive allosteric modulators (PAMs), with reliable pharmacological results comparable to those of the gold-standard manual patch-clamp technique while maintaining high-throughput capacity. Many ligand-gated ion channels exhibit rapid desensitization upon repetitive introduction of ligands; this loss of channel activity in the absence of pharmacological interaction poses a challenge for developing accurate, precise, and robust assays with high success rate, low run-down, and reliable pharmacological results. Here we present procedures to study nicotinic acetylcholine receptors (nAChRs) with the IonFlux™, an automated patch-clamp system with continuous flow and precise fluidic exchange; these procedures can also be generalized to the study of other ligand-gated ion channels. We present protocols to study agonist, antagonist, and PAM activities on nAChRs, particularly the rapidly desensitizing nAChR α7 receptors. The data demonstrate that the IonFlux™ system is a fast, robust, and reliable platform for the study of nAChRs and other ligand-gated ion channels, generating data that closely mimic those from manual patch-clamp conditions. © 2020 by John Wiley & Sons, Inc. Basic Protocol 1: Measuring agonist concentration-dependent response Basic Protocol 2: Measuring antagonist concentration-dependent response Basic Protocol 3: Measuring positive allosteric modulator (PAM) concentration-dependent response Support Protocol 1: Basic IonFlux system operation Support Protocol 2: Plate care and filling Support Protocol 3: Plate preparation for water rinsing Support Protocol 4: Water rinsing of plates Support Protocol 5: Plate priming Support Protocol 6: General assay Support Protocol 7: Editing the compound addition sequence (compound list) Support Protocol 8: Creating compound list for agonist concentration-dependent response Support Protocol 9: Creating compound list for antagonist or PAM concentration-dependent response Support Protocol 10: Defining the different compounds used or compound list Support Protocol 11: Maintenance Support Protocol 12: Data analysis Support Protocol 13: Cell culture.
Collapse
Affiliation(s)
- Ali Yehia
- Fluxion Biosciences, Alameda, California
| | - Haiyang Wei
- Eurofins Discovery, Eurofins Panlabs, Inc., St. Charles, Missouri
| |
Collapse
|
8
|
Bae M, Roh JD, Kim Y, Kim SS, Han HM, Yang E, Kang H, Lee S, Kim JY, Kang R, Jung H, Yoo T, Kim H, Kim D, Oh H, Han S, Kim D, Han J, Bae YC, Kim H, Ahn S, Chan AM, Lee D, Kim JW, Kim E. SLC6A20 transporter: a novel regulator of brain glycine homeostasis and NMDAR function. EMBO Mol Med 2021; 13:e12632. [PMID: 33428810 PMCID: PMC7863395 DOI: 10.15252/emmm.202012632] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 10/22/2020] [Accepted: 11/19/2020] [Indexed: 12/25/2022] Open
Abstract
Glycine transporters (GlyT1 and GlyT2) that regulate levels of brain glycine, an inhibitory neurotransmitter with co-agonist activity for NMDA receptors (NMDARs), have been considered to be important targets for the treatment of brain disorders with suppressed NMDAR function such as schizophrenia. However, it remains unclear whether other amino acid transporters expressed in the brain can also regulate brain glycine levels and NMDAR function. Here, we report that SLC6A20A, an amino acid transporter known to transport proline based on in vitro data but is understudied in the brain, regulates proline and glycine levels and NMDAR function in the mouse brain. SLC6A20A transcript and protein levels were abnormally increased in mice carrying a mutant PTEN protein lacking the C terminus through enhanced β-catenin binding to the Slc6a20a gene. These mice displayed reduced extracellular levels of brain proline and glycine and decreased NMDAR currents. Elevating glycine levels back to normal ranges by antisense oligonucleotide-induced SLC6A20 knockdown, or the competitive GlyT1 antagonist sarcosine, normalized NMDAR currents and repetitive climbing behavior observed in these mice. Conversely, mice lacking SLC6A20A displayed increased extracellular glycine levels and NMDAR currents. Lastly, both mouse and human SLC6A20 proteins mediated proline and glycine transports, and SLC6A20 proteins could be detected in human neurons. These results suggest that SLC6A20 regulates proline and glycine homeostasis in the brain and that SLC6A20 inhibition has therapeutic potential for brain disorders involving NMDAR hypofunction.
Collapse
Affiliation(s)
- Mihyun Bae
- Center for Synaptic Brain DysfunctionsInstitute for Basic Science (IBS)DaejeonKorea
| | - Junyeop Daniel Roh
- Department of Biological SciencesKorea Advanced Institute for Science and Technology (KAIST)DaejeonKorea
| | - Youjoung Kim
- Department of Biological SciencesKorea Advanced Institute for Science and Technology (KAIST)DaejeonKorea
| | - Seong Soon Kim
- Therapeutics and Biotechnology DivisionKorea Research Institute of Chemical Technology (KRICT)DaejeonKorea
| | - Hye Min Han
- Department of Anatomy and NeurobiologySchool of DentistryKyungpook National UniversityDaeguKorea
| | - Esther Yang
- Department of Anatomy and Division of Brain Korea 21Biomedical ScienceCollege of MedicineKorea UniversitySeoulKorea
| | - Hyojin Kang
- Division of National SupercomputingKISTIDaejeonKorea
| | - Suho Lee
- Center for Synaptic Brain DysfunctionsInstitute for Basic Science (IBS)DaejeonKorea
| | - Jin Yong Kim
- Department of Anatomy and Division of Brain Korea 21Biomedical ScienceCollege of MedicineKorea UniversitySeoulKorea
| | - Ryeonghwa Kang
- Department of Biological SciencesKorea Advanced Institute for Science and Technology (KAIST)DaejeonKorea
| | - Hwajin Jung
- Center for Synaptic Brain DysfunctionsInstitute for Basic Science (IBS)DaejeonKorea
| | - Taesun Yoo
- Center for Synaptic Brain DysfunctionsInstitute for Basic Science (IBS)DaejeonKorea
| | - Hyosang Kim
- Department of Biological SciencesKorea Advanced Institute for Science and Technology (KAIST)DaejeonKorea
| | - Doyoun Kim
- Center for Synaptic Brain DysfunctionsInstitute for Basic Science (IBS)DaejeonKorea
| | - Heejeong Oh
- Department of Biological SciencesKorea Advanced Institute for Science and Technology (KAIST)DaejeonKorea
| | - Sungwook Han
- Department of Biological SciencesKorea Advanced Institute for Science and Technology (KAIST)DaejeonKorea
| | - Dayeon Kim
- Graduate School of Medical Science and EngineeringKAISTDaejeonKorea
| | - Jinju Han
- Graduate School of Medical Science and EngineeringKAISTDaejeonKorea
| | - Yong Chul Bae
- Department of Anatomy and NeurobiologySchool of DentistryKyungpook National UniversityDaeguKorea
| | - Hyun Kim
- Department of Anatomy and Division of Brain Korea 21Biomedical ScienceCollege of MedicineKorea UniversitySeoulKorea
| | - Sunjoo Ahn
- Therapeutics and Biotechnology DivisionKorea Research Institute of Chemical Technology (KRICT)DaejeonKorea
| | - Andrew M Chan
- School of Biomedical SciencesThe Chinese University of Hong KongHong KongHong Kong SARChina
| | - Daeyoup Lee
- Department of Biological SciencesKorea Advanced Institute for Science and Technology (KAIST)DaejeonKorea
| | - Jin Woo Kim
- Department of Biological SciencesKorea Advanced Institute for Science and Technology (KAIST)DaejeonKorea
| | - Eunjoon Kim
- Center for Synaptic Brain DysfunctionsInstitute for Basic Science (IBS)DaejeonKorea
- Department of Biological SciencesKorea Advanced Institute for Science and Technology (KAIST)DaejeonKorea
| |
Collapse
|
9
|
Bell DC, Dallas ML. Advancing Ion Channel Research with Automated Patch Clamp (APC) Electrophysiology Platforms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1349:21-32. [DOI: 10.1007/978-981-16-4254-8_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
10
|
Alam MK, Koomson E, Zou H, Yi C, Li CW, Xu T, Yang M. Recent advances in microfluidic technology for manipulation and analysis of biological cells (2007–2017). Anal Chim Acta 2018; 1044:29-65. [DOI: 10.1016/j.aca.2018.06.054] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 06/19/2018] [Accepted: 06/19/2018] [Indexed: 12/17/2022]
|
11
|
Obergrussberger A, Goetze TA, Brinkwirth N, Becker N, Friis S, Rapedius M, Haarmann C, Rinke-Weiß I, Stölzle-Feix S, Brüggemann A, George M, Fertig N. An update on the advancing high-throughput screening techniques for patch clamp-based ion channel screens: implications for drug discovery. Expert Opin Drug Discov 2018; 13:269-277. [DOI: 10.1080/17460441.2018.1428555] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
12
|
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. [PMID: 28622411 DOI: 10.1111/bph.13916] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Automated patch clamp (APC) technology was first developed at the turn of the millennium. The increased throughput it afforded promised a new paradigm in ion channel recordings, offering the potential to overcome the time-consuming, low-throughput bottleneck, arising from manual patch clamp investigations. This has relevance to the fast-paced development of novel therapies for chronic pain. This review highlights the advances in technology, using select examples that have facilitated APC usage in both industry and academia. It covers both first generation and the latest developments in second-generation platforms. In addition, it also provides an overview of the pain research field and how APC platforms have furthered our understanding of ion channel research and the development of pharmacological tools and therapeutics. APC platforms have much to offer to the ion channel research community, and this review highlights areas of best practice for both academia and industry. The impact of APC platforms and the prospects of ion channel research and improved therapeutics for chronic pain will be evaluated. LINKED ARTICLES This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc.
Collapse
Affiliation(s)
| | - Mark L Dallas
- School of Pharmacy, University of Reading, Reading, UK
| |
Collapse
|
13
|
Zou B. Ion channel profiling to advance drug discovery and development. DRUG DISCOVERY TODAY. TECHNOLOGIES 2015; 18:18-23. [PMID: 26723888 DOI: 10.1016/j.ddtec.2015.10.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 10/07/2015] [Indexed: 06/05/2023]
Abstract
In vitro pharmacological profiling provides crucial information to eliminate drug candidates with potential toxicity early in drug discovery and reduce failure in later stages. It has become a common practice in industry to test lead compounds against a panel of ion channel targets for selectivity and safety liability at early drug discovery stages. Ion channel profiling technologies include binding assays, flux assays, fluorescent membrane potential assays, automated and conventional electrophysiology. Instead of examining compound effects on individual ion channel targets, automated current clamp, optical electrophysiology, and multi-electrode assays have evolved to investigate the integrated compound effects on cardiac myocytes. This review aims to provide an overview of ion channel profiling for cardiac safety and comparisons of various technologies.
Collapse
Affiliation(s)
- Beiyan Zou
- Molecular Devices LLC, 1311 Orleans Drive, Sunnyvale, CA 94089, United States.
| |
Collapse
|
14
|
Auzmendi JA, Smoler M, Moffatt L. Dynamics of T-Junction Solution Switching Aimed at Patch Clamp Experiments. PLoS One 2015; 10:e0133187. [PMID: 26177538 PMCID: PMC4503516 DOI: 10.1371/journal.pone.0133187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 06/23/2015] [Indexed: 11/29/2022] Open
Abstract
Solutions exchange systems are responsible for the timing of drug application on patch clamp experiments. There are two basic strategies for generating a solution exchange. When slow exchanges are bearable, it is easier to perform the exchange inside the tubing system upstream of the exit port. On the other hand, fast, reproducible, exchanges are usually performed downstream of the exit port. As both strategies are combinable, increasing the performance of upstream exchanges is desirable. We designed a simple method for manufacturing T-junctions (300 μm I.D.) and we measured the time profile of exchange of two saline solutions using a patch pipette with an open tip. Three factors were found to determine the timing of the solution switching: pressure, travelled distance and off-center distance. A linear relationship between the time delay and the travelled distance was found for each tested pressure, showing its dependence to the fluid velocity, which increased with pressure. The exchange time was found to increase quadratically with the delay, although a sizeable variability remains unexplained by this relationship. The delay and exchange times increased as the recording pipette moved away from the center of the stream. Those increases became dramatic as the pipette was moved close to the stream borders. Mass transport along the travelled distance between the slow fluid at the border and the fast fluid at the center seems to contribute to the time course of the solution exchange. This effect would be present in all tubing based devices. Present results might be of fundamental importance for the adequate design of serial compound exchangers which would be instrumental in the discovery of drugs that modulate the action of the physiological agonists of ion channels with the purpose of fine tuning their physiology.
Collapse
Affiliation(s)
- Jerónimo A. Auzmendi
- Instituto de Química Física de los Materiales, Medio Ambiente y Energía. Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mariano Smoler
- Instituto de Química Física de los Materiales, Medio Ambiente y Energía. Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Luciano Moffatt
- Instituto de Química Física de los Materiales, Medio Ambiente y Energía. Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
15
|
Wong LW, Tae HS, Cromer BA. Assembly, trafficking and function of α1β2γ2 GABAA receptors are regulated by N-terminal regions, in a subunit-specific manner. J Neurochem 2015; 134:819-32. [PMID: 26016529 DOI: 10.1111/jnc.13175] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 04/16/2015] [Accepted: 05/09/2015] [Indexed: 02/01/2023]
Abstract
GABAA receptors are pentameric ligand-gated ion channels that mediate inhibitory fast synaptic transmission in the central nervous system. Consistent with recent pentameric ligand-gated ion channels structures, sequence analysis predicts an α-helix near the N-terminus of each GABAA receptor subunit. Preceding each α-helix are 8-36 additional residues, which we term the N-terminal extension. In homomeric GABAC receptors and nicotinic acetylcholine receptors, the N-terminal α-helix is functionally essential. Here, we determined the role of the N-terminal extension and putative α-helix in heteromeric α1β2γ2 GABAA receptors. This role was most prominent in the α1 subunit, with deletion of the N-terminal extension or further deletion of the putative α-helix both dramatically reduced the number of functional receptors at the cell surface. Conversely, deletion of the β2 or γ2 N-terminal extension had little effect on the number of functional cell surface receptors. Additional deletion of the putative α-helix in the β2 or γ2 subunits did, however, decrease both functional cell surface receptors and incorporation of the γ2 subunit into mature receptors. In the β2 subunit only, α-helix deletions affected GABA sensitivity and desensitization. Our findings demonstrate that N-terminal extensions and α-helices make key subunit-specific contributions to assembly, consistent with both regions being involved in inter-subunit interactions. N-terminal α-helices and preceding sequences of eukaryotic pentameric ligand-gated ion channels are absent in prokaryotic homologues, suggesting they may not be functionally essential. Here, we show that in heteropentameric α1β2γ2 GABAA receptors, the role of these segments is highly subunit dependent. The extension preceding the α-helix in the α subunit is crucial for assembly and trafficking, but is of little importance in β and γ subunits. Indeed, robust receptor levels remain when the extension and α-helix are removed in β or γ subunits.
Collapse
Affiliation(s)
- Lik-Wei Wong
- Health Innovation Research Institute, School of Medical Sciences, RMIT University, Melbourne, Vic., Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Vic., Australia
| | - Han-Shen Tae
- Health Innovation Research Institute, School of Medical Sciences, RMIT University, Melbourne, Vic., Australia
| | - Brett A Cromer
- Health Innovation Research Institute, School of Medical Sciences, RMIT University, Melbourne, Vic., Australia
| |
Collapse
|
16
|
Thoma G, Smith AB, van Eis MJ, Vangrevelinghe E, Blanz J, Aichholz R, Littlewood-Evans A, Lee CC, Liu H, Zerwes HG. Discovery and Profiling of a Selective and Efficacious Syk Inhibitor. J Med Chem 2015; 58:1950-63. [DOI: 10.1021/jm5018863] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
| | | | | | | | | | | | | | - Christian C. Lee
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Hong Liu
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | | |
Collapse
|
17
|
Wong LW, Tae HS, Cromer BA. Role of the ρ1 GABA(C) receptor N-terminus in assembly, trafficking and function. ACS Chem Neurosci 2014; 5:1266-77. [PMID: 25347026 DOI: 10.1021/cn500220t] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The GABAC receptor and closely related GABAA receptor are members of the pentameric ligand-gated ion channels (pLGICs) superfamily and mediate inhibitory fast synaptic transmission in the nervous system. Each pLGIC subunit comprises an N-terminal extracellular agonist-binding domain followed by a channel domain and a variable intracellular domain. Available structural information shows that the core of the agonist-binding domain is a β sandwich of ten β-strands, which form the agonist-binding pocket at the subunit interface. This β-sandwich is preceded by an N-terminal α-helix in eukaryotic structures but not in prokaryotic structures. The N-terminal α-helix has been shown to be functionally essential in α7 nicotinic acetylcholine receptors. Sequence analysis of GABAC and GABAA receptors predicts an α-helix in a similar position but preceded by 8 to 46 additional residues, of unknown function, which we term the N-terminal extension. To test the functional role of both the N-terminal extension and the putative N-terminal α-helix in the ρ1 GABAC receptor, we created a series of deletions from the N-terminus. The N-terminal extension was not functionally essential, but its removal did reduce both cell surface expression and cooperativity of agonist-gated channel function. Further deletion of the putative N-terminal α-helix abolished receptor function by preventing cell-surface expression. Our results further demonstrate the essential role of the N-terminal α-helix in the assembly and trafficking of eukaryotic pLGICs. They also provide evidence that the N-terminal extension, although not essential, contributes to receptor assembly, trafficking and conformational changes associated with ligand gating.
Collapse
Affiliation(s)
- Lik-Wei Wong
- Health
Innovation Research Institute, School of Medical Sciences, RMIT University, Bundoora, Victoria 3083, Australia
- Department
of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Han-Shen Tae
- Health
Innovation Research Institute, School of Medical Sciences, RMIT University, Bundoora, Victoria 3083, Australia
| | - Brett A. Cromer
- Health
Innovation Research Institute, School of Medical Sciences, RMIT University, Bundoora, Victoria 3083, Australia
| |
Collapse
|
18
|
Kauthale RR, Dadarkar SS, Husain R, Karande VV, Gatne MM. Assessment of temperature-induced hERG channel blockade variation by drugs. J Appl Toxicol 2014; 35:799-805. [PMID: 25348819 DOI: 10.1002/jat.3074] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Revised: 08/20/2014] [Accepted: 08/20/2014] [Indexed: 12/21/2022]
Abstract
Drug-induced QT prolongation has been reported in humans and animals. This potentially lethal effect can be induced by drugs interacting with a cardiac potassium channel, namely hERG (human ether-a go-go-related gene) leading to arrhythmia or torsade de pointes (TdP). Hence, in vitro evaluation of therapeutics for their effects on the rapid delayed rectifier current (IKr) mediated by the K(+) ion channel encoded by hERG is a valuable tool for identifying potential arrhythmic side effects during drug safety testing. Our objective was to evaluate the temperature-induced hERG channel blockade variation by human and veterinary drugs using the IonFlux 16 system. A panel of eight drugs was tested for IKr inhibition at both ambient (23 °C) and physiological (37 °C) temperatures at various concentrations using IonFlux 16, an automated patch clamp system. Our results established that both amiodarone (IC(50) = 0.56 μM at 23 °C and 0.30 μM at 37 °C) and β-estradiol (IC(50) = 24.72 μM at 23 °C and 8.17 μM at 37 °C) showed a dose-dependent IKr blockade with a higher blockade at 37 °C. Whereas, blockade of IKr by both ivermectin (IC(50) = 12.52 μM at 23 °C and 24.41 μM at 37 °C) and frusemide (IC(50) = 12.58 μM at 23 °C and 25.55 μM at 37 °C) showed a dose-dependent IKr blockade with a lower blockade at 37 °C. Gentamicin, enrofloxacin, xylazine and albendazole did not block IKr at both the assessed temperatures. Collectively, these results demonstrate that the effect of temperature variation should be taken into consideration during the evaluation of test drugs for their hERG channel blockade potential.
Collapse
Affiliation(s)
- Rahul R Kauthale
- Department of Pharmacology and Toxicology, Bombay Veterinary College, Mumbai, 400012, Maharashtra, India
| | - Shruta S Dadarkar
- Department of Toxicology, Piramal Enterprises Limited, Mumbai, 400063, Maharashtra, India
| | - Raghib Husain
- Department of Toxicology, Piramal Enterprises Limited, Mumbai, 400063, Maharashtra, India
| | - Vikas V Karande
- Department of Pharmacology and Toxicology, Bombay Veterinary College, Mumbai, 400012, Maharashtra, India
| | - Madhumanjiri M Gatne
- Department of Pharmacology and Toxicology, Bombay Veterinary College, Mumbai, 400012, Maharashtra, India
| |
Collapse
|
19
|
Ferreiro SF, Vilariño N, Louzao MC, Nicolaou KC, Frederick MO, Botana LM. In vitro chronic effects on hERG channel caused by the marine biotoxin azaspiracid-2. Toxicon 2014; 91:69-75. [PMID: 25286396 DOI: 10.1016/j.toxicon.2014.09.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 09/16/2014] [Accepted: 09/25/2014] [Indexed: 11/27/2022]
Abstract
Azaspiracids (AZAs) are marine biotoxins produced by the dinoflagellate Azadinium spinosum that accumulate in many shellfish species. Azaspiracid poisoning caused by AZA-contaminated seafood consumption is primarily manifested by diarrhea in humans. To protect human health, AZA-1, AZA-2 and AZA-3 content in seafood has been regulated by food safety authorities in many countries. Recently AZAs have been reported as a low/moderate hERG channel blockers. Furthermore AZA-2 has been related to arrhythmia appearance in rats, suggesting potential heart toxicity. In this study AZA-2 in vitro effects on hERG channel after chronic exposure are analyzed to further explore potential cardiotoxicity. The amount of hERG channel in the plasma membrane, hERG channel trafficking and hERG currents were evaluated up to 12 h of toxin exposure. In these conditions AZA-2 caused an increase of hERG levels in the plasma membrane, probably related to hERG retrograde trafficking impairment. Although this alteration did not translate into an increase of hERG channel-related current, more studies will be necessary to understand its mechanism and to know what consequences could have in vivo. These findings suggest that azaspiracids might have chronic cardiotoxicity related to hERG channel trafficking and they should not be overlooked when evaluating the threat to human health.
Collapse
Affiliation(s)
- Sara F Ferreiro
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain
| | - Natalia Vilariño
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain.
| | - M Carmen Louzao
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain
| | - K C Nicolaou
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA; Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Michael O Frederick
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Luis M Botana
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain.
| |
Collapse
|
20
|
Danker T, Möller C. Early identification of hERG liability in drug discovery programs by automated patch clamp. Front Pharmacol 2014; 5:203. [PMID: 25228880 PMCID: PMC4151236 DOI: 10.3389/fphar.2014.00203] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 08/14/2014] [Indexed: 12/26/2022] Open
Abstract
Blockade of the cardiac ion channel coded by human ether-à-gogo-related gene (hERG) can lead to cardiac arrhythmia, which has become a major concern in drug discovery and development. Automated electrophysiological patch clamp allows assessment of hERG channel effects early in drug development to aid medicinal chemistry programs and has become routine in pharmaceutical companies. However, a number of potential sources of errors in setting up hERG channel assays by automated patch clamp can lead to misinterpretation of data or false effects being reported. This article describes protocols for automated electrophysiology screening of compound effects on the hERG channel current. Protocol details and the translation of criteria known from manual patch clamp experiments to automated patch clamp experiments to achieve good quality data are emphasized. Typical pitfalls and artifacts that may lead to misinterpretation of data are discussed. While this article focuses on hERG channel recordings using the QPatch (Sophion A/S, Copenhagen, Denmark) technology, many of the assay and protocol details given in this article can be transferred for setting up different ion channel assays by automated patch clamp and are similar on other planar patch clamp platforms.
Collapse
Affiliation(s)
| | - Clemens Möller
- Life Sciences Faculty, Albstadt-Sigmaringen University of Applied Sciences Sigmaringen, Germany
| |
Collapse
|
21
|
Thoma G, Blanz J, Bühlmayer P, Drückes P, Kittelmann M, Smith AB, van Eis M, Vangrevelinghe E, Zerwes HG, Che J(J, He X, Jin Y, Lee CC, Michellys PY, Uno T, Liu H. Syk inhibitors with high potency in presence of blood. Bioorg Med Chem Lett 2014; 24:2278-82. [DOI: 10.1016/j.bmcl.2014.03.075] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 03/24/2014] [Accepted: 03/25/2014] [Indexed: 11/16/2022]
|
22
|
HTS assays for developing the molecular pharmacology of ion channels. Curr Opin Pharmacol 2014; 15:91-6. [DOI: 10.1016/j.coph.2014.01.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 01/24/2014] [Accepted: 01/27/2014] [Indexed: 01/29/2023]
|
23
|
Tanzi S, Matteucci M, Christiansen TL, Friis S, Christensen MT, Garnaes J, Wilson S, Kutchinsky J, Taboryski R. Ion channel recordings on an injection-molded polymer chip. LAB ON A CHIP 2013; 13:4784-4793. [PMID: 24154831 DOI: 10.1039/c3lc50760b] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
In this paper, we demonstrate recordings of the ion channel activity across the cell membrane in a biological cell by employing the so-called patch clamping technique on an injection-molded polymer microfluidic device. The findings will allow direct recordings of ion channel activity to be made using the cheapest materials and production platform to date and with the potential for very high throughput. The employment of cornered apertures for cell capture allowed the fabrication of devices without through holes and via a scheme comprising master origination by dry etching in a silicon substrate, electroplating in nickel and injection molding of the final part. The most critical device parameters were identified as the length of the patching capillary and the very low surface roughness on the inside of the capillary. The cross-sectional shape of the orifice was found to be less critical, as both rectangular and semicircular profiles seemed to have almost the same ability to form tight seals with cells with negligible leak currents. The devices were functionally tested using human embryonic kidney cells expressing voltage-gated sodium channels (Nav1.7) and benchmarked against a commercial state-of-the-art system for automated ion channel recordings. These experiments considered current-voltage (IV) relationships for activation and inactivation of the Nav1.7 channels and their sensitivity to a local anesthetic, lidocaine. Both IVs and lidocaine dose-response curves obtained from the injection-molded polymer device were in good agreement with data obtained from the commercial system.
Collapse
Affiliation(s)
- Simone Tanzi
- Department of Micro- and Nanotechnology, Technical University of Denmark, Building 345E, DK-2800 Kongens Lyngby, Denmark.
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Raphemot R, Kadakia RJ, Olsen ML, Banerjee S, Days E, Smith SS, Weaver CD, Denton JS. Development and validation of fluorescence-based and automated patch clamp-based functional assays for the inward rectifier potassium channel Kir4.1. Assay Drug Dev Technol 2013; 11:532-43. [PMID: 24266659 DOI: 10.1089/adt.2013.544] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The inward rectifier potassium (Kir) channel Kir4.1 plays essential roles in modulation of neurotransmission and renal sodium transport and may represent a novel drug target for temporal lobe epilepsy and hypertension. The molecular pharmacology of Kir4.1 is limited to neurological drugs, such as fluoxetine (Prozac(©)), exhibiting weak and nonspecific activity toward the channel. The development of potent and selective small-molecule probes would provide critically needed tools for exploring the integrative physiology and therapeutic potential of Kir4.1. A fluorescence-based thallium (Tl(+)) flux assay that utilizes a tetracycline-inducible T-Rex-HEK293-Kir4.1 cell line to enable high-throughput screening (HTS) of small-molecule libraries was developed. The assay is dimethyl sulfoxide tolerant and exhibits robust screening statistics (Z'=0.75±0.06). A pilot screen of 3,655 small molecules and lipids revealed 16 Kir4.1 inhibitors (0.4% hit rate). 3,3-Diphenyl-N-(1-phenylethyl)propan-1-amine, termed VU717, inhibits Kir4.1-mediated thallium flux with an IC50 of ∼6 μM. An automated patch clamp assay using the IonFlux HT workbench was developed to facilitate compound characterization. Leak-subtracted ensemble "loose patch" recordings revealed robust tetracycline-inducible and Kir4.1 currents that were inhibited by fluoxetine (IC50=10 μM), VU717 (IC50=6 μM), and structurally related calcium channel blocker prenylamine (IC50=6 μM). Finally, we demonstrate that VU717 inhibits Kir4.1 channel activity in cultured rat astrocytes, providing proof-of-concept that the Tl(+) flux and IonFlux HT assays can enable the discovery of antagonists that are active against native Kir4.1 channels.
Collapse
Affiliation(s)
- Rene Raphemot
- 1 Department of Anesthesiology, Vanderbilt University School of Medicine , Nashville, Tennessee
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Elkins RC, Davies MR, Brough SJ, Gavaghan DJ, Cui Y, Abi-Gerges N, Mirams GR. Variability in high-throughput ion-channel screening data and consequences for cardiac safety assessment. J Pharmacol Toxicol Methods 2013; 68:112-22. [PMID: 23651875 PMCID: PMC4135079 DOI: 10.1016/j.vascn.2013.04.007] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 04/05/2013] [Accepted: 04/25/2013] [Indexed: 11/29/2022]
Abstract
Introduction Unwanted drug interactions with ionic currents in the heart can lead to an increased proarrhythmic risk to patients in the clinic. It is therefore a priority for safety pharmacology teams to detect block of cardiac ion channels, and new technologies have enabled the development of automated and high-throughput screening assays using cell lines. As a result of screening multiple ion-channels there is a need to integrate information, particularly for compounds affecting more than one current, and mathematical electrophysiology in-silico action potential models are beginning to be used for this. Methods We quantified the variability associated with concentration-effect curves fitted to recordings from high-throughput Molecular Devices IonWorks® Quattro™ screens when detecting block of IKr (hERG), INa (NaV1.5), ICaL (CaV1.2), IKs (KCNQ1/minK) and Ito (Kv4.3/KChIP2.2), and the Molecular Devices FLIPR® Tetra fluorescence screen for ICaL (CaV1.2), for control compounds used at AstraZeneca and GlaxoSmithKline. We examined how screening variability propagates through in-silico action potential models for whole cell electrical behaviour, and how confidence intervals on model predictions can be estimated with repeated simulations. Results There are significant levels of variability associated with high-throughput ion channel electrophysiology screens. This variability is of a similar magnitude for different cardiac ion currents and different compounds. Uncertainty in the Hill coefficients of reported concentration-effect curves is particularly high. Depending on a compound’s ion channel blocking profile, the uncertainty introduced into whole-cell predictions can become significant. Discussion Our technique allows confidence intervals to be placed on computational model predictions that are based on high-throughput ion channel screens. This allows us to suggest when repeated screens should be performed to reduce uncertainty in a compound’s action to acceptable levels, to allow a meaningful interpretation of the data.
Collapse
Affiliation(s)
- Ryan C Elkins
- Global Safety Pharmacology, Global Safety Assessment, AstraZeneca, Alderley Park SK10 4TG, UK
| | | | | | | | | | | | | |
Collapse
|
26
|
Su X, Theberge AB, January CT, Beebe DJ. Effect of microculture on cell metabolism and biochemistry: do cells get stressed in microchannels? Anal Chem 2013; 85:1562-70. [PMID: 23327437 DOI: 10.1021/ac3027228] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Microfluidics is emerging as a promising platform for cell culture, enabling increased microenvironment control and potential for integrated analysis compared to conventional macroculture systems such as well plates and Petri dishes. To advance the use of microfluidic devices for cell culture, it is necessary to better understand how miniaturization affects cell behavior. In particular, microfluidic devices have significantly higher surface-area-to-volume ratios than conventional platforms, resulting in lower volumes of media per cell, which can lead to cell stress. We investigated cell stress under a variety of culture conditions using three cell lines: parental HEK (human embryonic kidney) cells and transfected HEK cells that stably express wild-type (WT) and mutant (G601S) human ether-a-go-go related gene (hERG) potassium channel protein. These three cell lines provide a unique model system through which to study cell-type-specific responses in microculture because mutant hERG is known to be sensitive to environmental conditions, making its expression a particularly sensitive readout through which to compare macro- and microculture. While expression of WT-hERG was similar in microchannel and well culture, the expression of mutant G601S-hERG was reduced in microchannels. Expression of the endoplasmic reticulum (ER) stress marker immunoglobulin binding protein (BiP) was upregulated in all three cell lines in microculture. Using BiP expression, glucose consumption, and lactate accumulation as readouts we developed methods for reducing ER stress including properly increasing the frequency of media replacement, reducing cell seeding density, and adjusting the serum concentration and buffering capacity of culture medium. Indeed, increasing the buffering capacity of culture medium or frequency of media replacement partially restored the expression of the G601S-hERG in microculture. This work illuminates how biochemical properties of cells differ in macro- and microculture and suggests strategies that can be used to modify cell culture protocols for future studies involving miniaturized culture platforms.
Collapse
Affiliation(s)
- Xiaojing Su
- Department of Biomedical Engineering, University of Wisconsin-Madison, Wisconsin Institutes for Medical Research, 1111 Highland Avenue, Madison, Wisconsin 53705, United States
| | | | | | | |
Collapse
|
27
|
Gillie DJ, Novick SJ, Donovan BT, Payne LA, Townsend C. Development of a high-throughput electrophysiological assay for the human ether-à-go-go related potassium channel hERG. J Pharmacol Toxicol Methods 2013; 67:33-44. [DOI: 10.1016/j.vascn.2012.10.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Revised: 10/02/2012] [Accepted: 10/18/2012] [Indexed: 01/03/2023]
|
28
|
Chen Q, Yim PD, Yuan N, Johnson J, Cook JM, Smith S, Ionescu-Zanetti C, Wang ZJ, Arnold LA, Emala CW. Comparison of cell expression formats for the characterization of GABA(A) channels using a microfluidic patch clamp system. Assay Drug Dev Technol 2012; 10:325-35. [PMID: 22574655 DOI: 10.1089/adt.2011.415] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Ensemble recording and microfluidic perfusion are recently introduced techniques aimed at removing the laborious nature and low recording success rates of manual patch clamp. Here, we present assay characteristics for these features integrated into one automated electrophysiology platform as applied to the study of GABA(A) channels. A variety of cell types and methods of GABA(A) channel expression were successfully studied (defined as I(GABA)>500 pA), including stably transfected human embryonic kidney (HEK) cells expressing α(1)β(3)γ(2) GABA(A) channels, frozen ready-to-assay (RTA) HEK cells expressing α(1)β(3)γ(2) or α(3)β(3)γ(2) GABA(A) channels, transiently transfected HEK293T cells expressing α(1)β(3)γ(2) GABA(A) channels, and immortalized cultures of human airway smooth muscle cells endogenously expressing GABA(A) channels. Current measurements were successfully studied in multiple cell types with multiple modes of channel expression in response to several classic GABA(A) channel agonists, antagonists, and allosteric modulators. We obtained success rates above 95% for transiently or stably transfected HEK cells and frozen RTA HEK cells expressing GABA(A) channels. Tissue-derived immortalized cultures of airway smooth muscle cells exhibited a slightly lower recording success rate of 75% using automated patch, which was much higher than the 5% success rate using manual patch clamp technique by the same research group. Responses to agonists, antagonists, and allosteric modulators compared well to previously reported manual patch results. The data demonstrate that both the biophysics and pharmacologic characterization of GABA(A) channels in a wide variety of cell formats can be performed using this automated patch clamp system.
Collapse
Affiliation(s)
- Qin Chen
- Fluxion Biosciences, South San Francisco, California, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Spencer CI, Li N, Chen Q, Johnson J, Nevill T, Kammonen J, Ionescu-Zanetti C. Ion channel pharmacology under flow: automation via well-plate microfluidics. Assay Drug Dev Technol 2012; 10:313-24. [PMID: 22574656 DOI: 10.1089/adt.2011.414] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Automated patch clamping addresses the need for high-throughput screening of chemical entities that alter ion channel function. As a result, there is considerable utility in the pharmaceutical screening arena for novel platforms that can produce relevant data both rapidly and consistently. Here we present results that were obtained with an innovative microfluidic automated patch clamp system utilizing a well-plate that eliminates the necessity of internal robotic liquid handling. Continuous recording from cell ensembles, rapid solution switching, and a bench-top footprint enable a number of assay formats previously inaccessible to automated systems. An electro-pneumatic interface was employed to drive the laminar flow of solutions in a microfluidic network that delivered cells in suspension to ensemble recording sites. Whole-cell voltage clamp was applied to linear arrays of 20 cells in parallel utilizing a 64-channel voltage clamp amplifier. A number of unique assays requiring sequential compound applications separated by a second or less, such as rapid determination of the agonist EC(50) for a ligand-gated ion channel or the kinetics of desensitization recovery, are enabled by the system. In addition, the system was validated via electrophysiological characterizations of both voltage-gated and ligand-gated ion channel targets: hK(V)2.1 and human Ether-à-go-go-related gene potassium channels, hNa(V)1.7 and 1.8 sodium channels, and (α1) hGABA(A) and (α1) human nicotinic acetylcholine receptor receptors. Our results show that the voltage dependence, kinetics, and interactions of these channels with pharmacological agents were matched to reference data. The results from these IonFlux™ experiments demonstrate that the system provides high-throughput automated electrophysiology with enhanced reliability and consistency, in a user-friendly format.
Collapse
Affiliation(s)
- C Ian Spencer
- Fluxion Biosciences, Inc., South San Francisco, California 94080, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Polonchuk L. Toward a New Gold Standard for Early Safety: Automated Temperature-Controlled hERG Test on the PatchLiner. Front Pharmacol 2012; 3:3. [PMID: 22303293 PMCID: PMC3266667 DOI: 10.3389/fphar.2012.00003] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 01/06/2012] [Indexed: 11/13/2022] Open
Abstract
The Patchliner® temperature-controlled automated patch clamp system was evaluated for testing drug effects on potassium currents through human ether-à-go-go related gene (hERG) channels expressed in Chinese hamster ovary cells at 35-37°C. IC(50) values for a set of reference drugs were compared with those obtained using the conventional voltage clamp technique. The results showed good correlation between the data obtained using automated and conventional electrophysiology. Based on these results, the Patchliner(®) represents an innovative automated electrophysiology platform for conducting the hERG assay that substantially increases throughput and has the advantage of operating at physiological temperature. It allows fast, accurate, and direct assessment of channel function to identify potential proarrhythmic side effects and sets a new standard in ion channel research for drug safety testing.
Collapse
Affiliation(s)
- Liudmila Polonchuk
- Non-Clinical Safety, Pharma Research, F. Hoffmann-La Roche Ltd. Basel, Switzerland
| |
Collapse
|
31
|
Möller C, Witchel H. Automated electrophysiology makes the pace for cardiac ion channel safety screening. Front Pharmacol 2011; 2:73. [PMID: 22131974 PMCID: PMC3222877 DOI: 10.3389/fphar.2011.00073] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Accepted: 11/06/2011] [Indexed: 01/14/2023] Open
Abstract
The field of automated patch-clamp electrophysiology has emerged from the tension between the pharmaceutical industry’s need for high-throughput compound screening versus its need to be conservative due to regulatory requirements. On the one hand, hERG channel screening was increasingly requested for new chemical entities, as the correlation between blockade of the ion channel coded by hERG and torsades de pointes cardiac arrhythmia gained increasing attention. On the other hand, manual patch-clamping, typically quoted as the “gold-standard” for understanding ion channel function and modulation, was far too slow (and, consequently, too expensive) for keeping pace with the numbers of compounds submitted for hERG channel investigations from pharmaceutical R&D departments. In consequence it became more common for some pharmaceutical companies to outsource safety pharmacological investigations, with a focus on hERG channel interactions. This outsourcing has allowed those pharmaceutical companies to build up operational flexibility and greater independence from internal resources, and allowed them to obtain access to the latest technological developments that emerged in automated patch-clamp electrophysiology – much of which arose in specialized biotech companies. Assays for nearly all major cardiac ion channels are now available by automated patch-clamping using heterologous expression systems, and recently, automated action potential recordings from stem-cell derived cardiomyocytes have been demonstrated. Today, most of the large pharmaceutical companies have acquired automated electrophysiology robots and have established various automated cardiac ion channel safety screening assays on these, in addition to outsourcing parts of their needs for safety screening.
Collapse
Affiliation(s)
- Clemens Möller
- InViTe Research Institute, Albstadt-Sigmaringen University Sigmaringen, Germany
| | | |
Collapse
|