1
|
Aghavali R, Roberts EG, Kurokawa YK, Mak E, Chan MYC, Wong AOT, Li RA, Costa KD. Enhanced drug classification using machine learning with multiplexed cardiac contractility assays. Pharmacol Res 2024; 209:107459. [PMID: 39396765 DOI: 10.1016/j.phrs.2024.107459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 09/04/2024] [Accepted: 10/08/2024] [Indexed: 10/15/2024]
Abstract
Cardiac screening of newly discovered drugs remains a longstanding challenge for the pharmaceutical industry. While therapeutic efficacy and cardiotoxicity are evaluated through preclinical biochemical and animal testing, 90 % of lead compounds fail to meet safety and efficacy benchmarks during human clinical trials. A preclinical model more representative of the human cardiac response is needed; heart tissue engineered from human pluripotent stem cell derived cardiomyocytes offers such a platform. In this study, three functionally distinct and independently validated engineered cardiac tissue assays are exposed to increasing concentrations of known compounds representing 5 classes of mechanistic action, creating a robust electrophysiology and contractility dataset. Combining results from six individual models, the resulting ensemble algorithm can classify the mechanistic action of unknown compounds with 86.2 % predictive accuracy. This outperforms single-assay models and offers a strategy to enhance future clinical trial success aligned with the recent FDA Modernization Act 2.0.
Collapse
Affiliation(s)
- Reza Aghavali
- Novoheart, Medera Inc., 6 Tide St., Boston, MA 02210, USA.
| | - Erin G Roberts
- Novoheart, Medera Inc., 6 Tide St., Boston, MA 02210, USA.
| | | | - Erica Mak
- Novoheart, Medera Inc., 6 Tide St., Boston, MA 02210, USA.
| | | | - Andy O T Wong
- Novoheart, Medera Inc., 6 Tide St., Boston, MA 02210, USA.
| | - Ronald A Li
- Novoheart, Medera Inc., 6 Tide St., Boston, MA 02210, USA.
| | - Kevin D Costa
- Novoheart, Medera Inc., 6 Tide St., Boston, MA 02210, USA.
| |
Collapse
|
2
|
Lee SG, Song GE, Seok J, Kim J, Kim MW, Rhee J, Park S, Jeong KS, Lee S, Lee YH, Jeong Y, Chung HM, Kim CY. Evaluation of the cardiotoxicity potential of bisphenol analogues in human induced pluripotent stem cells derived cardiomyocytes. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 272:116108. [PMID: 38364764 DOI: 10.1016/j.ecoenv.2024.116108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 01/31/2024] [Accepted: 02/09/2024] [Indexed: 02/18/2024]
Abstract
The importance of evaluating the cardiotoxicity potential of common chemicals as well as new drugs is increasing as a result of the development of animal alternative test methods using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM). Bisphenol A (BPA), which is used as a main material in plastics, is known as an endocrine-disrupting chemical, and recently reported to cause cardiotoxicity through inhibition of ion channels in CMs even with acute exposure. Accordingly, the need for the development of alternatives to BPA has been highlighted, and structural analogues including bisphenol AF, C, E, F, and S have been developed. However, cardiotoxicity data for analogues of bisphenol are not well known. In this study, in order to evaluate the cardiotoxicity potential of analogues, including BPA, a survival test of hiPSC-CMs and a dual-cardiotoxicity evaluation based on a multi-electrode array were performed. Acute exposure to all bisphenol analogues did not affect survival rate, but spike amplitude, beat period, and field potential duration were decreased in a dose-dependent manner in most of the bisphenols except bisphenol S. In addition, bisphenols, except for bisphenol S, reduced the contractile force of hiPSC-CMs and resulted in beating arrest at high doses. Taken together, it can be suggested that the developed bisphenol analogues could cause cardiotoxicity even with acute exposure, and it is considered that the application of the MEA-based dual-cardiotoxicity evaluation method can be an effective help in the development of safe alternatives.
Collapse
Affiliation(s)
- Seul-Gi Lee
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, Seoul 05029, Republic of Korea; College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Gyeong-Eun Song
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Jin Seok
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Jin Kim
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Min Woo Kim
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Jooeon Rhee
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Shinhye Park
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Kyu Sik Jeong
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Suemin Lee
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Yun Hyeong Lee
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Youngin Jeong
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Hyung Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, Seoul 05029, Republic of Korea; Miraecell Bio Co. Ltd., Seoul 04795, Republic of Korea
| | - C-Yoon Kim
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea.
| |
Collapse
|
3
|
Teixeira-Fonseca JL, Santos-Miranda A, Marques ILS, Marques LP, Alcantara F, de Lima Conceição MR, Souza DS, Santana Gondim AN, Roman-Campos D. Eugenol delays the onset of ouabain-induced ventricular cardiac arrhythmias in guinea pigs. Basic Clin Pharmacol Toxicol 2023; 133:565-575. [PMID: 37675641 DOI: 10.1111/bcpt.13941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 08/22/2023] [Accepted: 09/01/2023] [Indexed: 09/08/2023]
Abstract
Eugenol is an aromatic compound used in the manufacture of medicines, perfumes, cosmetics and as an anaesthetic due to the ability of the drug to block the neuronal isoform of voltage-gated Na+ channels (NaV ). Some arrhythmias are associated with gain of function in the sodium current (INa ) found in cardiomyocytes, and antiarrhythmic sodium channel blockers are commonly used in the clinical practice. This study sought to elucidate the potential mechanisms of eugenol's protection in the arrhythmic model of ouabain-induced arrhythmias in guinea pig heart. Ex vivo arrhythmias were induced using 50 μM of ouabain. The antiarrhythmic properties of eugenol were evaluated in the ex vivo heart preparation and isolated ventricular cardiomyocytes. The compound's effects on cardiac sodium current and action potential using the patch-clamp technique were evaluated. In all, eugenol decreased the ex vivo cardiac arrhythmias induced by ouabain. Furthermore, eugenol showed concentration dependent effect upon peak INa , left-shifted the stationary inactivation curve and delayed the recovery from inactivation of the INa . All these aspects are considered to be antiarrhythmic. Our findings demonstrate that eugenol has antiarrhythmic activity, which may be partially explained by the ability of eugenol to change de biophysical properties of INa of cardiomyocytes.
Collapse
Affiliation(s)
- Jorge Lucas Teixeira-Fonseca
- Laboratório de Cardiobiologia, Departamento de Biofísica, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Artur Santos-Miranda
- Departamento de Fisiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Leisiane Pereira Marques
- Laboratório de Cardiobiologia, Departamento de Biofísica, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Fabiana Alcantara
- Laboratório de Cardiobiologia, Departamento de Biofísica, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Michael Ramon de Lima Conceição
- Laboratório de Cardiobiologia, Departamento de Biofísica, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Diego Santos Souza
- Laboratório de Cardiobiologia, Departamento de Biofísica, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Antonio Nei Santana Gondim
- Laboratório de Biofísica e Farmacologia do Coração, Departamento de Educação (Campus-XII), Universidade do Estado da Bahia (UNEB), Guanambi, Brazil
| | - Danilo Roman-Campos
- Laboratório de Cardiobiologia, Departamento de Biofísica, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| |
Collapse
|
4
|
Lee J, Gänswein T, Ulusan H, Emmenegger V, Saguner AM, Duru F, Hierlemann A. Repeated and On-Demand Intracellular Recordings of Cardiomyocytes Derived from Human-Induced Pluripotent Stem Cells. ACS Sens 2022; 7:3181-3191. [PMID: 36166837 PMCID: PMC7613763 DOI: 10.1021/acssensors.2c01678] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Pharmaceutical compounds may have cardiotoxic properties, triggering potentially life-threatening arrhythmias. To investigate proarrhythmic effects of drugs, the patch clamp technique has been used as the gold standard for characterizing the electrophysiology of cardiomyocytes in vitro. However, the applicability of this technology for drug screening is limited, as it is complex to use and features low throughput. Recent studies have demonstrated that 3D-nanostructured electrodes enable to obtain intracellular signals from many cardiomyocytes in parallel; however, the tedious electrode fabrication and limited measurement duration still remain major issues for cardiotoxicity testing. Here, we demonstrate how porous Pt-black electrodes, arranged in high-density microelectrode arrays, can be used to record intracellular-like signals of cardiomyocytes at large scale repeatedly over an extended period of time. The developed technique, which yields highly parallelized electroporations using stimulation voltages around 1 V peak-to-peak amplitude, enabled intracellular-like recordings at high success rates without causing significant alteration in key electrophysiological features. In a proof-of-concept study, we investigated electrophysiological modulations induced by two clinically applied drugs, nifedipine and quinidine. As the obtained results were in good agreement with previously published data, we are confident that the developed technique has the potential to be routinely used in in vitro platforms for cardiotoxicity screening.
Collapse
Affiliation(s)
- Jihyun Lee
- Corresponding Authors Jihyun Lee — Bio Engineering Laboratory, ETH Zurich, 4058 Basel, Switzerland; ® Phone: +41 (0)61 387 31 28; jihyun.lee@ bsse.ethz.ch; Andreas Hierlemann — Bio Engineering Laboratory, ETH Zurich, 4058 Basel, Switzerland; Phone: +41 (0)61 387 31 50;
| | | | | | | | | | | | - Andreas Hierlemann
- Corresponding Authors Jihyun Lee — Bio Engineering Laboratory, ETH Zurich, 4058 Basel, Switzerland; ® Phone: +41 (0)61 387 31 28; jihyun.lee@ bsse.ethz.ch; Andreas Hierlemann — Bio Engineering Laboratory, ETH Zurich, 4058 Basel, Switzerland; Phone: +41 (0)61 387 31 50;
| |
Collapse
|
5
|
Abstract
Purpose of Review The advent of induced pluripotent stem cells (iPSC) has paved the way for new in vitro models of human cardiomyopathy. Herein, we will review existing models of disease as well as strengths and limitations of the system. Recent Findings Preclinical studies have now demonstrated that iPSCs generated from patients with both acquired or heritable genetic diseases retain properties of the disease in vitro and can be used as a model to study novel therapeutics. iPSCs can be differentiated in vitro into the cardiomyocyte lineage into cells resembling adult ventricular myocytes that retain properties of cardiovascular disease from their respective donor. iPSC pluripotency allows for them to be frozen, stored, and continually used to generate iPSC-derived myocytes for future experiments without need for invasive procedures or repeat myocyte isolations to obtain animal or human cardiac tissues. Summary While not without their limitations, iPSC models offer new ways for studying patient-specific cardiomyopathies. iPSCs offer a high-throughput avenue for drug development, modeling of disease pathophysiology in vitro, and enabling experimental repair strategies without need for invasive procedures to obtain cardiac tissues.
Collapse
|
6
|
Lee SJ, Kim HA, Kim SJ, Lee HA. Improving Generation of Cardiac Organoids from Human Pluripotent Stem Cells Using the Aurora Kinase Inhibitor ZM447439. Biomedicines 2021; 9:biomedicines9121952. [PMID: 34944767 PMCID: PMC8698385 DOI: 10.3390/biomedicines9121952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/16/2021] [Accepted: 12/19/2021] [Indexed: 01/21/2023] Open
Abstract
Drug-induced cardiotoxicity reduces the success rates of drug development. Thus, the limitations of current evaluation methods must be addressed. Human cardiac organoids (hCOs) derived from induced pluripotent stem cells (hiPSCs) are useful as an advanced drug-testing model; they demonstrate similar electrophysiological functionality and drug reactivity as the heart. How-ever, similar to other organoid models, they have immature characteristics compared to adult hearts, and exhibit batch-to-batch variation. As the cell cycle is important for the mesodermal differentiation of stem cells, we examined the effect of ZM447439, an aurora kinase inhibitor that regulates the cell cycle, on cardiogenic differentiation. We determined the optimal concentration and timing of ZM447439 for the differentiation of hCOs from hiPSCs and developed a novel protocol for efficiently and reproducibly generating beating hCOs with improved electrophysiological functionality, contractility, and yield. We validated their maturity through electro-physiological- and image-based functional assays and gene profiling with next-generation sequencing, and then applied these cells to multi-electrode array platforms to monitor the cardio-toxicity of drugs related to cardiac arrhythmia; the results confirmed the drug reactivity of hCOs. These findings may enable determination of the regulatory mechanism of cell cycles underlying the generation of iPSC-derived hCOs, providing a valuable drug testing platform.
Collapse
Affiliation(s)
- Su-Jin Lee
- Department of Predictive Toxicology, Korea Institute of Toxicology (KIT), Daejeon 34114, Korea; (S.-J.L.); (H.-A.K.)
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hyeon-A Kim
- Department of Predictive Toxicology, Korea Institute of Toxicology (KIT), Daejeon 34114, Korea; (S.-J.L.); (H.-A.K.)
| | - Sung-Joon Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
- Correspondence: (S.-J.K.); (H.-A.L.); Tel.: +82-2-740-8230 (S.-J.K.); +82-42-610-8093 (H.-A.L.)
| | - Hyang-Ae Lee
- Department of Predictive Toxicology, Korea Institute of Toxicology (KIT), Daejeon 34114, Korea; (S.-J.L.); (H.-A.K.)
- Correspondence: (S.-J.K.); (H.-A.L.); Tel.: +82-2-740-8230 (S.-J.K.); +82-42-610-8093 (H.-A.L.)
| |
Collapse
|
7
|
Amend N, Worek F, Thiermann H, Wille T. Investigation of cardiac glycosides from oleander in a human induced pluripotent stem cells derived cardiomyocyte model. Toxicol Lett 2021; 350:261-266. [PMID: 34371141 DOI: 10.1016/j.toxlet.2021.07.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/13/2021] [Accepted: 07/27/2021] [Indexed: 11/25/2022]
Abstract
The ingestion of Nerium oleander and Thevetia peruviana are common causes for poisoning in Southeast Asia. All parts of the oleander shrub contain cardiac glycosides of the cardenolide type. These glycosides act via inhibition of a Na+/K+-ATPase which might cause severe arrhythmia and subsequent death in oleander-poisoned patients. The current study uses human induced pluripotent stem cells derived cardiomyocytes (hiPSC-CM) in a microelectrode array (MEA) system to assess the cardiac effects of neriifolin, oleandrin, digitoxigenin, peruvoside and thevetin A from the oleander plant. Digoxin was used as established reference compound. All tested compounds showed a corrected field potential duration (FPDc) shortening and was the lowest for 600 nM digitoxigenin with -36.9 ± 1.2 %. Next to the dose-dependent pro-arrhythmic potential, a complete beat arrest of the spontaneously beating hiPSC-CM was observed at a concentration of 300 nM for neriifolin, 600 nM for oleandrin and 1000 nM for digitoxigenin and peruvoside. Thevetin A did not cause arrhythmia up to a final concentration of 1000 nM. Thus, it was possible to establish a cardiac effect rank order of the tested substances: neriifolin > oleandrin > digitoxigenin = peruvoside > digoxin > thevetin A.
Collapse
Affiliation(s)
- N Amend
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, München, Germany
| | - F Worek
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, München, Germany
| | - H Thiermann
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, München, Germany
| | - T Wille
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, München, Germany.
| |
Collapse
|
8
|
Lee SG, Kim J, Oh MS, Ryu B, Kang KR, Baek J, Lee JM, Choi SO, Kim CY, Chung HM. Development and validation of dual-cardiotoxicity evaluation method based on analysis of field potential and contractile force of human iPSC-derived cardiomyocytes / multielectrode assay platform. Biochem Biophys Res Commun 2021; 555:67-73. [PMID: 33813278 DOI: 10.1016/j.bbrc.2021.03.039] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 03/08/2021] [Indexed: 11/24/2022]
Abstract
A recent in vitro cardiovascular safety pharmacology test uses cardiomyocytes derived from human induced pluripotent stem cells (hiPSCs) to overcome the limitations of the classical test systems, such as species differences and local channel analysis. The Comprehensive in vitro Proarrhythmia Assay (CiPA) is a new proarrhythmia screening paradigm proposed by a CiPA steering expert group, which essentially requires iPSCs derived cardiomyocyte-based electrophysiological evaluation technology. Moreover, the measurement of the contractile force is also emerging as an important parameter to recapitulate non-proarrhythmic cardiotoxicity. Therefore, we constructed an multielectrode assay (MEA) evaluation method that can measure the electrophysiological changes with 6 reference drugs in hiPSC-derived cardiomyocytes. Subsequently, it was confirmed that the electrophysiological were changed in accordance with the mechanism of action of the drugs. Furthermore, based on the multi-probe impedance, we confirmed the decrease in contractile force due to treatment with drugs, and developed a platform to evaluate cardiotoxicity according to drugs along with field potential changes. Our excitation-contraction coupling cardiotoxicity assessment is considered to be more supportive in cardiac safety studies on pharmacologic sensitivity by complementing each assessment parameter.
Collapse
Affiliation(s)
- Seul-Gi Lee
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, Seoul, 143-701, Republic of Korea
| | - Jin Kim
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Min-Seok Oh
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, Seoul, 143-701, Republic of Korea
| | - Bokyeong Ryu
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Kyu-Ree Kang
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, Seoul, 143-701, Republic of Korea
| | - Jieun Baek
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, Seoul, 143-701, Republic of Korea
| | - Jin-Moo Lee
- Pharmacological Research Division, Toxicological Evaluation and Research Department, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Republic of Korea
| | - Sun-Ok Choi
- Pharmacological Research Division, Toxicological Evaluation and Research Department, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Republic of Korea
| | - C-Yoon Kim
- College of Veterinary Medicine, Konkuk University, Seoul, 05029, Republic of Korea.
| | - Hyung Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, Seoul, 143-701, Republic of Korea.
| |
Collapse
|
9
|
Fletcher S, Maddock H, James RS, Wallis R, Gharanei M. The cardiac work-loop technique: An in vitro model for identifying and profiling drug-induced changes in inotropy using rat papillary muscles. Sci Rep 2020; 10:5258. [PMID: 32210283 PMCID: PMC7093439 DOI: 10.1038/s41598-020-58935-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/26/2019] [Indexed: 11/09/2022] Open
Abstract
The cardiac work-loop technique closely mimics the intrinsic in vivo movement and characteristics of cardiac muscle function. In this study, six known inotropes were profiled using the work-loop technique to evaluate the potential of this method to predict inotropy. Papillary muscles from male Sprague-Dawley rats were mounted onto an organ bath perfused with Krebs-Henseleit buffer. Following optimisation, work-loop contractions were performed that included an initial stabilisation period followed by vehicle control or drug administration. Six known inotropes were tested: digoxin, dobutamine, isoprenaline, flecainide, verapamil and atenolol. Muscle performance was evaluated by calculating power output during work-loop contraction. Digoxin, dobutamine and isoprenaline caused a significant increase in power output of muscles when compared to vehicle control. Flecainide, verapamil and atenolol significantly reduced power output of muscles. These changes in power output were reflected in alterations in work loop shapes. This is the first study in which changes in work-loop shape detailing for example the activation, shortening or passive re-lengthening have been linked to the mechanism of action of a compound. This study has demonstrated that the work-loop technique can provide an important novel method with which to assess detailed mechanisms of drug-induced effects on cardiac muscle contractility.
Collapse
Affiliation(s)
- Sophie Fletcher
- Centre for Sport, Exercise and Life Sciences, Coventry University, Coventry, United Kingdom.,InoCardia Ltd, Technocentre, Puma Way, Coventry, CV1 2TT, UK
| | - Helen Maddock
- Centre for Sport, Exercise and Life Sciences, Coventry University, Coventry, United Kingdom. .,InoCardia Ltd, Technocentre, Puma Way, Coventry, CV1 2TT, UK.
| | - Rob S James
- Centre for Sport, Exercise and Life Sciences, Coventry University, Coventry, United Kingdom
| | - Rob Wallis
- InoCardia Ltd, Technocentre, Puma Way, Coventry, CV1 2TT, UK
| | - Mayel Gharanei
- Centre for Sport, Exercise and Life Sciences, Coventry University, Coventry, United Kingdom.,InoCardia Ltd, Technocentre, Puma Way, Coventry, CV1 2TT, UK
| |
Collapse
|
10
|
Nemade H, Acharya A, Chaudhari U, Nembo E, Nguemo F, Riet N, Abken H, Hescheler J, Papadopoulos S, Sachinidis A. Cyclooxygenases Inhibitors Efficiently Induce Cardiomyogenesis in Human Pluripotent Stem Cells. Cells 2020; 9:cells9030554. [PMID: 32120775 PMCID: PMC7140528 DOI: 10.3390/cells9030554] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 01/28/2020] [Accepted: 02/26/2020] [Indexed: 12/12/2022] Open
Abstract
Application of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) is limited by the challenges in their efficient differentiation. Recently, the Wingless (Wnt) signaling pathway has emerged as the key regulator of cardiomyogenesis. In this study, we evaluated the effects of cyclooxygenase inhibitors on cardiac differentiation of hPSCs. Cardiac differentiation was performed by adherent monolayer based method using 4 hPSC lines (HES3, H9, IMR90, and ES4SKIN). The efficiency of cardiac differentiation was evaluated by flow cytometry and RT-qPCR. Generated hPSC-CMs were characterised using immunocytochemistry, electrophysiology, electron microscopy, and calcium transient measurements. Our data show that the COX inhibitors Sulindac and Diclofenac in combination with CHIR99021 (GSK-3 inhibitor) efficiently induce cardiac differentiation of hPSCs. In addition, inhibition of COX using siRNAs targeted towards COX-1 and/or COX-2 showed that inhibition of COX-2 alone or COX-1 and COX-2 in combination induce cardiomyogenesis in hPSCs within 12 days. Using IMR90-Wnt reporter line, we showed that inhibition of COX-2 led to downregulation of Wnt signalling activity in hPSCs. In conclusion, this study demonstrates that COX inhibition efficiently induced cardiogenesis via modulation of COX and Wnt pathway and the generated cardiomyocytes express cardiac-specific structural markers as well as exhibit typical calcium transients and action potentials. These cardiomyocytes also responded to cardiotoxicants and can be relevant as an in vitro cardiotoxicity screening model.
Collapse
Affiliation(s)
- Harshal Nemade
- Institute of Neurophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany; (H.N.); (A.A.); (U.C.); (E.N.); (F.N.); (J.H.); (S.P.)
| | - Aviseka Acharya
- Institute of Neurophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany; (H.N.); (A.A.); (U.C.); (E.N.); (F.N.); (J.H.); (S.P.)
| | - Umesh Chaudhari
- Institute of Neurophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany; (H.N.); (A.A.); (U.C.); (E.N.); (F.N.); (J.H.); (S.P.)
| | - Erastus Nembo
- Institute of Neurophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany; (H.N.); (A.A.); (U.C.); (E.N.); (F.N.); (J.H.); (S.P.)
| | - Filomain Nguemo
- Institute of Neurophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany; (H.N.); (A.A.); (U.C.); (E.N.); (F.N.); (J.H.); (S.P.)
| | - Nicole Riet
- Department I Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne (UKK), Robert-Koch-Str. 21, 50931 Cologne, Germany;
| | - Hinrich Abken
- Regensburg Centre for Interventional Immunology (RCI), Deptartment Genetic Immunotherapy, University Hospital Regensburg, 93053 Regensburg, Germany;
| | - Jürgen Hescheler
- Institute of Neurophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany; (H.N.); (A.A.); (U.C.); (E.N.); (F.N.); (J.H.); (S.P.)
| | - Symeon Papadopoulos
- Institute of Neurophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany; (H.N.); (A.A.); (U.C.); (E.N.); (F.N.); (J.H.); (S.P.)
| | - Agapios Sachinidis
- Institute of Neurophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany; (H.N.); (A.A.); (U.C.); (E.N.); (F.N.); (J.H.); (S.P.)
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 21, 50931 Cologne, Germany
- Correspondence: ; Tel.: +49-0221-4787373
| |
Collapse
|
11
|
Garg P, Garg V, Shrestha R, Sanguinetti MC, Kamp TJ, Wu JC. Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes as Models for Cardiac Channelopathies: A Primer for Non-Electrophysiologists. Circ Res 2019; 123:224-243. [PMID: 29976690 DOI: 10.1161/circresaha.118.311209] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Life threatening ventricular arrhythmias leading to sudden cardiac death are a major cause of morbidity and mortality. In the absence of structural heart disease, these arrhythmias, especially in the younger population, are often an outcome of genetic defects in specialized membrane proteins called ion channels. In the heart, exceptionally well-orchestrated activity of a diversity of ion channels mediates the cardiac action potential. Alterations in either the function or expression of these channels can disrupt the configuration of the action potential, leading to abnormal electrical activity of the heart that can sometimes initiate an arrhythmia. Understanding the pathophysiology of inherited arrhythmias can be challenging because of the complexity of the disorder and lack of appropriate cellular and in vivo models. Recent advances in human induced pluripotent stem cell technology have provided remarkable progress in comprehending the underlying mechanisms of ion channel disorders or channelopathies by modeling these complex arrhythmia syndromes in vitro in a dish. To fully realize the potential of induced pluripotent stem cells in elucidating the mechanistic basis and complex pathophysiology of channelopathies, it is crucial to have a basic knowledge of cardiac myocyte electrophysiology. In this review, we will discuss the role of the various ion channels in cardiac electrophysiology and the molecular and cellular mechanisms of arrhythmias, highlighting the promise of human induced pluripotent stem cell-cardiomyocytes as a model for investigating inherited arrhythmia syndromes and testing antiarrhythmic strategies. Overall, this review aims to provide a basic understanding of the electrical activity of the heart and related channelopathies, especially to clinicians or research scientists in the cardiovascular field with limited electrophysiology background.
Collapse
Affiliation(s)
- Priyanka Garg
- From the Stanford Cardiovascular Institute (P.G., R.S., J.C.W.).,Department of Medicine, Division of Cardiology (P.G., R.S., J.C.W.).,Institute for Stem Cell Biology and Regenerative Medicine (P.G., R.S., J.C.W.)
| | - Vivek Garg
- Stanford University School of Medicine, CA; Department of Physiology, University of California San Francisco (V.G.)
| | - Rajani Shrestha
- From the Stanford Cardiovascular Institute (P.G., R.S., J.C.W.).,Department of Medicine, Division of Cardiology (P.G., R.S., J.C.W.).,Institute for Stem Cell Biology and Regenerative Medicine (P.G., R.S., J.C.W.)
| | | | - Timothy J Kamp
- Department of Medicine, University of Wisconsin-Madison (T.J.K.)
| | - Joseph C Wu
- From the Stanford Cardiovascular Institute (P.G., R.S., J.C.W.) .,Department of Medicine, Division of Cardiology (P.G., R.S., J.C.W.).,Institute for Stem Cell Biology and Regenerative Medicine (P.G., R.S., J.C.W.)
| |
Collapse
|
12
|
van Meer BJ, Krotenberg A, Sala L, Davis RP, Eschenhagen T, Denning C, Tertoolen LGJ, Mummery CL. Simultaneous measurement of excitation-contraction coupling parameters identifies mechanisms underlying contractile responses of hiPSC-derived cardiomyocytes. Nat Commun 2019; 10:4325. [PMID: 31541103 PMCID: PMC6754438 DOI: 10.1038/s41467-019-12354-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 09/04/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiomyocytes from human induced pluripotent stem cells (hiPSC-CMs) are increasingly recognized as valuable for determining the effects of drugs on ion channels but they do not always accurately predict contractile responses of the human heart. This is in part attributable to their immaturity but the sensitivity of measurement tools may also be limiting. Measuring action potential, calcium flux or contraction individually misses critical information that is captured when interrogating the complete excitation-contraction coupling cascade simultaneously. Here, we develop an hypothesis-based statistical algorithm that identifies mechanisms of action. We design and build a high-speed optical system to measure action potential, cytosolic calcium and contraction simultaneously using fluorescent sensors. These measurements are automatically processed, quantified and then assessed by the algorithm. Multiplexing these three critical physical features of hiPSC-CMs allows identification of all major drug classes affecting contractility with detection sensitivities higher than individual measurement of action potential, cytosolic calcium or contraction.
Collapse
Affiliation(s)
- Berend J van Meer
- Dept. of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZD, Leiden, The Netherlands
| | - Ana Krotenberg
- Dept. of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZD, Leiden, The Netherlands
| | - Luca Sala
- Dept. of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZD, Leiden, The Netherlands.,Istituto Auxologico Italiano, IRCCS, Center for Cardiac Arrhythmias of Genetic Origin, Laboratory of Cardiovascular Genetics, Via Zucchi 18, 20095, Cusano Milanino, MI, Italy
| | - Richard P Davis
- Dept. of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZD, Leiden, The Netherlands
| | - Thomas Eschenhagen
- Dept. of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Chris Denning
- Dept. of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Leon G J Tertoolen
- Dept. of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZD, Leiden, The Netherlands
| | - Christine L Mummery
- Dept. of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZD, Leiden, The Netherlands. .,Dept. of Applied Stem Cell Technologies, University of Twente, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands.
| |
Collapse
|
13
|
Guth BD, Engwall M, Eldridge S, Foley CM, Guo L, Gintant G, Koerner J, Parish ST, Pierson JB, Ribeiro AJS, Zabka T, Chaudhary KW, Kanda Y, Berridge B. Considerations for an In Vitro, Cell-Based Testing Platform for Detection of Adverse Drug-Induced Inotropic Effects in Early Drug Development. Part 1: General Considerations for Development of Novel Testing Platforms. Front Pharmacol 2019; 10:884. [PMID: 31447679 PMCID: PMC6697071 DOI: 10.3389/fphar.2019.00884] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 07/15/2019] [Indexed: 01/10/2023] Open
Abstract
Drug-induced effects on cardiac contractility can be assessed through the measurement of the maximal rate of pressure increase in the left ventricle (LVdP/dtmax) in conscious animals, and such studies are often conducted at the late stage of preclinical drug development. Detection of such effects earlier in drug research using simpler, in vitro test systems would be a valuable addition to our strategies for identifying the best possible drug development candidates. Thus, testing platforms with reasonably high throughput, and affordable costs would be helpful for early screening purposes. There may also be utility for testing platforms that provide mechanistic information about how a given drug affects cardiac contractility. Finally, there could be in vitro testing platforms that could ultimately contribute to the regulatory safety package of a new drug. The characteristics needed for a successful cell or tissue-based testing platform for cardiac contractility will be dictated by its intended use. In this article, general considerations are presented with the intent of guiding the development of new testing platforms that will find utility in drug research and development. In the following article (part 2), specific aspects of using human-induced stem cell-derived cardiomyocytes for this purpose are addressed.
Collapse
Affiliation(s)
- Brian D Guth
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany.,PreClinical Drug Development Platform (PCDDP), North-West University, Potchefstroom, South Africa
| | - Michael Engwall
- Safety Pharmacology and Animal Research Center, Amgen Research, Thousand Oaks, CA, United States
| | - Sandy Eldridge
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - C Michael Foley
- Department of Integrative Pharmacology, Integrated Sciences and Technology, AbbVie, North Chicago, IL, United States
| | - Liang Guo
- Laboratory of Investigative Toxicology, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | - Gary Gintant
- Department of Integrative Pharmacology, Integrated Sciences and Technology, AbbVie, North Chicago, IL, United States
| | - John Koerner
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Stanley T Parish
- Health and Environmental Sciences Institute, Washington, DC, United States
| | - Jennifer B Pierson
- Health and Environmental Sciences Institute, Washington, DC, United States
| | - Alexandre J S Ribeiro
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translation Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Tanja Zabka
- Department of Safety Assessment, Genentech, South San Francisco, CA, United States
| | - Khuram W Chaudhary
- Global Safety Pharmacology, GlaxoSmithKline plc, Collegeville, PA, United States
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, Kanagawa, Japan
| | - Brian Berridge
- National Toxicology Program, National Institute of Environmental Health Sciences, Durham, NC, United States
| |
Collapse
|
14
|
Kim TW, Che JH, Yun JW. Use of stem cells as alternative methods to animal experimentation in predictive toxicology. Regul Toxicol Pharmacol 2019; 105:15-29. [DOI: 10.1016/j.yrtph.2019.03.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 03/23/2019] [Accepted: 03/25/2019] [Indexed: 12/16/2022]
|
15
|
Electrophysiological characteristics and pharmacological sensitivity of two lines of human induced pluripotent stem cell derived cardiomyocytes coming from two different suppliers. J Pharmacol Toxicol Methods 2018; 90:58-66. [DOI: 10.1016/j.vascn.2017.12.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 11/21/2017] [Accepted: 12/18/2017] [Indexed: 01/08/2023]
|
16
|
Rehnelt S, Malan D, Juhasz K, Wolters B, Doerr L, Beckler M, Kettenhofen R, Bohlen H, Bruegmann T, Sasse P. Frequency-Dependent Multi-Well Cardiotoxicity Screening Enabled by Optogenetic Stimulation. Int J Mol Sci 2017; 18:E2634. [PMID: 29211031 PMCID: PMC5751237 DOI: 10.3390/ijms18122634] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 11/28/2017] [Accepted: 11/30/2017] [Indexed: 11/17/2022] Open
Abstract
Side effects on cardiac ion channels causing lethal arrhythmias are one major reason for drug withdrawals from the market. Field potential (FP) recording from cardiomyocytes, is a well-suited tool to assess such cardiotoxic effects of drug candidates in preclinical drug development, but it is currently limited to the spontaneous beating of the cardiomyocytes and manual analysis. Herein, we present a novel optogenetic cardiotoxicity screening system suited for the parallel automated frequency-dependent analysis of drug effects on FP recorded from human-induced pluripotent stem cell-derived cardiomyocytes. For the expression of the light-sensitive cation channel Channelrhodopsin-2, we optimised protocols using virus transduction or transient mRNA transfection. Optical stimulation was performed with a new light-emitting diode lid for a 96-well FP recording system. This enabled reliable pacing at physiologically relevant heart rates and robust recording of FP. Thereby we detected rate-dependent effects of drugs on Na⁺, Ca2+ and K⁺ channel function indicated by FP prolongation, FP shortening and the slowing of the FP downstroke component, as well as generation of afterdepolarisations. Taken together, we present a scalable approach for preclinical frequency-dependent screening of drug effects on cardiac electrophysiology. Importantly, we show that the recording and analysis can be fully automated and the technology is readily available using commercial products.
Collapse
Affiliation(s)
- Susanne Rehnelt
- Institute of Physiology I, Medical Faculty, University of Bonn, 53127 Bonn, Germany.
| | - Daniela Malan
- Institute of Physiology I, Medical Faculty, University of Bonn, 53127 Bonn, Germany.
| | - Krisztina Juhasz
- Nanion Technologies GmbH, 80636 Munich, Germany.
- Present address: Institute for Nanoelectronics, Department of Electrical Engineering and Information Technology, Technische Universität München, 80339 Munich, Germany.
| | - Benjamin Wolters
- Part of the Ncardia Group, Axiogenesis AG, 50829 Cologne, Germany.
| | - Leo Doerr
- Nanion Technologies GmbH, 80636 Munich, Germany.
| | | | - Ralf Kettenhofen
- Part of the Ncardia Group, Axiogenesis AG, 50829 Cologne, Germany.
| | - Heribert Bohlen
- Part of the Ncardia Group, Axiogenesis AG, 50829 Cologne, Germany.
| | - Tobias Bruegmann
- Institute of Physiology I, Medical Faculty, University of Bonn, 53127 Bonn, Germany.
- Research Training Group 1873, University of Bonn, 53127 Bonn, Germany.
| | - Philipp Sasse
- Institute of Physiology I, Medical Faculty, University of Bonn, 53127 Bonn, Germany.
| |
Collapse
|
17
|
Lee EK, Tran DD, Keung W, Chan P, Wong G, Chan CW, Costa KD, Li RA, Khine M. Machine Learning of Human Pluripotent Stem Cell-Derived Engineered Cardiac Tissue Contractility for Automated Drug Classification. Stem Cell Reports 2017; 9:1560-1572. [PMID: 29033305 PMCID: PMC5829317 DOI: 10.1016/j.stemcr.2017.09.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 09/11/2017] [Accepted: 09/12/2017] [Indexed: 01/07/2023] Open
Abstract
Accurately predicting cardioactive effects of new molecular entities for therapeutics remains a daunting challenge. Immense research effort has been focused toward creating new screening platforms that utilize human pluripotent stem cell (hPSC)-derived cardiomyocytes and three-dimensional engineered cardiac tissue constructs to better recapitulate human heart function and drug responses. As these new platforms become increasingly sophisticated and high throughput, the drug screens result in larger multidimensional datasets. Improved automated analysis methods must therefore be developed in parallel to fully comprehend the cellular response across a multidimensional parameter space. Here, we describe the use of machine learning to comprehensively analyze 17 functional parameters derived from force readouts of hPSC-derived ventricular cardiac tissue strips (hvCTS) electrically paced at a range of frequencies and exposed to a library of compounds. A generated metric is effective for then determining the cardioactivity of a given drug. Furthermore, we demonstrate a classification model that can automatically predict the mechanistic action of an unknown cardioactive drug.
Collapse
Affiliation(s)
- Eugene K Lee
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Novoheart LTD, Shatin, Hong Kong
| | | | - Wendy Keung
- Dr. Li Dak-Sum Research Centre, The University of Hong Kong - Karolinska Institutet Collaboration in Regenerative Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong; Ming-Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Stockholm 17177, Sweden
| | - Patrick Chan
- Dr. Li Dak-Sum Research Centre, The University of Hong Kong - Karolinska Institutet Collaboration in Regenerative Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong; Ming-Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Stockholm 17177, Sweden
| | | | | | - Kevin D Costa
- Novoheart LTD, Shatin, Hong Kong; Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA
| | - Ronald A Li
- Novoheart LTD, Shatin, Hong Kong; Dr. Li Dak-Sum Research Centre, The University of Hong Kong - Karolinska Institutet Collaboration in Regenerative Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong; Ming-Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Stockholm 17177, Sweden
| | - Michelle Khine
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Novoheart LTD, Shatin, Hong Kong.
| |
Collapse
|
18
|
Nozaki Y, Honda Y, Watanabe H, Saiki S, Koyabu K, Itoh T, Nagasawa C, Nakamori C, Nakayama C, Iwasaki H, Suzuki S, Tanaka K, Takahashi E, Miyamoto K, Morimura K, Yamanishi A, Endo H, Shinozaki J, Nogawa H, Shinozawa T, Saito F, Kunimatsu T. CSAHi study-2: Validation of multi-electrode array systems (MEA60/2100) for prediction of drug-induced proarrhythmia using human iPS cell-derived cardiomyocytes: Assessment of reference compounds and comparison with non-clinical studies and clinical information. Regul Toxicol Pharmacol 2017. [PMID: 28634147 DOI: 10.1016/j.yrtph.2017.06.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
With the aim of reconsidering ICH S7B and E14 guidelines, a new in vitro assay system has been subjected to worldwide validation to establish a better prediction platform for potential drug-induced QT prolongation and the consequent TdP in clinical practice. In Japan, CSAHi HEART team has been working on hiPS-CMs in the MEA (hiPS-CMs/MEA) under a standardized protocol and found no inter-facility or lot-to-lot variability for proarrhythmic risk assessment of 7 reference compounds. In this study, we evaluated the responses of hiPS-CMs/MEA to another 31 reference compounds associated with cardiac toxicities, and gene expression to further clarify the electrophysiological characteristics over the course of culture period. The hiPS-CMs/MEA assay accurately predicted reference compounds potential for arrhythmogenesis, and yielded results that showed better correlation with target concentrations of QTc prolongation or TdP in clinical setting than other current in vitro and in vivo assays. Gene expression analyses revealed consistent profiles in all samples within and among the testing facilities. This report would provide CiPA with informative guidance on the use of the hiPS-CMs/MEA assay, and promote the establishment of a new paradigm, beyond conventional in vitro and in vivo assays for cardiac safety assessment of new drugs.
Collapse
Affiliation(s)
- Yumiko Nozaki
- Preclinical Research Laboratories, Sumitomo Dainippon Pharma Co., Ltd., 3-1-98 Kasugade-naka, Konohana-ku, Osaka 554-0022, Japan.
| | - Yayoi Honda
- Preclinical Research Laboratories, Sumitomo Dainippon Pharma Co., Ltd., 3-1-98 Kasugade-naka, Konohana-ku, Osaka 554-0022, Japan
| | - Hitoshi Watanabe
- Preclinical Research Laboratories, Sumitomo Dainippon Pharma Co., Ltd., 3-1-98 Kasugade-naka, Konohana-ku, Osaka 554-0022, Japan
| | - Shota Saiki
- Research Laboratory for Development, Shionogi & Co., Ltd., 3-1-1, Futaba-cho, Toyonaka, Osaka 561-0825, Japan; Consortium for Safety Assessment using Human iPS Cells (CSAHi), Japan
| | - Kiyotaka Koyabu
- Research Laboratory for Development, Shionogi & Co., Ltd., 3-1-1, Futaba-cho, Toyonaka, Osaka 561-0825, Japan
| | - Tetsuji Itoh
- Research Laboratory for Development, Shionogi & Co., Ltd., 3-1-1, Futaba-cho, Toyonaka, Osaka 561-0825, Japan
| | - Chiho Nagasawa
- Drug Safety, Taisho Pharmaceutical Co., Ltd., 1-403, Yoshino-cho, Kita-ku, Saitama-shi, Saitama 331-9530, Japan; Consortium for Safety Assessment using Human iPS Cells (CSAHi), Japan
| | - Chiaki Nakamori
- Drug Safety, Taisho Pharmaceutical Co., Ltd., 1-403, Yoshino-cho, Kita-ku, Saitama-shi, Saitama 331-9530, Japan
| | - Chiaki Nakayama
- Drug Safety, Taisho Pharmaceutical Co., Ltd., 1-403, Yoshino-cho, Kita-ku, Saitama-shi, Saitama 331-9530, Japan
| | - Hiroshi Iwasaki
- Drug Safety, Taisho Pharmaceutical Co., Ltd., 1-403, Yoshino-cho, Kita-ku, Saitama-shi, Saitama 331-9530, Japan
| | - Shinobu Suzuki
- Nippon Boehringer Ingelheim Co., Ltd., 6-7-5, Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Kohji Tanaka
- Nippon Boehringer Ingelheim Co., Ltd., 6-7-5, Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Etsushi Takahashi
- Research Laboratories, Toyama Chemical Co., Ltd., 4-1, Shimookui 2-chome, Toyama 930-8508, Japan; Consortium for Safety Assessment using Human iPS Cells (CSAHi), Japan
| | - Kaori Miyamoto
- Research Laboratories, Toyama Chemical Co., Ltd., 4-1, Shimookui 2-chome, Toyama 930-8508, Japan
| | - Kaoru Morimura
- Research Laboratories, Toyama Chemical Co., Ltd., 4-1, Shimookui 2-chome, Toyama 930-8508, Japan
| | - Atsuhiro Yamanishi
- Toxicology Research Laboratory, Kyorin Pharmaceutical Co., Ltd., 1848, Nogi, Nogi-machi, Shimotsuga-gun, Tochigi 329-0114, Japan; Consortium for Safety Assessment using Human iPS Cells (CSAHi), Japan
| | - Hiroko Endo
- Toxicology Research Laboratory, Kyorin Pharmaceutical Co., Ltd., 1848, Nogi, Nogi-machi, Shimotsuga-gun, Tochigi 329-0114, Japan
| | - Junko Shinozaki
- Toxicology Research Laboratory, Kyorin Pharmaceutical Co., Ltd., 1848, Nogi, Nogi-machi, Shimotsuga-gun, Tochigi 329-0114, Japan
| | - Hisashi Nogawa
- Toxicology Research Laboratory, Kyorin Pharmaceutical Co., Ltd., 1848, Nogi, Nogi-machi, Shimotsuga-gun, Tochigi 329-0114, Japan
| | - Tadahiro Shinozawa
- Drug Safety Research Laboratories, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome Fujisawa, Kanagawa 251-8555, Japan; Consortium for Safety Assessment using Human iPS Cells (CSAHi), Japan; Japan Pharmaceutical Manufacturers Association, Drug Evaluation Committee, Non-Clinical Evaluation Expert Committee, Japan
| | - Fumiyo Saito
- Chemicals Assessment and Research Center, Chemicals Evaluation and Research Institute, Japan (CERI), 1600, Shimotakano, Sugito-machi, Kitakatsushika-gun, Saitama 345-0043, Japan; Consortium for Safety Assessment using Human iPS Cells (CSAHi), Japan
| | - Takeshi Kunimatsu
- Preclinical Research Laboratories, Sumitomo Dainippon Pharma Co., Ltd., 3-1-98 Kasugade-naka, Konohana-ku, Osaka 554-0022, Japan; Consortium for Safety Assessment using Human iPS Cells (CSAHi), Japan; Japan Pharmaceutical Manufacturers Association, Drug Evaluation Committee, Non-Clinical Evaluation Expert Committee, Japan.
| |
Collapse
|
19
|
Identification of Na+/K+-ATPase inhibition-independent proarrhythmic ionic mechanisms of cardiac glycosides. Sci Rep 2017; 7:2465. [PMID: 28550304 PMCID: PMC5446409 DOI: 10.1038/s41598-017-02496-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 04/13/2017] [Indexed: 12/17/2022] Open
Abstract
The current study explored the Na+/K+-ATPase (NKA) inhibition-independent proarrhythmic mechanisms of cardiac glycosides (CGs) which are well-known NKA inhibitors. With the cytosolic Ca2+ chelated by EGTA and BAPTA or extracellular Ca2+ replaced by Ba2+, effects of bufadienolides (bufalin (BF) and cinobufagin (CBG)) and cardenolides (ouabain (Oua) and pecilocerin A (PEA)) on the L-type calcium current (ICa,L) were recorded in heterologous expression Cav1.2-CHO cells and human embryonic stem cell-derived cardiomyocytes (hESC-CMs). BF and CBG demonstrated a concentration-dependent (0.1 to 100 µM) ICa,L inhibition (maximal ≥50%) without and with the NKA activity blocked by 10 µM Oua. BF significantly shortened the action potential duration at 1.0 µM and shortened the extracellular field potential duration at 0.01~1.0 µM. On the other hand, BF and CBG at 100 µM demonstrated a strong inhibition (≥40%) of the rapidly activating component of the delayed rectifier K+ current (IKr) in heterologous expression HEK293 cells and prolonged the APD of the heart of day-3 Zebrafish larva with disrupted rhythmic contractions. Moreover, hESC-CMs treated with BF (10 nM) for 24 hours showed moderate yet significant prolongation in APD90. In conclusion, our data indicate that CGs particularly bufadienolides possess cytosolic [Ca2+]i- and NKA inhibition- independent proarrhythmic potential through ICa,L and IKr inhibitions.
Collapse
|
20
|
Kitaguchi T, Moriyama Y, Taniguchi T, Maeda S, Ando H, Uda T, Otabe K, Oguchi M, Shimizu S, Saito H, Toratani A, Asayama M, Yamamoto W, Matsumoto E, Saji D, Ohnaka H, Miyamoto N. CSAHi study: Detection of drug-induced ion channel/receptor responses, QT prolongation, and arrhythmia using multi-electrode arrays in combination with human induced pluripotent stem cell-derived cardiomyocytes. J Pharmacol Toxicol Methods 2017; 85:73-81. [DOI: 10.1016/j.vascn.2017.02.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 12/05/2016] [Accepted: 02/01/2017] [Indexed: 12/20/2022]
|
21
|
Hyun SW, Kim BR, Hyun SA, Seo JW. The assessment of electrophysiological activity in human-induced pluripotent stem cell-derived cardiomyocytes exposed to dimethyl sulfoxide and ethanol by manual patch clamp and multi-electrode array system. J Pharmacol Toxicol Methods 2017; 87:93-98. [PMID: 28377112 DOI: 10.1016/j.vascn.2017.03.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 03/14/2017] [Accepted: 03/31/2017] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Recently, electrophysiological activity has been effectively measured in human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) to predict drug-induced arrhythmia. Dimethyl sulfoxide (DMSO) and ethanol have been used as diluting agents in many experiments. However, the maximum DMSO and ethanol concentrations that can be effectively used in the measurement of electrophysiological parameters in hiPSC-CMs-based patch clamp and multi-electrode array (MEA) have not been fully elucidated. METHODS We investigated the effects of varying concentrations of DMSO and ethanol used as diluting agents on several electrophysiological parameters in hiPSC-CMs using patch clamp and MEA. RESULTS Both DMSO and ethanol at concentrations>1% in external solution resulted in osmolality >400mOsmol/kg, but pH was not affected by either agent. Neither DMSO nor ethanol led to cell death at the concentrations examined. However, resting membrane potential, action potential amplitude, action potential duration at 90% and 40%, and corrected field potential duration were decreased significantly at 1% ethanol concentration. DMSO at 1% also significantly decreased the sodium spike amplitude. In addition, the waveform of action potential and field potential was recorded as irregular at 3% concentrations of both DMSO and ethanol. Concentrations of up to 0.3% of either agent did not affect osmolality, pH, cell death, or electrophysiological parameters in hiPSC-CMs. DISCUSSION Our findings suggest that 0.3% is the maximum concentration at which DMSO or ethanol should be used for dilution purposes in hiPSC-CMs-based patch clamp and MEA.
Collapse
Affiliation(s)
- Soo-Wang Hyun
- Research Group for Safety Pharmacology, Korea Institute of Toxicology, KRICT, 34114 Daejeon, Republic of Korea.
| | - Bo-Ram Kim
- Research Group for Safety Pharmacology, Korea Institute of Toxicology, KRICT, 34114 Daejeon, Republic of Korea.
| | - Sung-Ae Hyun
- Research Group for Safety Pharmacology, Korea Institute of Toxicology, KRICT, 34114 Daejeon, Republic of Korea.
| | - Joung-Wook Seo
- Research Group for Safety Pharmacology, Korea Institute of Toxicology, KRICT, 34114 Daejeon, Republic of Korea.
| |
Collapse
|
22
|
Dempsey GT, Chaudhary KW, Atwater N, Nguyen C, Brown BS, McNeish JD, Cohen AE, Kralj JM. Cardiotoxicity screening with simultaneous optogenetic pacing, voltage imaging and calcium imaging. J Pharmacol Toxicol Methods 2016; 81:240-50. [DOI: 10.1016/j.vascn.2016.05.003] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 05/02/2016] [Accepted: 05/09/2016] [Indexed: 12/23/2022]
|
23
|
Clements M. Multielectrode Array (MEA) Assay for Profiling Electrophysiological Drug Effects in Human Stem Cell-Derived Cardiomyocytes. CURRENT PROTOCOLS IN TOXICOLOGY 2016; 68:22.4.1-22.4.32. [PMID: 27145112 DOI: 10.1002/cptx.2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
More relevant and reliable preclinical cardiotoxicity tests are required to improve drug safety and reduce the cost of drug development. Human stem cell-derived cardiomyocytes (hSC-CMs) provide a potential model for the development of superior assays for preclinical drug safety screening. One such hSC-CM assay that has shown significant potential for enabling more predictive drug cardiac risk assessment is the MEA assay. The Multi-electrode Array (MEA) assay is an electrophysiology-based technique that uses microelectrodes embedded in the culture surface of each well to measure fluctuations in extracellular field potential (FP) generated from spontaneously beating hSC-CMs. Perturbations to the recorded FP waveform can be used as an unbiased method of predicting the identity of ion channel(s) impacted on drug exposure. Here, a higher throughput MEA assay using hSC-CMs in 48-well MEA plates is described for profiling compound-induced effects on cardiomyocyte electrophysiology. Techniques for preparing hSC-CM monolayers in MEA plates and methods to contextualize MEA assay experimental results are also covered. © 2016 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Mike Clements
- GE Healthcare Life Sciences, Maynard Centre, Forest Farm, Whitchurch, Cardiff, United Kingdom
| |
Collapse
|
24
|
Safety pharmacology studies using EFP and impedance. J Pharmacol Toxicol Methods 2016; 81:223-32. [PMID: 27084108 DOI: 10.1016/j.vascn.2016.04.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/01/2016] [Accepted: 04/05/2016] [Indexed: 12/17/2022]
Abstract
INTRODUCTION While extracellular field potential (EFP) recordings using multi-electrode arrays (MEAs) are a well-established technique for monitoring changes in cardiac and neuronal function, impedance is a relatively unexploited technology. The combination of EFP, impedance and human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) has important implications for safety pharmacology as functional information about contraction and field potentials can be gleaned from human cardiomyocytes in a beating monolayer. The main objectives of this study were to demonstrate, using a range of different compounds, that drug effects on contraction and electrophysiology can be detected using a beating monolayer of hiPSC-CMs on the CardioExcyte 96. METHODS hiPSC-CMs were grown as a monolayer on NSP-96 plates for the CardioExcyte 96 (Nanion Technologies) and recordings were made in combined EFP and impedance mode at physiological temperature. The effect of the hERG blockers, E4031 and dofetilide, hERG trafficking inhibitor, pentamidine, β-adrenergic receptor agonist, isoproterenol, and calcium channel blocker, nifedipine, was tested on the EFP and impedance signals. RESULTS Combined impedance and EFP measurements were made from hiPSC-CMs using the CardioExcyte 96 (Nanion Technologies). E4031 and dofetilide, known to cause arrhythmia and Torsades de Pointes (TdP) in humans, decreased beat rate in impedance and EFP modes. Early afterdepolarization (EAD)-like events, an in vitro marker of TdP, could also be detected using this system. Isoproterenol and nifedipine caused an increase in beat rate. A long-term study (over 30h) of pentamidine, a hERG trafficking inhibitor, showed a concentration and time-dependent effect of pentamidine. DISCUSSION In the light of the new Comprehensive in Vitro Proarrhythmia Assay (CiPA) initiative to improve guidelines and standardize assays and protocols, the use of EFP and impedance measurements from hiPSCs may become critical in determining the proarrhythmic risk of potential drug candidates. The combination of EFP offering information about cardiac electrophysiology, and impedance, providing information about contractility from the same area of a synchronously beating monolayer of human cardiomyocytes in a 96-well plate format has important implications for future cardiac safety testing.
Collapse
|
25
|
Evaluation of nefazodone-induced cardiotoxicity in human induced pluripotent stem cell-derived cardiomyocytes. Toxicol Appl Pharmacol 2016; 296:42-53. [DOI: 10.1016/j.taap.2016.01.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 01/12/2016] [Accepted: 01/21/2016] [Indexed: 01/10/2023]
|
26
|
Del Álamo JC, Lemons D, Serrano R, Savchenko A, Cerignoli F, Bodmer R, Mercola M. High throughput physiological screening of iPSC-derived cardiomyocytes for drug development. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1717-27. [PMID: 26952934 DOI: 10.1016/j.bbamcr.2016.03.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/01/2016] [Accepted: 03/03/2016] [Indexed: 12/25/2022]
Abstract
Cardiac drug discovery is hampered by the reliance on non-human animal and cellular models with inadequate throughput and physiological fidelity to accurately identify new targets and test novel therapeutic strategies. Similarly, adverse drug effects on the heart are challenging to model, contributing to costly failure of drugs during development and even after market launch. Human induced pluripotent stem cell derived cardiac tissue represents a potentially powerful means to model aspects of heart physiology relevant to disease and adverse drug effects, providing both the human context and throughput needed to improve the efficiency of drug development. Here we review emerging technologies for high throughput measurements of cardiomyocyte physiology, and comment on the promises and challenges of using iPSC-derived cardiomyocytes to model disease and introduce the human context into early stages of drug discovery. This article is part of a Special Issue entitled: Cardiomyocyte biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.
Collapse
Affiliation(s)
- Juan C Del Álamo
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, 9500 Gilman Drive MC 0411, La Jolla, CA 92093-0411, USA
| | - Derek Lemons
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive MC 0412, La Jolla, CA 92093-0412, USA; Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, CA 92037, USA
| | - Ricardo Serrano
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, 9500 Gilman Drive MC 0411, La Jolla, CA 92093-0411, USA
| | - Alex Savchenko
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive MC 0412, La Jolla, CA 92093-0412, USA; Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, CA 92037, USA; Stanford Cardiovascular Institute, 265 Campus Dr., Stanford, CA 94305-5454, USA
| | - Fabio Cerignoli
- ACEA Biosciences, Inc., 6779 Mesa Ridge Road, San Diego, CA 92121, USA
| | - Rolf Bodmer
- Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, CA 92037, USA
| | - Mark Mercola
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive MC 0412, La Jolla, CA 92093-0412, USA; Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, CA 92037, USA; Stanford Cardiovascular Institute, 265 Campus Dr., Stanford, CA 94305-5454, USA.
| |
Collapse
|
27
|
Maturing human pluripotent stem cell-derived cardiomyocytes in human engineered cardiac tissues. Adv Drug Deliv Rev 2016; 96:110-34. [PMID: 25956564 DOI: 10.1016/j.addr.2015.04.019] [Citation(s) in RCA: 174] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 04/24/2015] [Accepted: 04/25/2015] [Indexed: 12/19/2022]
Abstract
Engineering functional human cardiac tissue that mimics the native adult morphological and functional phenotype has been a long held objective. In the last 5 years, the field of cardiac tissue engineering has transitioned from cardiac tissues derived from various animal species to the production of the first generation of human engineered cardiac tissues (hECTs), due to recent advances in human stem cell biology. Despite this progress, the hECTs generated to date remain immature relative to the native adult myocardium. In this review, we focus on the maturation challenge in the context of hECTs, the present state of the art, and future perspectives in terms of regenerative medicine, drug discovery, preclinical safety testing and pathophysiological studies.
Collapse
|
28
|
Hunsberger JG, Efthymiou AG, Malik N, Behl M, Mead IL, Zeng X, Simeonov A, Rao M. Induced Pluripotent Stem Cell Models to Enable In Vitro Models for Screening in the Central Nervous System. Stem Cells Dev 2015; 24:1852-64. [PMID: 25794298 PMCID: PMC4533087 DOI: 10.1089/scd.2014.0531] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 03/20/2015] [Indexed: 12/23/2022] Open
Abstract
There is great need to develop more predictive drug discovery tools to identify new therapies to treat diseases of the central nervous system (CNS). Current nonpluripotent stem cell-based models often utilize non-CNS immortalized cell lines and do not enable the development of personalized models of disease. In this review, we discuss why in vitro models are necessary for translational research and outline the unique advantages of induced pluripotent stem cell (iPSC)-based models over those of current systems. We suggest that iPSC-based models can be patient specific and isogenic lines can be differentiated into many neural cell types for detailed comparisons. iPSC-derived cells can be combined to form small organoids, or large panels of lines can be developed that enable new forms of analysis. iPSC and embryonic stem cell-derived cells can be readily engineered to develop reporters for lineage studies or mechanism of action experiments further extending the utility of iPSC-based systems. We conclude by describing novel technologies that include strategies for the development of diversity panels, novel genomic engineering tools, new three-dimensional organoid systems, and modified high-content screens that may bring toxicology into the 21st century. The strategic integration of these technologies with the advantages of iPSC-derived cell technology, we believe, will be a paradigm shift for toxicology and drug discovery efforts.
Collapse
Affiliation(s)
| | | | - Nasir Malik
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland
| | - Mamta Behl
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Ivy L. Mead
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina
| | - Xianmin Zeng
- Buck Institute for Age Research, Novato, California
| | - Anton Simeonov
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland
| | - Mahendra Rao
- New York Stem Cell Foundation, New York, New York
| |
Collapse
|
29
|
Gilchrist KH, Lewis GF, Gay EA, Sellgren KL, Grego S. High-throughput cardiac safety evaluation and multi-parameter arrhythmia profiling of cardiomyocytes using microelectrode arrays. Toxicol Appl Pharmacol 2015; 288:249-57. [PMID: 26232523 DOI: 10.1016/j.taap.2015.07.024] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 07/16/2015] [Accepted: 07/24/2015] [Indexed: 12/21/2022]
Abstract
Microelectrode arrays (MEAs) recording extracellular field potentials of human-induced pluripotent stem cell-derived cardiomyocytes (hiPS-CM) provide a rich data set for functional assessment of drug response. The aim of this work is the development of a method for a systematic analysis of arrhythmia using MEAs, with emphasis on the development of six parameters accounting for different types of cardiomyocyte signal irregularities. We describe a software approach to carry out such analysis automatically including generation of a heat map that enables quick visualization of arrhythmic liability of compounds. We also implemented signal processing techniques for reliable extraction of the repolarization peak for field potential duration (FPD) measurement even from recordings with low signal to noise ratios. We measured hiPS-CM's on a 48 well MEA system with 5minute recordings at multiple time points (0.5, 1, 2 and 4h) after drug exposure. We evaluated concentration responses for seven compounds with a combination of hERG, QT and clinical proarrhythmia properties: Verapamil, Ranolazine, Flecainide, Amiodarone, Ouabain, Cisapride, and Terfenadine. The predictive utility of MEA parameters as surrogates of these clinical effects were examined. The beat rate and FPD results exhibited good correlations with previous MEA studies in stem cell derived cardiomyocytes and clinical data. The six-parameter arrhythmia assessment exhibited excellent predictive agreement with the known arrhythmogenic potential of the tested compounds, and holds promise as a new method to predict arrhythmic liability.
Collapse
Affiliation(s)
- Kristin H Gilchrist
- RTI International, 3040 E. Cornwallis Road, Research Triangle Park, NC 27709, USA.
| | - Gregory F Lewis
- RTI International, 3040 E. Cornwallis Road, Research Triangle Park, NC 27709, USA
| | - Elaine A Gay
- RTI International, 3040 E. Cornwallis Road, Research Triangle Park, NC 27709, USA
| | - Katelyn L Sellgren
- RTI International, 3040 E. Cornwallis Road, Research Triangle Park, NC 27709, USA
| | - Sonia Grego
- RTI International, 3040 E. Cornwallis Road, Research Triangle Park, NC 27709, USA
| |
Collapse
|
30
|
Lee S, Lee HA, Kim SJ, Kim KS. Cellular mechanisms for trazodone-induced cardiotoxicity. Hum Exp Toxicol 2015; 35:501-10. [DOI: 10.1177/0960327115595683] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The second-generation selective 5-HT2 receptor antagonists and reuptake inhibitors (SARIs) class antidepressants are known to have fewer cardiovascular side effects than the older ones. However, several case reports showed that trazodone, one of the second-generation SARIs, induces QT prolongation, cardiac arrhythmia, and ventricular tachycardia. Although these clinical cases suggested trazodone-induced cardiotoxicity, the toxicological actions of trazodone on cardiac action potentials (APs) beyond the human ether-a-go-go related gene (hERG) remain unclear. To elucidate the cellular mechanism for the adverse cardiac effects of trazodone, we investigated its effects on cardiac APs and ion channels using whole-cell patch clamp techniques in human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and transiently transfected human embryonic kidney cells (HEK293) with cardiac ion channel complementary DNA. Trazodone dose-dependently decreased the maximum upstroke velocity ( Vmax) and prolonged the AP duration, inducing early after depolarizations at 3 and 10 μM that triggered ventricular arrhythmias in hiPSC-CMs. Trazodone also inhibited all of the major ion channels ( IKr, IKs, INa, and ICa), with an especially high inhibitory potency on hERG. These data indicate that the prolonged AP duration and decreased Vmax due to trazodone are mainly the result of hERG and sodium ion inhibition, and its inhibitory effects on cardiac ion channels can be exhibited in hiPSC-CMs.
Collapse
Affiliation(s)
- S Lee
- Next-Generation Pharmaceutical Research Center, Korea Institute of Toxicology, Korea Research Institute of Chemical Technology, Yuseong-gu, Daejeon, Korea
| | - H-A Lee
- Next-Generation Pharmaceutical Research Center, Korea Institute of Toxicology, Korea Research Institute of Chemical Technology, Yuseong-gu, Daejeon, Korea
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - SJ Kim
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - K-S Kim
- Next-Generation Pharmaceutical Research Center, Korea Institute of Toxicology, Korea Research Institute of Chemical Technology, Yuseong-gu, Daejeon, Korea
| |
Collapse
|
31
|
Khan JM, Lyon AR, Harding SE. The case for induced pluripotent stem cell-derived cardiomyocytes in pharmacological screening. Br J Pharmacol 2014; 169:304-17. [PMID: 22845396 DOI: 10.1111/j.1476-5381.2012.02118.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The current drug screening models are deficient, particularly in detecting cardiac side effects. Human stem cell-derived cardiomyocytes could aid both early cardiotoxicity detection and novel drug discovery. Work over the last decade has generated human embryonic stem cells as potentially accurate sources of human cardiomyocytes, but ethical constraints and poor efficacy in establishing cell lines limit their use. Induced pluripotent stem cells do not require the use of human embryos and have the added advantage of producing patient-specific cardiomyocytes, allowing both generic and disease- and patient-specific pharmacological screening, as well as drug development through disease modelling. A critical question is whether sufficient standards have been achieved in the reliable and reproducible generation of 'adult-like' cardiomyocytes from human fibroblast tissue to progress from validation to safe use in practice and drug discovery. This review will highlight the need for a new experimental system, assess the validity of human induced pluripotent stem cell-derived cardiomyocytes and explore what the future may hold for their use in pharmacology.
Collapse
Affiliation(s)
- Jaffar M Khan
- Royal Brompton and Harefield NHS Trust, London, UK National Heart and Lung Institute, Imperial College, London, UK
| | | | | |
Collapse
|
32
|
Availability of human induced pluripotent stem cell-derived cardiomyocytes in assessment of drug potential for QT prolongation. Toxicol Appl Pharmacol 2014; 278:72-7. [DOI: 10.1016/j.taap.2014.04.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 03/18/2014] [Accepted: 04/05/2014] [Indexed: 11/20/2022]
|
33
|
Clements M, Thomas N. High-throughput multi-parameter profiling of electrophysiological drug effects in human embryonic stem cell derived cardiomyocytes using multi-electrode arrays. Toxicol Sci 2014; 140:445-61. [PMID: 24812011 DOI: 10.1093/toxsci/kfu084] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Human stem cell derived cardiomyocytes (hESC-CM) provide a potential model for development of improved assays for pre-clinical predictive drug safety screening. We have used multi-electrode array (MEA) analysis of hESC-CM to generate multi-parameter data to profile drug impact on cardiomyocyte electrophysiology using a panel of 21 compounds active against key cardiac ion channels. Our study is the first to apply multi-parameter phenotypic profiling and clustering techniques commonly used for high-content imaging and microarray data to the analysis of electrophysiology data obtained by MEA analysis. Our data show good correlations with previous studies in stem cell derived cardiomyocytes and demonstrate improved specificity in compound risk assignment over convention single-parametric approaches. These analyses indicate great potential for multi-parameter MEA data acquired from hESC-CM to enable drug electrophysiological liabilities to be assessed in pre-clinical cardiotoxicity assays, facilitating informed decision making and liability management at the optimum point in drug development.
Collapse
Affiliation(s)
| | - Nick Thomas
- GE Healthcare Life Sciences, Cardiff CF14 7YT, UK
| |
Collapse
|
34
|
Analysis of beat fluctuations and oxygen consumption in cardiomyocytes by scanning electrochemical microscopy. Anal Biochem 2014; 447:39-42. [DOI: 10.1016/j.ab.2013.11.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 10/18/2013] [Accepted: 11/07/2013] [Indexed: 11/19/2022]
|
35
|
Sarić T, Halbach M, Khalil M, Er F. Induced pluripotent stem cells as cardiac arrhythmic in vitro models and the impact for drug discovery. Expert Opin Drug Discov 2013; 9:55-76. [PMID: 24294840 DOI: 10.1517/17460441.2014.863275] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The development of new antiarrhythmic agents is challenging and is hampered by high attrition rate of novel drug candidates. One of the reasons for this is limited predictability of existing preclinical models for drug assessment. Cardiomyocytes (CMs) derived from disease-specific induced pluripotent stem cells (iPSC) represent a novel in vitro cellular model of cardiac arrhythmias with an unprecedented potential for generating new mechanistic insight into disease pathophysiology and improving the process of drug development. AREAS COVERED This review outlines recent studies demonstrating the suitability and limitations of iPSC-derived CMs (iPS-CMs) for in vitro modeling inherited arrhythmias and drug testing. The authors focus on channelopathies and outline the properties of iPS-CMs, highlighting their utility and limitations for investigating the mechanism of cardiac arrhythmias and drug discovery. EXPERT OPINION The iPS-CMs represent a valuable addition to the already existing armamentarium of cardiac arrhythmic models. However, the superiority of iPS-CMs over other arrhythmia models has not yet been rigorously established and the limitations of the model must be overcome before its full potential for antiarrhythmic drug discovery can be realized. Nevertheless, iPS cell-based platforms hold a great potential for increasing our knowledge about cellular arrhythmia mechanisms and improving the drug discovery process.
Collapse
Affiliation(s)
- Tomo Sarić
- University of Cologne, Institute for Neurophysiology, Center for Physiology and Pathophysiology, Medical Center , Robert Koch Str. 39, 50931 Cologne , Germany +49 221 478 86686 ; +49 221 478-3834 ;
| | | | | | | |
Collapse
|
36
|
Agarwal A, Goss JA, Cho A, McCain ML, Parker KK. Microfluidic heart on a chip for higher throughput pharmacological studies. LAB ON A CHIP 2013; 13:3599-608. [PMID: 23807141 PMCID: PMC3786400 DOI: 10.1039/c3lc50350j] [Citation(s) in RCA: 336] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
We present the design of a higher throughput "heart on a chip" which utilizes a semi-automated fabrication technique to process sub millimeter sized thin film cantilevers of soft elastomers. Anisotropic cardiac microtissues which recapitulate the laminar architecture of the heart ventricle are engineered on these cantilevers. Deflection of these cantilevers, termed Muscular Thin Films (MTFs), during muscle contraction allows calculation of diastolic and systolic stresses generated by the engineered tissues. We also present the design of a reusable one channel fluidic microdevice completely built out of autoclavable materials which incorporates various features required for an optical cardiac contractility assay: metallic base which fits on a heating element for temperature control, transparent top for recording cantilever deformation and embedded electrodes for electrical field stimulation of the tissue. We employ the microdevice to test the positive inotropic effect of isoproterenol on cardiac contractility at dosages ranging from 1 nM to 100 μM. The higher throughput fluidic heart on a chip has applications in testing of cardiac tissues built from rare or expensive cell sources and for integration with other organ mimics. These advances will help alleviate translational barriers for commercial adoption of these technologies by improving the throughput and reproducibility of readout, standardization of the platform and scalability of manufacture.
Collapse
Affiliation(s)
- Ashutosh Agarwal
- Disease Biophysics Group, Wyss Institute of Biologically Inspired Engineering, Harvard Stem Cell Institute, School of Engineering and Applied Sciences, Harvard University, 29 Oxford St, Pierce Hall Rm 321, Cambridge, MA, 02138, USA
| | - Josue Adrian Goss
- Disease Biophysics Group, Wyss Institute of Biologically Inspired Engineering, Harvard Stem Cell Institute, School of Engineering and Applied Sciences, Harvard University, 29 Oxford St, Pierce Hall Rm 321, Cambridge, MA, 02138, USA
| | - Alexander Cho
- Disease Biophysics Group, Wyss Institute of Biologically Inspired Engineering, Harvard Stem Cell Institute, School of Engineering and Applied Sciences, Harvard University, 29 Oxford St, Pierce Hall Rm 321, Cambridge, MA, 02138, USA
| | - Megan Laura McCain
- Disease Biophysics Group, Wyss Institute of Biologically Inspired Engineering, Harvard Stem Cell Institute, School of Engineering and Applied Sciences, Harvard University, 29 Oxford St, Pierce Hall Rm 321, Cambridge, MA, 02138, USA
| | - Kevin Kit Parker
- Disease Biophysics Group, Wyss Institute of Biologically Inspired Engineering, Harvard Stem Cell Institute, School of Engineering and Applied Sciences, Harvard University, 29 Oxford St, Pierce Hall Rm 321, Cambridge, MA, 02138, USA
- ; Fax: +(617) 495-9837; Tel: +(617) 495-2850
| |
Collapse
|
37
|
Human embryonic stem cell derived cardiac myocytes detect hERG-mediated repolarization effects, but not Nav1.5 induced depolarization delay. J Pharmacol Toxicol Methods 2013; 68:74-81. [DOI: 10.1016/j.vascn.2013.03.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 03/09/2013] [Accepted: 03/11/2013] [Indexed: 01/05/2023]
|
38
|
Fasina FO, Olaokun OO, Oladipo OO, Fasina MM, Makinde AA, Heath L, Bastos ADS. Phytochemical analysis and in-vitro anti-African swine fever virus activity of extracts and fractions of Ancistrocladus uncinatus, Hutch and Dalziel (Ancistrocladaceae). BMC Vet Res 2013; 9:120. [PMID: 23777548 PMCID: PMC3694037 DOI: 10.1186/1746-6148-9-120] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 06/10/2013] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND African swine fever (ASF), a highly contagious fatal acute haemorrhagic viral disease of pigs currently has no treatment or vaccination protocol and it threatens the pig industry worldwide. Recent outbreaks were managed by farmers with ethnoveterinary preparations with various claims of effectiveness. RESULTS We identified 35 compounds using GC-MS protocol and ASF virus (NIG 99) was significantly reduced by some extracts and fractions of the plant. However, the plant was poorly extracted by water and cytotoxicity was found to be a major problem with the use of the plant since its extracts also reduced the primary cells used in the assay. CONCLUSION It is confirmed that the plant has antiviral potentials against ASF virus and farmers' claims seem to have certain degree of veracity, but finding the best means of exploring the potential of the plant while reducing its cytotoxic effect in-vitro and in-vivo will be necessary.
Collapse
Affiliation(s)
- Folorunso O Fasina
- Department of Production Animal Studies, Faculty of Veterinary Science, University of Pretoria, Onderstepoort 0110, South Africa
- Mammal Research Institute, Department of Zoology and Entomology, University of Pretoria, Hatfield, South Africa
| | - Oyinlola O Olaokun
- Phytomedicine Programme, Department of Paraclinical Science, Faculty of Veterinary Science, University of Pretoria, Onderstepoort 0110, South Africa
| | | | - Margaret M Fasina
- Maximum Farms, P. O. Box 23, Vom Post Office, Plateau State, Nigeria
| | | | - Livio Heath
- Transboundary Animal Disease Programme, ARC-Onderstepoort Veterinary Institute, Onderstepoort, South Africa
| | - Armanda DS Bastos
- Mammal Research Institute, Department of Zoology and Entomology, University of Pretoria, Hatfield, South Africa
| |
Collapse
|
39
|
Harris K, Aylott M, Cui Y, Louttit JB, McMahon NC, Sridhar A. Comparison of Electrophysiological Data From Human-Induced Pluripotent Stem Cell–Derived Cardiomyocytes to Functional Preclinical Safety Assays. Toxicol Sci 2013; 134:412-26. [DOI: 10.1093/toxsci/kft113] [Citation(s) in RCA: 177] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
40
|
Himmel HM. Drug-induced functional cardiotoxicity screening in stem cell-derived human and mouse cardiomyocytes: effects of reference compounds. J Pharmacol Toxicol Methods 2013; 68:97-111. [PMID: 23702537 DOI: 10.1016/j.vascn.2013.05.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 05/10/2013] [Accepted: 05/10/2013] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Early prediction of drug-induced functional cardiotoxicity requires robust in-vitro systems suitable for medium/high throughput and easily accessible cardiomyocytes with defined reproducible properties. The xCELLigence Cardio system uses 96-well plates with interdigitated electrodes that detect the impedance changes of rhythmic contractions of stem cell-derived cardiomyocyte (SC-CM) layers. Here, we report on our initial screening experience in comparison to established (multi)cellular and in-vivo models. METHODS Impedance signals from human iPSC-CM (iCells™) and mouse eSC-CM (Cor.At™) were analyzed for contraction amplitude (CA) and duration, rise/fall time, beating rate (BR) and irregularity. RESULTS Following solution exchange, impedance signals re-approximated steady-state conditions after about 2 (Cor.At™) and 3h (iCells™); these time points were used to analyze drug effects. The solvent DMSO (≤1%) hardly influenced contraction parameters in Cor.At™, whereas in iCells™ DMSO (>0.1%) reduced CA and enhanced BR. The selective hERG K⁺ channel blockers E-4031 and dofetilide reduced CA and accelerated BR (≥30 nM) according to the analysis software. The latter, however, was due to burst-like contractions (300 nM) that could be detected only by visual inspection of recordings, and were more pronounced in Cor.At™ as in iCells™. In cardiac myocytes and tissue preparations, however, E4031 and dofetilide have been reported to increase cell shortening and contractile force and to reduce BR. Compounds (pentamidine, HMR1556, ATX2, TTX, and verapamil) with other mechanisms of action were also investigated; their effects differed partially between cell lines (e.g. TTX) and compared to established (multi)cellular models (e.g. HMR1556, ouabain). CONCLUSION Mouse and human stem cell-derived cardiomyocytes respond differently to drugs and these responses occasionally also differ from those originating from established in-vitro and in-vivo models. Hence, drug-induced cardiotoxic effects may be detected with this system, however, the predictive or even translational value of results is considered limited and not yet firmly established.
Collapse
|
41
|
Ragone MI, Torres NS, Consolini AE. Energetic study of cardioplegic hearts under ischaemia/reperfusion and [Ca(2+)] changes in cardiomyocytes of guinea-pig: mitochondrial role. Acta Physiol (Oxf) 2013; 207:369-84. [PMID: 23171431 DOI: 10.1111/apha.12027] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 09/08/2012] [Accepted: 10/02/2012] [Indexed: 12/25/2022]
Abstract
AIM To study the role of mitochondria in the recovery of guinea-pig hearts exposed to high-K(+)-cardioplegia (CPG) and ischaemia/reperfusion (I/R) METHODS: We measured contractility and heat release in perfused guinea-pig hearts and cytosolic and mitochondrial Ca(2+) by epifluorescence and confocal microscopy in isolated cardiomyocytes loaded with Fluo-4 or Rhod-2. RESULTS In hearts, CPG increased the postischaemic contractile recovery, and this was potentiated by the mNCX blocker clonazepam and the mKATP opener diazoxide, which also prevented the fall in muscle economy. Moreover, CPG prevented the stunning induced by ouabain, which was reduced by clonazepam. In cardiomyocytes, CPG increased fluorescent signals of cytosolic and mitochondrial Ca(2+), while the addition of a mNCX blocker (CGP37157) increased cytosolic but reduced mitochondrial [Ca(2+)]. Ouabain in CPG increased cytosolic Ca(2+) and resting heat, but the addition of CGP37157 reduced them, as well as mitochondrial Ca(2+). CONCLUSIONS CPG, diazoxide and clonazepam improve postischaemic recovery, respectively, by increasing the Ca(2+) cycling and by reducing the mitochondrial Ca(2+) uptake either by uniporter or by mNCX. The mitochondria compete with the leaky sarcoplasmic reticulum (SR) as sink of Ca(2+) in guinea-pig hearts, affecting the postischaemic contractility. CPG also prevented the ouabain-induced dysfunction by avoiding the Ca(2+) overload. Ouabain reduced the synergism between CPG and clonazepam suggesting that [Na(+)]i and SR load influence the mNCX role.
Collapse
Affiliation(s)
- M. I. Ragone
- Cátedra de Farmacología, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas; Universidad Nacional de La Plata (UNLP); La Plata; Argentina
| | - N. S. Torres
- The Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI); Salt Lake City; UT; USA
| | - A. E. Consolini
- Cátedra de Farmacología, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas; Universidad Nacional de La Plata (UNLP); La Plata; Argentina
| |
Collapse
|
42
|
Deployment of in silico and in vitro safety assays in early-stage drug discovery. Future Med Chem 2012; 4:1211-3. [PMID: 22800365 DOI: 10.4155/fmc.12.71] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
43
|
Blazeski A, Zhu R, Hunter DW, Weinberg SH, Zambidis ET, Tung L. Cardiomyocytes derived from human induced pluripotent stem cells as models for normal and diseased cardiac electrophysiology and contractility. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2012; 110:166-77. [PMID: 22971665 PMCID: PMC3910285 DOI: 10.1016/j.pbiomolbio.2012.07.013] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 07/30/2012] [Indexed: 12/21/2022]
Abstract
Since the first description of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), these cells have garnered tremendous interest for their potential use in patient-specific analysis and therapy. Additionally, hiPSC-CMs can be derived from donor cells from patients with specific cardiac disorders, enabling in vitro human disease models for mechanistic study and therapeutic drug assessment. However, a full understanding of their electrophysiological and contractile function is necessary before this potential can be realized. Here, we review this emerging field from a functional perspective, with particular emphasis on beating rate, action potential, ionic currents, multicellular conduction, calcium handling and contraction. We further review extant hiPSC-CM disease models that recapitulate genetic myocardial disease.
Collapse
Affiliation(s)
- Adriana Blazeski
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD
| | - Renjun Zhu
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD
| | - David W. Hunter
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD
| | - Seth H. Weinberg
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD
| | - Elias T. Zambidis
- Institute for Cell Engineering and Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University, Baltimore, MD
| | - Leslie Tung
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD
| |
Collapse
|
44
|
Puppala D, Collis LP, Sun SZ, Bonato V, Chen X, Anson B, Pletcher M, Fermini B, Engle SJ. Comparative gene expression profiling in human-induced pluripotent stem cell--derived cardiocytes and human and cynomolgus heart tissue. Toxicol Sci 2012; 131:292-301. [PMID: 22982684 DOI: 10.1093/toxsci/kfs282] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Cardiotoxicity is one of the leading causes of drug attrition. Current in vitro models insufficiently predict cardiotoxicity, and there is a need for alternative physiologically relevant models. Here we describe the gene expression profile of human-induced pluripotent stem cell-derived cardiocytes (iCC) postthaw over a period of 42 days in culture and compare this profile to human fetal and adult as well as adult cynomolgus nonhuman primate (NHP, Macaca fascicularis) heart tissue. Our results indicate that iCC express relevant cardiac markers such as ion channels (SCN5A, KCNJ2, CACNA1C, KCNQ1, and KCNH2), tissue-specific structural markers (MYH6, MYLPF, MYBPC3, DES, TNNT2, and TNNI3), and transcription factors (NKX2.5, GATA4, and GATA6) and lack the expression of stem cell markers (FOXD3, GBX2, NANOG, POU5F1, SOX2, and ZFP42). Furthermore, we performed a functional evaluation of contractility of the iCC and showed functional and pharmacological correlations with myocytes isolated from adult NHP hearts. These results suggest that stem cell-derived cardiocytes may represent a novel in vitro model to study human cardiac toxicity with potential ex vivo and in vivo translation.
Collapse
Affiliation(s)
- Dinesh Puppala
- Compound Safety Prediction, Pfizer, Inc., Groton, Connecticut 06340, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Sirenko O, Crittenden C, Callamaras N, Hesley J, Chen YW, Funes C, Rusyn I, Anson B, Cromwell EF. Multiparameter in vitro assessment of compound effects on cardiomyocyte physiology using iPSC cells. ACTA ACUST UNITED AC 2012; 18:39-53. [PMID: 22972846 DOI: 10.1177/1087057112457590] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A large percentage of drugs fail in clinical studies due to cardiac toxicity; thus, development of sensitive in vitro assays that can evaluate potential adverse effects on cardiomyocytes is extremely important for drug development. Human cardiomyocytes derived from stem cell sources offer more clinically relevant cell-based models than those presently available. Human-induced pluripotent stem cell-derived cardiomyocytes are especially attractive because they express ion channels and demonstrate spontaneous mechanical and electrical activity similar to adult cardiomyocytes. Here we demonstrate techniques for measuring the impact of pharmacologic compounds on the beating rate of cardiomyocytes with ImageXpress Micro and FLIPR Tetra systems. The assays employ calcium-sensitive dyes to monitor changes in Ca(2+) fluxes synchronous with cell beating, which allows monitoring of the beat rate, amplitude, and other parameters. We demonstrate here that the system is able to detect concentration-dependent atypical patterns caused by hERG inhibitors and other ion channel blockers. We also show that both positive and negative chronotropic effects on cardiac rate can be observed and IC(50) values determined. This methodology is well suited for safety testing and can be used to estimate efficacy and dosing of drug candidates prior to clinical studies.
Collapse
|
46
|
Rajamohan D, Matsa E, Kalra S, Crutchley J, Patel A, George V, Denning C. Current status of drug screening and disease modelling in human pluripotent stem cells. Bioessays 2012; 35:281-98. [PMID: 22886688 PMCID: PMC3597971 DOI: 10.1002/bies.201200053] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The emphasis in human pluripotent stem cell (hPSC) technologies has shifted from cell therapy to in vitro disease modelling and drug screening. This review examines why this shift has occurred, and how current technological limitations might be overcome to fully realise the potential of hPSCs. Details are provided for all disease-specific human induced pluripotent stem cell lines spanning a dozen dysfunctional organ systems. Phenotype and pharmacology have been examined in only 17 of 63 lines, primarily those that model neurological and cardiac conditions. Drug screening is most advanced in hPSC-cardiomyocytes. Responses for almost 60 agents include examples of how careful tests in hPSC-cardiomyocytes have improved on existing in vitro assays, and how these cells have been integrated into high throughput imaging and electrophysiology industrial platforms. Such successes will provide an incentive to overcome bottlenecks in hPSC technology such as improving cell maturity and industrial scalability whilst reducing cost.
Collapse
Affiliation(s)
- Divya Rajamohan
- Department of Stem Cells, Tissue Engineering & Modelling, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | | | | | | | | | | | | |
Collapse
|
47
|
Blazeski A, Zhu R, Hunter DW, Weinberg SH, Boheler KR, Zambidis ET, Tung L. Electrophysiological and contractile function of cardiomyocytes derived from human embryonic stem cells. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2012; 110:178-95. [PMID: 22958937 DOI: 10.1016/j.pbiomolbio.2012.07.012] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 07/30/2012] [Indexed: 12/23/2022]
Abstract
Human embryonic stem cells have emerged as the prototypical source from which cardiomyocytes can be derived for use in drug discovery and cell therapy. However, such applications require that these cardiomyocytes (hESC-CMs) faithfully recapitulate the physiology of adult cells, especially in relation to their electrophysiological and contractile function. We review what is known about the electrophysiology of hESC-CMs in terms of beating rate, action potential characteristics, ionic currents, and cellular coupling as well as their contractility in terms of calcium cycling and contraction. We also discuss the heterogeneity in cellular phenotypes that arises from variability in cardiac differentiation, maturation, and culture conditions, and summarize present strategies that have been implemented to reduce this heterogeneity. Finally, we present original electrophysiological data from optical maps of hESC-CM clusters.
Collapse
Affiliation(s)
- Adriana Blazeski
- Department of Biomedical Engineering, The Johns Hopkins University, 720 Rutland Ave., Baltimore, MD 21205, USA
| | | | | | | | | | | | | |
Collapse
|