1
|
Kyro GW, Martin MT, Watt ED, Batista VS. CardioGenAI: a machine learning-based framework for re-engineering drugs for reduced hERG liability. J Cheminform 2025; 17:30. [PMID: 40045386 PMCID: PMC11881490 DOI: 10.1186/s13321-025-00976-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 02/21/2025] [Indexed: 03/09/2025] Open
Abstract
The link between in vitro hERG ion channel inhibition and subsequent in vivo QT interval prolongation, a critical risk factor for the development of arrythmias such as Torsade de Pointes, is so well established that in vitro hERG activity alone is often sufficient to end the development of an otherwise promising drug candidate. It is therefore of tremendous interest to develop advanced methods for identifying hERG-active compounds in the early stages of drug development, as well as for proposing redesigned compounds with reduced hERG liability and preserved primary pharmacology. In this work, we present CardioGenAI, a machine learning-based framework for re-engineering both developmental and commercially available drugs for reduced hERG activity while preserving their pharmacological activity. The framework incorporates novel state-of-the-art discriminative models for predicting hERG channel activity, as well as activity against the voltage-gated NaV1.5 and CaV1.2 channels due to their potential implications in modulating the arrhythmogenic potential induced by hERG channel blockade. We applied the complete framework to pimozide, an FDA-approved antipsychotic agent that demonstrates high affinity to the hERG channel, and generated 100 refined candidates. Remarkably, among the candidates is fluspirilene, a compound which is of the same class of drugs as pimozide (diphenylmethanes) and therefore has similar pharmacological activity, yet exhibits over 700-fold weaker binding to hERG. Furthermore, we demonstrated the framework's ability to optimize hERG, NaV1.5 and CaV1.2 profiles of multiple FDA-approved compounds while maintaining the physicochemical nature of the original drugs. We envision that this method can effectively be applied to developmental compounds exhibiting hERG liabilities to provide a means of rescuing drug development programs that have stalled due to hERG-related safety concerns. Additionally, the discriminative models can also serve independently as effective components of virtual screening pipelines. We have made all of our software open-source at https://github.com/gregory-kyro/CardioGenAI to facilitate integration of the CardioGenAI framework for molecular hypothesis generation into drug discovery workflows.Scientific contributionThis work introduces CardioGenAI, an open-source machine learning-based framework designed to re-engineer drugs for reduced hERG liability while preserving their pharmacological activity. The complete CardioGenAI framework can be applied to developmental compounds exhibiting hERG liabilities to provide a means of rescuing drug discovery programs facing hERG-related challenges. In addition, the framework incorporates novel state-of-the-art discriminative models for predicting hERG, NaV1.5 and CaV1.2 channel activity, which can function independently as effective components of virtual screening pipelines.
Collapse
Affiliation(s)
- Gregory W Kyro
- Department of Chemistry, Yale University, New Haven, CT, 06511, USA.
- Drug Safety Research & Development, Pfizer Research & Development, Groton, CT, 06340, USA.
| | - Matthew T Martin
- Drug Safety Research & Development, Pfizer Research & Development, Groton, CT, 06340, USA
| | - Eric D Watt
- Drug Safety Research & Development, Pfizer Research & Development, Groton, CT, 06340, USA
| | - Victor S Batista
- Department of Chemistry, Yale University, New Haven, CT, 06511, USA.
| |
Collapse
|
2
|
Kim J, Won D, Kim TH, Kim CY, Ko SH. Rapid prototyping and facile customization of conductive hydrogel bioelectronics based on all laser process. Biosens Bioelectron 2024; 258:116327. [PMID: 38703496 DOI: 10.1016/j.bios.2024.116327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/05/2024] [Accepted: 04/22/2024] [Indexed: 05/06/2024]
Abstract
Proper customization in size and shape is essential in implantable bioelectronics for stable bio-signal recording. Over the past decades, many researchers have heavily relied on conventional photolithography processes to fabricate implantable bioelectronics. Therefore, they could not avoid the critical limitation of high cost and complex processing steps to optimize bioelectronic devices for target organs with various sizes and shapes. Here, we propose rapid prototyping using all laser processes to fabricate customized bioelectronics. PEDOT:PSS is selectively irradiated by an ultraviolet (UV) pulse laser to form wet-stable conductive hydrogels that can softly interact with biological tissues (50 μm line width). The encapsulation layer is selectively patterned using the same laser source by UV-curing polymer networks (110 μm line width). For high stretchability (over 100%), mesh structures are made by the selective laser cutting process. Our rapid prototyping strategy minimizes the use of high-cost equipment, using only a single UV laser source to process the electrodes, encapsulation, and substrates that constitute bioelectronics without a photomask, enabling the prototyping stretchable microelectrode array with an area of 1 cm2 less than 10 min. We fabricated an optimized stretchable microelectrode array with low impedances (∼1.1 kΩ at 1 kHz) that can effectively record rat's cardiac signals with various health states.
Collapse
Affiliation(s)
- Jin Kim
- Department of Physiology, College of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Daeyeon Won
- Applied Nano and Thermal Science Lab, Department of Mechanical Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Tae Hyun Kim
- Applied Nano and Thermal Science Lab, Department of Mechanical Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea; School of Mechanical Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - C-Yoon Kim
- College of Veterinary Medicine, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea.
| | - Seung Hwan Ko
- Applied Nano and Thermal Science Lab, Department of Mechanical Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea; Institute of Engineering Research / Institute of Advanced Machinery and Design (SNU-IAMD), Seoul National University, Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea; Interdisciplinary Program in Bioengineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
| |
Collapse
|
3
|
Xiang Y, Shi K, Li Y, Xue J, Tong Z, Li H, Li Z, Teng C, Fang J, Hu N. Active Micro-Nano-Collaborative Bioelectronic Device for Advanced Electrophysiological Recording. NANO-MICRO LETTERS 2024; 16:132. [PMID: 38411852 PMCID: PMC10899154 DOI: 10.1007/s40820-024-01336-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 12/28/2023] [Indexed: 02/28/2024]
Abstract
The development of precise and sensitive electrophysiological recording platforms holds the utmost importance for research in the fields of cardiology and neuroscience. In recent years, active micro/nano-bioelectronic devices have undergone significant advancements, thereby facilitating the study of electrophysiology. The distinctive configuration and exceptional functionality of these active micro-nano-collaborative bioelectronic devices offer the potential for the recording of high-fidelity action potential signals on a large scale. In this paper, we review three-dimensional active nano-transistors and planar active micro-transistors in terms of their applications in electro-excitable cells, focusing on the evaluation of the effects of active micro/nano-bioelectronic devices on electrophysiological signals. Looking forward to the possibilities, challenges, and wide prospects of active micro-nano-devices, we expect to advance their progress to satisfy the demands of theoretical investigations and medical implementations within the domains of cardiology and neuroscience research.
Collapse
Affiliation(s)
- Yuting Xiang
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 310058, People's Republic of China
- Department of Obstetrics and Gynecology, The Tenth Affiliated Hospital, Southern Medical University, Dongguan, 523059, People's Republic of China
- Dongguan Key Laboratory of Major Diseases in Obstetrics and Gynecology, Dongguan, 523059, People's Republic of China
| | - Keda Shi
- Department of Lung Transplantation and General Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Ying Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
| | - Jiajin Xue
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou, 310052, People's Republic of China
| | - Zhicheng Tong
- Department of Orthopedics, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322005, People's Republic of China
| | - Huiming Li
- Department of Orthopedics, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322005, People's Republic of China
| | - Zhongjun Li
- Department of Obstetrics and Gynecology, The Tenth Affiliated Hospital, Southern Medical University, Dongguan, 523059, People's Republic of China.
- Dongguan Key Laboratory of Major Diseases in Obstetrics and Gynecology, Dongguan, 523059, People's Republic of China.
| | - Chong Teng
- Department of Orthopedics, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322005, People's Republic of China.
| | - Jiaru Fang
- School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China.
| | - Ning Hu
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 310058, People's Republic of China.
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou, 310052, People's Republic of China.
| |
Collapse
|
4
|
Yang W, Ouyang Q, Zhu Z, Wu Y, Fan M, Liao Y, Guo X, Xu Z, Zhang X, Zhang Y, Hu N, Zhang D. A biosensing system employing nonlinear dynamic analysis-assisted neural network for drug-induced cardiotoxicity assessment. Biosens Bioelectron 2023; 222:114923. [PMID: 36455375 DOI: 10.1016/j.bios.2022.114923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 11/16/2022] [Indexed: 11/18/2022]
Abstract
Preclinical investigation of drug-induced cardiotoxicity is of importance for drug development. To evaluate such cardiotoxicity, in vitro high-throughput interdigitated electrode-based recording of cardiomyocytes mechanical beating is widely used. To automatically analyze the features from the beating signals for drug-induced cardiotoxicity assessment, artificial neural network analysis is conventionally employed and signals are segmented into cycles and feature points are located in the cycles. However, signal segmentation and location of feature points for different signal shapes require design of specific algorithms. Consequently, this may lower the efficiency of research and the applications of such algorithms in signals with different morphologies are limited. Here, we present a biosensing system that employs nonlinear dynamic analysis-assisted neural network (NDANN) to avoid the signal segmentation process and directly extract features from beating signal time series. By processing beating time series with fixed time duration to avoid the signal segmentation process, this NDANN-based biosensing system can identify drug-induced cardiotoxicity with accuracy over 0.99. The individual drugs were classified with high accuracies over 0.94 and drug-induced cardiotoxicity levels were accurately predicted. We also evaluated the generalization performance of the NDANN-based biosensing system in assessing drug-induced cardiotoxicity through an independent dataset. This system achieved accuracy of 0.85-0.95 for different drug concentrations in identification of drug-induced cardiotoxicity. This result demonstrates that our NDANN-based biosensing system has the capacity of screening newly developed drugs, which is crucial in practical applications. This NDANN-based biosensing system can work as a new screening platform for drug-induced cardiotoxicity and improve the efficiency of bio-signal processing.
Collapse
Affiliation(s)
- Wenjian Yang
- Research Center for Intelligent Sensing Systems, Zhejiang Laboratory, Hangzhou, 311100, China
| | - Qiangqiang Ouyang
- First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Zhijing Zhu
- Key Laboratory of Novel Target and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, School of Computer & Computing Science, Zhejiang University City College, Hangzhou, 310015, China; School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Yue Wu
- Research Center for Intelligent Sensing Systems, Zhejiang Laboratory, Hangzhou, 311100, China.
| | - Minzhi Fan
- Research Center for Intelligent Sensing Systems, Zhejiang Laboratory, Hangzhou, 311100, China
| | - Yuheng Liao
- Research Center for Intelligent Sensing Systems, Zhejiang Laboratory, Hangzhou, 311100, China
| | - Xinyu Guo
- Research Center for Intelligent Sensing Systems, Zhejiang Laboratory, Hangzhou, 311100, China
| | - Zhongyuan Xu
- Research Center for Intelligent Sensing Systems, Zhejiang Laboratory, Hangzhou, 311100, China
| | - Xiaoyu Zhang
- Research Center for Intelligent Sensing Systems, Zhejiang Laboratory, Hangzhou, 311100, China
| | - Yunshan Zhang
- Research Center for Intelligent Sensing Systems, Zhejiang Laboratory, Hangzhou, 311100, China
| | - Ning Hu
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311200, China; Stoddart Institute of Molecular Science, Department of Chemistry, Zhejiang University, Hangzhou, 310058, China
| | - Diming Zhang
- Research Center for Intelligent Sensing Systems, Zhejiang Laboratory, Hangzhou, 311100, China.
| |
Collapse
|
5
|
Feng H, Wei GW. Virtual screening of DrugBank database for hERG blockers using topological Laplacian-assisted AI models. Comput Biol Med 2023; 153:106491. [PMID: 36599209 PMCID: PMC10120853 DOI: 10.1016/j.compbiomed.2022.106491] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 11/29/2022] [Accepted: 12/27/2022] [Indexed: 12/29/2022]
Abstract
The human ether-a-go-go (hERG) potassium channel (Kv11.1) plays a critical role in mediating cardiac action potential. The blockade of this ion channel can potentially lead fatal disorder and/or long QT syndrome. Many drugs have been withdrawn because of their serious hERG-cardiotoxicity. It is crucial to assess the hERG blockade activity in the early stage of drug discovery. We are particularly interested in the hERG-cardiotoxicity of compounds collected in the DrugBank database considering that many DrugBank compounds have been approved for therapeutic treatments or have high potential to become drugs. Machine learning-based in silico tools offer a rapid and economical platform to virtually screen DrugBank compounds. We design accurate and robust classifiers for blockers/non-blockers and then build regressors to quantitatively analyze the binding potency of the DrugBank compounds on the hERG channel. Molecular sequences are embedded with two natural language processing (NLP) methods, namely, autoencoder and transformer. Complementary three-dimensional (3D) molecular structures are embedded with two advanced mathematical approaches, i.e., topological Laplacians and algebraic graphs. With our state-of-the-art tools, we reveal that 227 out of the 8641 DrugBank compounds are potential hERG blockers, suggesting serious drug safety problems. Our predictions provide guidance for the further experimental interrogation of DrugBank compounds' hERG-cardiotoxicity.
Collapse
Affiliation(s)
- Hongsong Feng
- Department of Mathematics, Michigan State University, MI 48824, USA
| | - Guo-Wei Wei
- Department of Mathematics, Michigan State University, MI 48824, USA; Department of Electrical and Computer Engineering, Michigan State University, MI 48824, USA; Department of Biochemistry and Molecular Biology, Michigan State University, MI 48824, USA.
| |
Collapse
|
6
|
Mai P, Hampl J, Baca M, Brauer D, Singh S, Weise F, Borowiec J, Schmidt A, Küstner JM, Klett M, Gebinoga M, Schroeder IS, Markert UR, Glahn F, Schumann B, Eckstein D, Schober A. MatriGrid® Based Biological Morphologies: Tools for 3D Cell Culturing. Bioengineering (Basel) 2022; 9:bioengineering9050220. [PMID: 35621498 PMCID: PMC9138054 DOI: 10.3390/bioengineering9050220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 02/06/2023] Open
Abstract
Recent trends in 3D cell culturing has placed organotypic tissue models at another level. Now, not only is the microenvironment at the cynosure of this research, but rather, microscopic geometrical parameters are also decisive for mimicking a tissue model. Over the years, technologies such as micromachining, 3D printing, and hydrogels are making the foundation of this field. However, mimicking the topography of a particular tissue-relevant substrate can be achieved relatively simply with so-called template or morphology transfer techniques. Over the last 15 years, in one such research venture, we have been investigating a micro thermoforming technique as a facile tool for generating bioinspired topographies. We call them MatriGrid®s. In this research account, we summarize our learning outcome from this technique in terms of the influence of 3D micro morphologies on different cell cultures that we have tested in our laboratory. An integral part of this research is the evolution of unavoidable aspects such as possible label-free sensing and fluidic automatization. The development in the research field is also documented in this account.
Collapse
Affiliation(s)
- Patrick Mai
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Jörg Hampl
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
- Correspondence: (J.H.); (A.S.); Tel.: +49-3677-6933387 (A.S.)
| | - Martin Baca
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Dana Brauer
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Sukhdeep Singh
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Frank Weise
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Justyna Borowiec
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - André Schmidt
- Placenta Lab, Department of Obstetrics, Jena University Hospital, 07747 Jena, Germany; (A.S.); (U.R.M.)
| | - Johanna Merle Küstner
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Maren Klett
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Michael Gebinoga
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Insa S. Schroeder
- Biophysics Division, GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany;
| | - Udo R. Markert
- Placenta Lab, Department of Obstetrics, Jena University Hospital, 07747 Jena, Germany; (A.S.); (U.R.M.)
| | - Felix Glahn
- Institute of Environmental Toxicology, Martin-Luther-University Halle-Wittenberg, 06097 Halle, Germany; (F.G.); (B.S.); (D.E.)
| | - Berit Schumann
- Institute of Environmental Toxicology, Martin-Luther-University Halle-Wittenberg, 06097 Halle, Germany; (F.G.); (B.S.); (D.E.)
| | - Diana Eckstein
- Institute of Environmental Toxicology, Martin-Luther-University Halle-Wittenberg, 06097 Halle, Germany; (F.G.); (B.S.); (D.E.)
| | - Andreas Schober
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
- Correspondence: (J.H.); (A.S.); Tel.: +49-3677-6933387 (A.S.)
| |
Collapse
|
7
|
Youhanna S, Kemas AM, Preiss L, Zhou Y, Shen JX, Cakal SD, Paqualini FS, Goparaju SK, Shafagh RZ, Lind JU, Sellgren CM, Lauschke VM. Organotypic and Microphysiological Human Tissue Models for Drug Discovery and Development-Current State-of-the-Art and Future Perspectives. Pharmacol Rev 2022; 74:141-206. [PMID: 35017176 DOI: 10.1124/pharmrev.120.000238] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 10/12/2021] [Indexed: 12/11/2022] Open
Abstract
The number of successful drug development projects has been stagnant for decades despite major breakthroughs in chemistry, molecular biology, and genetics. Unreliable target identification and poor translatability of preclinical models have been identified as major causes of failure. To improve predictions of clinical efficacy and safety, interest has shifted to three-dimensional culture methods in which human cells can retain many physiologically and functionally relevant phenotypes for extended periods of time. Here, we review the state of the art of available organotypic culture techniques and critically review emerging models of human tissues with key importance for pharmacokinetics, pharmacodynamics, and toxicity. In addition, developments in bioprinting and microfluidic multiorgan cultures to emulate systemic drug disposition are summarized. We close by highlighting important trends regarding the fabrication of organotypic culture platforms and the choice of platform material to limit drug absorption and polymer leaching while supporting the phenotypic maintenance of cultured cells and allowing for scalable device fabrication. We conclude that organotypic and microphysiological human tissue models constitute promising systems to promote drug discovery and development by facilitating drug target identification and improving the preclinical evaluation of drug toxicity and pharmacokinetics. There is, however, a critical need for further validation, benchmarking, and consolidation efforts ideally conducted in intersectoral multicenter settings to accelerate acceptance of these novel models as reliable tools for translational pharmacology and toxicology. SIGNIFICANCE STATEMENT: Organotypic and microphysiological culture of human cells has emerged as a promising tool for preclinical drug discovery and development that might be able to narrow the translation gap. This review discusses recent technological and methodological advancements and the use of these systems for hit discovery and the evaluation of toxicity, clearance, and absorption of lead compounds.
Collapse
Affiliation(s)
- Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Aurino M Kemas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Lena Preiss
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Joanne X Shen
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Selgin D Cakal
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Francesco S Paqualini
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Sravan K Goparaju
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Reza Zandi Shafagh
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Johan Ulrik Lind
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Carl M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| |
Collapse
|
8
|
Kim JS, Choi SW, Park YG, Kim SJ, Choi CH, Cha MJ, Chang JH. Impact of High-Dose Irradiation on Human iPSC-Derived Cardiomyocytes Using Multi-Electrode Arrays: Implications for the Antiarrhythmic Effects of Cardiac Radioablation. Int J Mol Sci 2021; 23:351. [PMID: 35008778 PMCID: PMC8745341 DOI: 10.3390/ijms23010351] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 12/19/2022] Open
Abstract
Cardiac radioablation is emerging as an alternative option for refractory ventricular arrhythmias. However, the immediate acute effect of high-dose irradiation on human cardiomyocytes remains poorly known. We measured the electrical activities of human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) upon irradiation with 0, 20, 25, 30, 40, and 50 Gy using a multi-electrode array, and cardiomyocyte function gene levels were evaluated. iPSC-CMs showed to recover their electrophysiological activities (total active electrode, spike amplitude and slope, and corrected field potential duration) within 3-6 h from the acute effects of high-dose irradiation. The beat rate immediately increased until 3 h after irradiation, but it steadily decreased afterward. Conduction velocity slowed in cells irradiated with ≥25 Gy until 6-12 h and recovered within 24 h; notably, 20 and 25 Gy-treated groups showed subsequent continuous increase. At day 7 post-irradiation, except for cTnT, cardiomyocyte function gene levels increased with increasing irradiation dose, but uniquely peaked at 25-30 Gy. Altogether, high-dose irradiation immediately and reversibly modifies the electrical conduction of cardiomyocytes. Thus, compensatory mechanisms at the cellular level may be activated after the high-dose irradiation acute effects, thereby, contributing to the immediate antiarrhythmic outcome of cardiac radioablation for refractory ventricular arrhythmias.
Collapse
Affiliation(s)
- Jae Sik Kim
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul 03080, Korea; (J.S.K.); (C.H.C.)
- Department of Radiation Oncology, Kyung Hee University Hospital at Gangdong, Seoul 05278, Korea
| | - Seong Woo Choi
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Korea;
| | - Yun-Gwi Park
- Stem Cell Research Institute, T&R Biofab Co., Ltd., Siheung 15073, Korea;
| | - Sung Joon Kim
- Department of Physiology & Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03080, Korea;
| | - Chang Heon Choi
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul 03080, Korea; (J.S.K.); (C.H.C.)
- Department of Radiation Oncology, Seoul National University Hospital, Seoul 03080, Korea
| | - Myung-Jin Cha
- Division of Cardiology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Ji Hyun Chang
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul 03080, Korea; (J.S.K.); (C.H.C.)
- Department of Radiation Oncology, Seoul National University Hospital, Seoul 03080, Korea
| |
Collapse
|
9
|
Chen L, He Y, Wang X, Ge J, Li H. Ventricular voltage-gated ion channels: Detection, characteristics, mechanisms, and drug safety evaluation. Clin Transl Med 2021; 11:e530. [PMID: 34709746 PMCID: PMC8516344 DOI: 10.1002/ctm2.530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiac voltage-gated ion channels (VGICs) play critical roles in mediating cardiac electrophysiological signals, such as action potentials, to maintain normal heart excitability and contraction. Inherited or acquired alterations in the structure, expression, or function of VGICs, as well as VGIC-related side effects of pharmaceutical drug delivery can result in abnormal cellular electrophysiological processes that induce life-threatening cardiac arrhythmias or even sudden cardiac death. Hence, to reduce possible heart-related risks, VGICs must be acknowledged as important targets in drug discovery and safety studies related to cardiac disease. In this review, we first summarize the development and application of electrophysiological techniques that are employed in cardiac VGIC studies alone or in combination with other techniques such as cryoelectron microscopy, optical imaging and optogenetics. Subsequently, we describe the characteristics, structure, mechanisms, and functions of various well-studied VGICs in ventricular myocytes and analyze their roles in and contributions to both physiological cardiac excitability and inherited cardiac diseases. Finally, we address the implications of the structure and function of ventricular VGICs for drug safety evaluation. In summary, multidisciplinary studies on VGICs help researchers discover potential targets of VGICs and novel VGICs in heart, enrich their knowledge of the properties and functions, determine the operation mechanisms of pathological VGICs, and introduce groundbreaking trends in drug therapy strategies, and drug safety evaluation.
Collapse
Affiliation(s)
- Lulan Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular DiseasesShanghai Xuhui District Central Hospital & Zhongshan‐xuhui Hospital, Zhongshan Hospital, Fudan UniversityShanghaiChina
| | - Yue He
- Department of CardiologyShanghai Xuhui District Central Hospital & Zhongshan‐xuhui HospitalShanghaiChina
| | - Xiangdong Wang
- Institute of Clinical Science, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular DiseasesShanghai Xuhui District Central Hospital & Zhongshan‐xuhui Hospital, Zhongshan Hospital, Fudan UniversityShanghaiChina
| | - Hua Li
- Department of Cardiology, Shanghai Institute of Cardiovascular DiseasesShanghai Xuhui District Central Hospital & Zhongshan‐xuhui Hospital, Zhongshan Hospital, Fudan UniversityShanghaiChina
| |
Collapse
|
10
|
Xu D, Mo J, Xie X, Hu N. In-Cell Nanoelectronics: Opening the Door to Intracellular Electrophysiology. NANO-MICRO LETTERS 2021; 13:127. [PMID: 34138366 PMCID: PMC8124030 DOI: 10.1007/s40820-021-00655-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/13/2021] [Indexed: 05/07/2023]
Abstract
Establishing a reliable electrophysiological recording platform is crucial for cardiology and neuroscience research. Noninvasive and label-free planar multitransistors and multielectrode arrays are conducive to perform the large-scale cellular electrical activity recordings, but the signal attenuation limits these extracellular devices to record subthreshold activities. In recent decade, in-cell nanoelectronics have been rapidly developed to open the door to intracellular electrophysiology. With the unique three-dimensional nanotopography and advanced penetration strategies, high-throughput and high-fidelity action potential like signal recordings is expected to be realized. This review summarizes in-cell nanoelectronics from versatile nano-biointerfaces, penetration strategies, active/passive nanodevices, systematically analyses the applications in electrogenic cells and especially evaluates the influence of nanodevices on the high-quality intracellular electrophysiological signals. Further, the opportunities, challenges and broad prospects of in-cell nanoelectronics are prospected, expecting to promote the development of in-cell electrophysiological platforms to meet the demand of theoretical investigation and clinical application.
Collapse
Affiliation(s)
- Dongxin Xu
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China
| | - Jingshan Mo
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China
| | - Xi Xie
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China
- The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
| | - Ning Hu
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China.
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China.
| |
Collapse
|
11
|
Iachetta G, Colistra N, Melle G, Deleye L, Tantussi F, De Angelis F, Dipalo M. Improving reliability and reducing costs of cardiotoxicity assessments using laser-induced cell poration on microelectrode arrays. Toxicol Appl Pharmacol 2021; 418:115480. [PMID: 33689843 DOI: 10.1016/j.taap.2021.115480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/07/2021] [Accepted: 03/02/2021] [Indexed: 10/22/2022]
Abstract
Drug-induced cardiotoxicity is a major barrier to drug development and a main cause of withdrawal of marketed drugs. Drugs can strongly alter the spontaneous functioning of the heart by interacting with the cardiac membrane ion channels. If these effects only surface during in vivo preclinical tests, clinical trials or worse after commercialization, the societal and economic burden will be significant and seriously hinder the efficient drug development process. Hence, cardiac safety pharmacology requires in vitro electrophysiological screening assays of all drug candidates to predict cardiotoxic effects before clinical trials. In the past 10 years, microelectrode array (MEA) technology began to be considered a valuable approach in pharmaceutical applications. However, an effective tool for high-throughput intracellular measurements, compatible with pharmaceutical standards, is not yet available. Here, we propose laser-induced optoacoustic poration combined with CMOS-MEA technology as a reliable and effective platform to detect cardiotoxicity. This approach enables the acquisition of high-quality action potential recordings from large numbers of cardiomyocytes within the same culture well, providing reliable data using single-well MEA devices and single cardiac syncytia per each drug. Thus, this technology could be applied in drug safety screening platforms reducing times and costs of cardiotoxicity assessments, while simultaneously improving the data reliability.
Collapse
Affiliation(s)
| | - Nicolò Colistra
- Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Giovanni Melle
- Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Lieselot Deleye
- Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | | | | | - Michele Dipalo
- Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy.
| |
Collapse
|
12
|
Automated feature extraction from large cardiac electrophysiological data sets. J Electrocardiol 2021; 65:157-162. [PMID: 33640635 DOI: 10.1016/j.jelectrocard.2021.02.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 01/19/2021] [Accepted: 02/11/2021] [Indexed: 12/28/2022]
Abstract
RATIONALE A new multi-electrode array-based application for the long-term recording of action potentials from electrogenic cells makes possible exciting cardiac electrophysiology studies in health and disease. With hundreds of simultaneous electrode recordings being acquired over a period of days, the main challenge becomes achieving reliable signal identification and quantification. OBJECTIVE We set out to develop an algorithm capable of automatically extracting regions of high-quality action potentials from terabyte size experimental results and to map the trains of action potentials into a low-dimensional feature space for analysis. METHODS AND RESULTS Our automatic segmentation algorithm finds regions of acceptable action potentials in large data sets of electrophysiological readings. We use spectral methods and support vector machines to classify our readings and to extract relevant features. We are able to show that action potentials from the same cell site can be recorded over days without detrimental effects to the cell membrane. The variability between measurements 24 h apart is comparable to the natural variability of the features at a single time point. CONCLUSIONS Our work contributes towards a non-invasive approach for cardiomyocyte functional maturation, as well as developmental, pathological and pharmacological studies. As the human-derived cardiac model tissue has the genetic makeup of its donor, a powerful tool for individual drug toxicity screening emerges.
Collapse
|
13
|
Visone R, Ugolini GS, Cruz-Moreira D, Marzorati S, Piazza S, Pesenti E, Redaelli A, Moretti M, Occhetta P, Rasponi M. Micro-electrode channel guide (µECG) technology: an online method for continuous electrical recording in a human beating heart-on-chip. Biofabrication 2021; 13. [PMID: 33561845 DOI: 10.1088/1758-5090/abe4c4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 02/09/2021] [Indexed: 12/19/2022]
Abstract
Cardiac toxicity still represents a common adverse outcome causing drug attrition and post-marketing withdrawal. The development of relevant in vitro models resembling the human heart recently opened the path towards a more accurate detection of drug-induced human cardiac toxicity early in the drug development process. Organs-on-chip (OoC) have been proposed as promising tools to recapitulate in vitro the key aspects of the in vivo cardiac physiology and to provide a means to directly analyze functional readouts. In this scenario, a new device capable of continuous monitoring of electrophysiological signals from functional in vitro human hearts-on-chip is here presented. The development of cardiac microtissues was achieved through a recently published method to control the mechanical environment, while the introduction of a technology consisting in micro-electrode coaxial guides (µECG) allowed to conduct direct and non-destructive electrophysiology studies. The generated human cardiac microtissues exhibited synchronous spontaneous beating, as demonstrated by multi-point and continuous acquisition of cardiac field potential, and expression of relevant genes encoding for cardiac ion-channels. A proof-of-concept pharmacological validation on 3 drugs proved the proposed model to potentially be a powerful tool to evaluate functional cardiac toxicity.
Collapse
Affiliation(s)
- Roberta Visone
- Politecnico di Milano Dipartimento di Elettronica Informazione e Bioingegneria, Via Ponzio 34/5, Milano, Lombardia, 20133, ITALY
| | - Giovanni Stefano Ugolini
- Politecnico di Milano Dipartimento di Elettronica Informazione e Bioingegneria, Via Ponzio 34/5, Milano, Lombardia, 20133, ITALY
| | - Daniela Cruz-Moreira
- Politecnico di Milano Dipartimento di Elettronica Informazione e Bioingegneria, Via Ponzio 34/5, Milano, Lombardia, 20133, ITALY
| | - Simona Marzorati
- Translational Medicine, Accelera Srl, via Pasteur, Nerviano, Nerviano, MI, 20100, ITALY
| | - Stefano Piazza
- BiomimX Srl, Via Giovanni Durando 38/A, Milan, 20158, ITALY
| | | | - Alberto Redaelli
- Politecnico di Milano Dipartimento di Elettronica Informazione e Bioingegneria, Via Ponzio 34/5, Milano, Lombardia, 20133, ITALY
| | - Matteo Moretti
- Cell and Tissue Engineering Lab, IRCCS Galeazzi Orthopaedic Institute, via R Galeazzi 4, Milan, 20161, ITALY
| | - Paola Occhetta
- Politecnico di Milano Dipartimento di Elettronica Informazione e Bioingegneria, Via Ponzio 34/5, Milano, Lombardia, 20133, ITALY
| | - Marco Rasponi
- Politecnico di Milano Dipartimento di Elettronica Informazione e Bioingegneria, Via Ponzio 34/5, Milano, Lombardia, 20133, ITALY
| |
Collapse
|
14
|
Sekhar Pagadala N. Computational prediction of hERG blockers using homology modelling, molecular docking and QuaSAR studies. RESULTS IN CHEMISTRY 2021. [DOI: 10.1016/j.rechem.2021.100101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
15
|
Kumar P, Ghosh A, Sundaresan L, Kathirvel P, Sankaranarayanan K, Chatterjee S. Ectopic release of nitric oxide modulates the onset of cardiac development in avian model. In Vitro Cell Dev Biol Anim 2020; 56:593-603. [PMID: 32959218 DOI: 10.1007/s11626-020-00495-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 08/10/2020] [Indexed: 01/01/2023]
Abstract
Heart development is one of the earliest developmental events, and its pumping action is directly linked to the intensity of development of other organs. Heart contractions mediate the circulation of the nutrients and signalling molecules to the focal points of developing embryos. In the present study, we used in vivo, ex vivo, in vitro, and in silico methods for chick embryo model to characterize and identify molecular targets under the influence of ectopic nitric oxide in reference to cardiogenesis. Spermine NONOate (SpNO) treatment of 10 μM increased the percentage of chick embryos having beating heart at 40th h of incubation by 2.2-fold (p < 0.001). In an ex vivo chick embryo culture, SpNO increased the percentage of embryos having beats by 1.56-fold (p < 0.05) compared with control after 2 h of treatment. Total body weight of SpNO-treated chick embryos at the Hamburger and Hamilton (HH) stage 29 was increased by 1.22-fold (p < 0.005). Cardiac field potential (FP) recordings of chick embryo at HH29 showed 2.5-fold (p < 0.001) increased in the amplitude, 3.2-fold (p < 0.001) increased in frequency of SpNO-treated embryos over that of the control group, whereas FP duration was unaffected. In cultured cardiac progenitors cells (CPCs), SpNO treatment decreased apoptosis and cell death by twofold (p < 0.001) and 1.7-fold (p < 0.001), respectively. Transcriptome analysis of chick embryonic heart isolated from HH15 stage pre-treated with SpNO at HH8 stage showed upregulation of genes involved in heart morphogenesis, heart contraction, cardiac cell development, calcium signalling, structure, and development whereas downregulated genes were enriched under the terms extracellular matrix, wnt pathway, and BMP pathway. The key upstream molecules predicted to be activated were p38 MAPK, MEF2C, TBX5, and GATA4 while KDM5α, DNMT3A, and HNF1α were predicted to be inhibited. This study suggests that the ectopic nitric oxide modulates the onset of cardiac development.
Collapse
Affiliation(s)
- Pavitra Kumar
- Vascular Biology Laboratory, AU-KBC Research Centre, M.I.T Campus of Anna University, Chromepet, Chennai, Tamil Nadu, 600044, India
| | - Anuran Ghosh
- Department of Biotechnology, Anna University, Chennai, Tamil Nadu, India
| | - Lakshmikirupa Sundaresan
- Vascular Biology Laboratory, AU-KBC Research Centre, M.I.T Campus of Anna University, Chromepet, Chennai, Tamil Nadu, 600044, India.,Department of Biotechnology, Anna University, Chennai, Tamil Nadu, India
| | | | | | - Suvro Chatterjee
- Vascular Biology Laboratory, AU-KBC Research Centre, M.I.T Campus of Anna University, Chromepet, Chennai, Tamil Nadu, 600044, India. .,Department of Biotechnology, Anna University, Chennai, Tamil Nadu, India.
| |
Collapse
|
16
|
Garrido A, Lepailleur A, Mignani SM, Dallemagne P, Rochais C. hERG toxicity assessment: Useful guidelines for drug design. Eur J Med Chem 2020; 195:112290. [PMID: 32283295 DOI: 10.1016/j.ejmech.2020.112290] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/27/2020] [Accepted: 03/27/2020] [Indexed: 02/06/2023]
Abstract
All along the drug development process, one of the most frequent adverse side effects, leading to the failure of drugs, is the cardiac arrhythmias. Such failure is mostly related to the capacity of the drug to inhibit the human ether-à-go-go-related gene (hERG) cardiac potassium channel. The early identification of hERG inhibition properties of biological active compounds has focused most of attention over the years. In order to prevent the cardiac side effects, a great number of in silico, in vitro and in vivo assays have been performed. The main goal of these studies is to understand the reasons of these effects, and then to give information or instructions to scientists involved in drug development to avoid the cardiac side effects. To evaluate anticipated cardiovascular effects, early evaluation of hERG toxicity has been strongly recommended for instance by the regulatory agencies such as U.S. Food and Drug Administration (FDA) and European Medicines Agency (EMA). Thus, following an initial screening of a collection of compounds to find hits, a great number of pharmacomodulation studies on the novel identified chemical series need to be performed including activity evaluation towards hERG. We provide in this concise review clear guidelines, based on described examples, illustrating successful optimization process to avoid hERG interactions as cases studies and to spur scientists to develop safe drugs.
Collapse
Affiliation(s)
- Amanda Garrido
- Normandie Univ, UNICAEN, Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), Caen, France
| | - Alban Lepailleur
- Normandie Univ, UNICAEN, Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), Caen, France
| | - Serge M Mignani
- UMR 860, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologique, Université Paris Descartes, PRES Sorbonne Paris Cité, CNRS, 45 rue des Saints Pères, 75006, Paris, France; CQM - Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9020-105, Funchal, Portugal
| | - Patrick Dallemagne
- Normandie Univ, UNICAEN, Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), Caen, France
| | - Christophe Rochais
- Normandie Univ, UNICAEN, Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), Caen, France.
| |
Collapse
|
17
|
|
18
|
Wei X, Qin C, Gu C, He C, Yuan Q, Liu M, Zhuang L, Wan H, Wang P. A novel bionic in vitro bioelectronic tongue based on cardiomyocytes and microelectrode array for bitter and umami detection. Biosens Bioelectron 2019; 145:111673. [PMID: 31546200 DOI: 10.1016/j.bios.2019.111673] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 08/22/2019] [Accepted: 09/02/2019] [Indexed: 12/27/2022]
Abstract
Electronic tongues (ETs) have been developed and widely used in food, beverage and pharmaceutical fields, but limited in sensitivity and specificity. In recent years, bioelectronic tongues (BioETs) integrating biological materials and various types of transducers are proposed to bridge the gap between ET system and biological taste. In this work, a bionic in vitro cell-based BioET is developed for bitter and umami detection, utilizing rat cardiomyocytes as a primary taste sensing element and microelectrode arrays (MEAs) as a secondary transducer for the first time. The primary cardiomyocytes of Sprague Dawley (SD) rats, which endogenously express bitter and umami taste receptors, were cultured on MEAs. Cells attached and grew well on the sensor surface, and syncytium was formed for potential conduction and mechanical beating, indicating the good biocompatibility of surface coating. The specificity of this BioET was verified by testing different tastants and bitter compounds. The results show that the BioET responds to bitter and umami compounds specifically among five basic tastants. For bitter recognition, only those can activate receptors in cardiomyocytes can be recognized by the BioET, and different bitter substances could be discriminated by principal component analysis (PCA). Moreover, the specific detections of two bitters (Denatonium Benzoate, Diphenidol) and an umami compound (Monosodium Glutamate) were realized with a detection limit of 10-6 M. The cardiomyocytes-based BioET proposed in this work provides a new approach for the construction of BioETs and has promising applications in taste detection and pharmaceutical study.
Collapse
Affiliation(s)
- Xinwei Wei
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China; State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Chunlian Qin
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Chenlei Gu
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Chuanjiang He
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Qunchen Yuan
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Mengxue Liu
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Liujing Zhuang
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Hao Wan
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China; State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Ping Wang
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China; State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, China.
| |
Collapse
|
19
|
Gu X, Yeung SY, Chadda A, Poon ENY, Boheler KR, Hsing IM. Organic Electrochemical Transistor Arrays for In Vitro Electrophysiology Monitoring of 2D and 3D Cardiac Tissues. ACTA ACUST UNITED AC 2018; 3:e1800248. [DOI: 10.1002/adbi.201800248] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 10/26/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Xi Gu
- Bioengineering Graduate Program; Department of Chemical and Biological Engineering; The Hong Kong University of Science and Technology; Hong Kong China
| | - Sin Yu Yeung
- Bioengineering Graduate Program; Department of Chemical and Biological Engineering; The Hong Kong University of Science and Technology; Hong Kong China
| | - Akriti Chadda
- Bioengineering Graduate Program; Department of Chemical and Biological Engineering; The Hong Kong University of Science and Technology; Hong Kong China
| | - Ellen Ngar Yun Poon
- The Stem Cell and Regenerative Consortium; The University of Hong Kong; Hong Kong China
| | - Kenneth R. Boheler
- The Stem Cell and Regenerative Consortium; The University of Hong Kong; Hong Kong China
- The Department of Biomedical Engineering; Johns Hopkins University; Baltimore MD 21205 USA
| | - I.-Ming Hsing
- Bioengineering Graduate Program; Department of Chemical and Biological Engineering; The Hong Kong University of Science and Technology; Hong Kong China
| |
Collapse
|
20
|
Kumar P, Kumar HA, Sundaresan L, Ghosh A, Kathirvel P, Thilak A, Katakia YT, Sankaranarayanan K, Chatterjee S. Thalidomide remodels developing heart in chick embryo: discovery of a thalidomide mediated hematoma in heart muscle. Naunyn Schmiedebergs Arch Pharmacol 2018; 391:1093-1105. [PMID: 29982937 DOI: 10.1007/s00210-018-1532-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 06/22/2018] [Indexed: 12/28/2022]
Abstract
Despite of medical disaster caused by thalidomide in 1960s, the drug came to clinical use again for the treatment of erythema nodosum leprosum (ENL) and multiple myeloma. Recently, a new generation of children affected by thalidomide intake by their mothers during pregnancy has been identified in Brazil. In the past few years, there is the great enhancement in our understanding of the molecular mechanisms and targets of thalidomide with the help of modern OMICS technologies. However, understanding of cardiac-specific anomalies in fetus due to thalidomide intake by the respective mother has not been explored fully. At organ level, thalidomide causes congenital heart diseases, limb deformities in addition to ocular, and neural and ear abnormalities. The period of morning sickness and cardiogenesis is synchronized in pregnant women. Therefore, thalidomide intake during the first trimester could affect cardiogenesis severely. Thalidomide intake in pregnant women either causes miscarriage or heart abnormalities such as patent ductus arteriosus, ventricular septal defect (VSD), atrial septal defect (ASD), and pulmonary stenosis in survivors. In the present study, we identified a novel morphological defect (lump) in the heart of thalidomide-treated chick embryos. We characterized the lump at morphological, histo-pathological, oxidative stress, electro-physiological, and gene expression level. To our knowledge, here, we report the very first electrophysiological characterization of embryonic heart affected by thalidomide treatment.
Collapse
Affiliation(s)
- Pavitra Kumar
- Vascular Biology Laboratory, AU-KBC Research Centre, Chennai, Tamil Nadu, India
| | - Harish A Kumar
- Department of Biotechnology, Anna University, Chennai, Tamil Nadu, India
| | - Lakshmikirupa Sundaresan
- Vascular Biology Laboratory, AU-KBC Research Centre, Chennai, Tamil Nadu, India.,Department of Biotechnology, Anna University, Chennai, Tamil Nadu, India
| | - Anuran Ghosh
- Department of Biotechnology, Anna University, Chennai, Tamil Nadu, India
| | | | - Apurva Thilak
- Department of Biotechnology, Anna University, Chennai, Tamil Nadu, India
| | - Yash T Katakia
- Vascular Biology Laboratory, AU-KBC Research Centre, Chennai, Tamil Nadu, India.,Department of Biotechnology, Anna University, Chennai, Tamil Nadu, India
| | | | - Suvro Chatterjee
- Vascular Biology Laboratory, AU-KBC Research Centre, Chennai, Tamil Nadu, India. .,Department of Biotechnology, Anna University, Chennai, Tamil Nadu, India. .,AU-KBC Research Centre, M.I.T Campus of Anna University, Chromepet, Chennai, 600044, India.
| |
Collapse
|
21
|
Graphene Microelectrode Arrays for Electrical and Optical Measurements of Human Stem Cell-Derived Cardiomyocytes. Cell Mol Bioeng 2018; 11:407-418. [PMID: 31719891 DOI: 10.1007/s12195-018-0525-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 04/26/2018] [Indexed: 12/17/2022] Open
Abstract
Introduction Cell-cell communication plays a pivotal role in biological systems' coordination and function. Electrical properties have been linked to specification and differentiation of stem cells into targeted progeny, such as neurons and cardiomyocytes. Currently, there is a critical need in developing new ways to complement fluorescent indicators, such as Ca2+-sensitive dyes, for direct electrophysiological measurements of cells and tissue. Here, we report a unique transparent and biocompatible graphene-based electrical platform that enables electrical and optical investigation of human embryonic stem cell-derived cardiomyocytes' (hESC-CMs) intracellular processes and intercellular communication. Methods Graphene, a honeycomb sp2 hybridized two-dimensional carbon lattice, was synthesized using low pressure chemical vapor deposition system, and was tested for biocompatibility. Au and graphene microelectrode arrays (MEAs) were fabricated using well-established microfabrication methods. Au and graphene MEAs were interfaced with hESC-CMs to perform both optical and electrical recordings. Results Optical imaging and Raman spectroscopy confirmed the presence of monolayer graphene. Viability assay showed biocompatibility of graphene. Electrochemical characterization proved graphene's functional activity. Nitric acid treatment further enhanced the electrochemical properties of graphene. Graphene electrodes' transparency enabled both optical and electrical recordings from hESC-CMs. Graphene MEA detected changes in beating frequency and field potential duration upon β-adrenergic receptor agonist treatment. Conclusion The transparent graphene platform enables the investigation of both intracellular and intercellular communication processes and will create new avenues for bidirectional communication (sensing and stimulation) with electrically active tissues and will set the ground for investigations reported diseases such as Alzheimer, Parkinson's disease and arrhythmias.
Collapse
|
22
|
A biopotential optrode array: operation principles and simulations. Sci Rep 2018; 8:2690. [PMID: 29426924 PMCID: PMC5807498 DOI: 10.1038/s41598-018-20182-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 12/21/2017] [Indexed: 01/11/2023] Open
Abstract
We propose an optical electrode ’optrode’ sensor array for biopotential measurements. The transduction mechanism is based on deformed helix ferroelectric liquid crystals which realign, altering the optrode’s light reflectance properties, relative to applied potential fields of biological cells and tissue. A computational model of extracellular potential recording by the optrode including the electro-optical transduction mechanism is presented, using a combination of time-domain and frequency-domain finite element analysis. Simulations indicate that the device has appropriate temporal response to faithfully transduce neuronal spikes, and spatial resolution to capture impulse propagation along a single neuron. These simulations contribute to the development of multi-channel optrode arrays for spatio-temporal mapping of electric events in excitable biological tissue.
Collapse
|
23
|
Tixier E, Raphel F, Lombardi D, Gerbeau JF. Composite Biomarkers Derived from Micro-Electrode Array Measurements and Computer Simulations Improve the Classification of Drug-Induced Channel Block. Front Physiol 2018; 8:1096. [PMID: 29354067 PMCID: PMC5762138 DOI: 10.3389/fphys.2017.01096] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 12/13/2017] [Indexed: 12/19/2022] Open
Abstract
The Micro-Electrode Array (MEA) device enables high-throughput electrophysiology measurements that are less labor-intensive than patch-clamp based techniques. Combined with human-induced pluripotent stem cells cardiomyocytes (hiPSC-CM), it represents a new and promising paradigm for automated and accurate in vitro drug safety evaluation. In this article, the following question is addressed: which features of the MEA signals should be measured to better classify the effects of drugs? A framework for the classification of drugs using MEA measurements is proposed. The classification is based on the ion channels blockades induced by the drugs. It relies on an in silico electrophysiology model of the MEA, a feature selection algorithm and automatic classification tools. An in silico model of the MEA is developed and is used to generate synthetic measurements. An algorithm that extracts MEA measurements features designed to perform well in a classification context is described. These features are called composite biomarkers. A state-of-the-art machine learning program is used to carry out the classification of drugs using experimental MEA measurements. The experiments are carried out using five different drugs: mexiletine, flecainide, diltiazem, moxifloxacin, and dofetilide. We show that the composite biomarkers outperform the classical ones in different classification scenarios. We show that using both synthetic and experimental MEA measurements improves the robustness of the composite biomarkers and that the classification scores are increased.
Collapse
Affiliation(s)
- Eliott Tixier
- Inria Paris, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie-Paris 6, UMR 7598 LJLL, Paris, France
| | - Fabien Raphel
- Inria Paris, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie-Paris 6, UMR 7598 LJLL, Paris, France
| | - Damiano Lombardi
- Inria Paris, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie-Paris 6, UMR 7598 LJLL, Paris, France
| | - Jean-Frédéric Gerbeau
- Inria Paris, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie-Paris 6, UMR 7598 LJLL, Paris, France
| |
Collapse
|
24
|
Wang YI, Carmona C, Hickman JJ, Shuler ML. Multiorgan Microphysiological Systems for Drug Development: Strategies, Advances, and Challenges. Adv Healthc Mater 2018; 7:10.1002/adhm.201701000. [PMID: 29205920 PMCID: PMC5805562 DOI: 10.1002/adhm.201701000] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 09/18/2017] [Indexed: 12/19/2022]
Abstract
Traditional cell culture and animal models utilized for preclinical drug screening have led to high attrition rates of drug candidates in clinical trials due to their low predictive power for human response. Alternative models using human cells to build in vitro biomimetics of the human body with physiologically relevant organ-organ interactions hold great potential to act as "human surrogates" and provide more accurate prediction of drug effects in humans. This review is a comprehensive investigation into the development of tissue-engineered human cell-based microscale multiorgan models, or multiorgan microphysiological systems for drug testing. The evolution from traditional models to macro- and microscale multiorgan systems is discussed in regards to the rationale for recent global efforts in multiorgan microphysiological systems. Current advances in integrating cell culture and on-chip analytical technologies, as well as proof-of-concept applications for these multiorgan microsystems are discussed. Major challenges for the field, such as reproducibility and physiological relevance, are discussed with comparisons of the strengths and weaknesses of various systems to solve these challenges. Conclusions focus on the current development stage of multiorgan microphysiological systems and new trends in the field.
Collapse
Affiliation(s)
- Ying I Wang
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Carlos Carmona
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | - James J Hickman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
- Hesperos, Inc., 3259 Progress Dr, Room 158, Orlando, FL 32826
| | - Michael L Shuler
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
- Hesperos, Inc., 3259 Progress Dr, Room 158, Orlando, FL 32826
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
25
|
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
|
26
|
Schneider JJ. Vertically Aligned Carbon Nanotubes as Platform for Biomimetically Inspired Mechanical Sensing, Bioactive Surfaces, and Electrical Cell Interfacing. ACTA ACUST UNITED AC 2017; 1:e1700101. [PMID: 32646166 DOI: 10.1002/adbi.201700101] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 07/04/2017] [Indexed: 12/30/2022]
Abstract
Vertically aligned carbon nanotubes (VACNTs) are one dimensional carbon objects anchored atop of a solid substrate. They are geometrically fixed in contrast to their counterparts, randomly oriented carbon nanotubes (CNTs). In this progress report, the breadth in which these one dimensional, mechanically flexible, though robust and electrical conducting carbon nanostructures can be employed as functional material is shown and our research is put in perspective to work in the last five to ten years. The connection between the different areas touched in this report is the biomimetic-materials approach, which rely on the hairy morphology of VACNTs. These properties in connection with their electrical conductivity offer possibilities towards new functional features and applications of VACNTs. To appreciate the possibilities of biomimetic research with VACNTs, first their material characteristics are given to make the reader familiar with specific features of their synthesis, the peculiarities in arranging and controlling the morphology of CNTs in a vertical alignment as well as a current understanding of these properties on a microscopic basis. In doing so, similarities as well as differences, which offer new possibilities for biomimetic studies of VACNTS with respect to multiwalled randomly oriented CNTs, will become clear.
Collapse
Affiliation(s)
- Jörg J Schneider
- Eduard-Zintl-Institut für Anorganische und Physikalische Chemie, Technische Universität Darmstadt, Alarich-Weiss Str. 12, 64287, Darmstadt, Germany
| |
Collapse
|
27
|
Li X, Zhang Y, Li H, Zhao Y. Modeling of the hERG K+ Channel Blockage Using Online Chemical Database and Modeling Environment (OCHEM). Mol Inform 2017; 36. [PMID: 28857516 DOI: 10.1002/minf.201700074] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 08/08/2017] [Indexed: 11/06/2022]
Abstract
Human ether-a-go-go related gene (hERG) K+ channel plays an important role in cardiac action potential. Blockage of hERG channel may result in long QT syndrome (LQTS), even cause sudden cardiac death. Many drugs have been withdrawn from the market because of the serious hERG-related cardiotoxicity. Therefore, it is quite essential to estimate the chemical blockage of hERG in the early stage of drug discovery. In this study, a diverse set of 3721 compounds with hERG inhibition data was assembled from literature. Then, we make full use of the Online Chemical Modeling Environment (OCHEM), which supplies rich machine learning methods and descriptor sets, to build a series of classification models for hERG blockage. We also generated two consensus models based on the top-performing individual models. The consensus models performed much better than the individual models both on 5-fold cross validation and external validation. Especially, consensus model II yielded the prediction accuracy of 89.5 % and MCC of 0.670 on external validation. This result indicated that the predictive power of consensus model II should be stronger than most of the previously reported models. The 17 top-performing individual models and the consensus models and the data sets used for model development are available at https://ochem.eu/article/103592.
Collapse
Affiliation(s)
- Xiao Li
- Beijing Computing Center, Beijing Academy of Science and Technology, 7 Fengxian road, Beijing, 100094, China.,Beijing Beike Deyuan Bio-Pharm Technology Co.Ltd, 7 Fengxian road, Beijing, 100094, China
| | - Yuan Zhang
- Beijing Beike Deyuan Bio-Pharm Technology Co.Ltd, 7 Fengxian road, Beijing, 100094, China
| | - Huanhuan Li
- Beijing Beike Deyuan Bio-Pharm Technology Co.Ltd, 7 Fengxian road, Beijing, 100094, China
| | - Yong Zhao
- Beijing Computing Center, Beijing Academy of Science and Technology, 7 Fengxian road, Beijing, 100094, China.,Beijing Beike Deyuan Bio-Pharm Technology Co.Ltd, 7 Fengxian road, Beijing, 100094, China
| |
Collapse
|
28
|
Lapp H, Bruegmann T, Malan D, Friedrichs S, Kilgus C, Heidsieck A, Sasse P. Frequency-dependent drug screening using optogenetic stimulation of human iPSC-derived cardiomyocytes. Sci Rep 2017; 7:9629. [PMID: 28851973 PMCID: PMC5575076 DOI: 10.1038/s41598-017-09760-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 07/31/2017] [Indexed: 11/29/2022] Open
Abstract
Side effects on cardiac ion channels are one major reason for new drugs to fail during preclinical evaluation. Herein we propose a simple optogenetic screening tool measuring extracellular field potentials (FP) from paced cardiomyocytes to identify drug effects over the whole physiological heart range, which is essential given the rate-dependency of ion channel function and drug action. Human induced pluripotent stem cell-derived cardiomyocytes were transduced with an adeno-associated virus to express Channelrhodopsin2 and plated on micro-electrode arrays. Global pulsed illumination (470 nm, 1 ms, 0.9 mW/mm2) was applied at frequencies from 1 to 2.5 Hz, which evoked FP simultaneously in all cardiomyocytes. This synchronized activation allowed averaging of FP from all electrodes resulting in one robust FP signal for analysis. Field potential duration (FPD) was ~25% shorter at 2.5 Hz compared to 1 Hz. Inhibition of hERG channels prolonged FPD only at low heart rates whereas Ca2+ channel block shortened FPD at all heart rates. Optogenetic pacing also allowed analysis of the maximum downstroke velocity of the FP to detect drug effects on Na+ channel availability. In principle, the presented method is well scalable for high content cardiac toxicity screening or personalized medicine for inherited cardiac channelopathies.
Collapse
Affiliation(s)
- Hendrik Lapp
- Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Sigmund-Freud-Str. 25, 53127, Bonn, Germany
| | - Tobias Bruegmann
- Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Sigmund-Freud-Str. 25, 53127, Bonn, Germany
- Research Training Group 1873, University of Bonn, 53127, Bonn, Germany
| | - Daniela Malan
- Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Sigmund-Freud-Str. 25, 53127, Bonn, Germany
| | - Stephanie Friedrichs
- Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Sigmund-Freud-Str. 25, 53127, Bonn, Germany
| | - Carsten Kilgus
- Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Sigmund-Freud-Str. 25, 53127, Bonn, Germany
| | - Alexandra Heidsieck
- Zentralinstitut für Medizintechnik, Technische Universität München, München, Germany
| | - Philipp Sasse
- Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Sigmund-Freud-Str. 25, 53127, Bonn, Germany.
| |
Collapse
|
29
|
Abi-Gerges N, Pointon A, Oldman KL, Brown MR, Pilling MA, Sefton CE, Garside H, Pollard CE. Assessment of extracellular field potential and Ca2+ transient signals for early QT/pro-arrhythmia detection using human induced pluripotent stem cell-derived cardiomyocytes. J Pharmacol Toxicol Methods 2017; 83:1-15. [DOI: 10.1016/j.vascn.2016.09.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 09/02/2016] [Accepted: 09/08/2016] [Indexed: 12/29/2022]
|
30
|
Influence of field potential duration on spontaneous beating rate of human induced pluripotent stem cell-derived cardiomyocytes: Implications for data analysis and test system selection. J Pharmacol Toxicol Methods 2016; 82:74-82. [DOI: 10.1016/j.vascn.2016.08.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 07/19/2016] [Accepted: 08/04/2016] [Indexed: 11/15/2022]
|
31
|
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
|
32
|
|
33
|
Chavan S, Abdelaziz A, Wiklander JG, Nicholls IA. A k-nearest neighbor classification of hERG K(+) channel blockers. J Comput Aided Mol Des 2016; 30:229-36. [PMID: 26860111 PMCID: PMC4802000 DOI: 10.1007/s10822-016-9898-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 01/28/2016] [Indexed: 01/08/2023]
Abstract
A series of 172 molecular structures that block the hERG K+ channel were used to develop a classification model where, initially, eight types of PaDEL fingerprints were used for k-nearest neighbor model development. A consensus model constructed using Extended-CDK, PubChem and Substructure count fingerprint-based models was found to be a robust predictor of hERG activity. This consensus model demonstrated sensitivity and specificity values of 0.78 and 0.61 for the internal dataset compounds and 0.63 and 0.54 for the external (PubChem) dataset compounds, respectively. This model has identified the highest number of true positives (i.e. 140) from the PubChem dataset so far, as compared to other published models, and can potentially serve as a basis for the prediction of hERG active compounds. Validating this model against FDA-withdrawn substances indicated that it may even be useful for differentiating between mechanisms underlying QT prolongation.
Collapse
Affiliation(s)
- Swapnil Chavan
- Bioorganic and Biophysical Chemistry Laboratory, Department of Chemistry and Biomedical Sciences, Linnaeus University Centre for Biomaterials Chemistry, Linnaeus University, 391 82, Kalmar, Sweden.
| | - Ahmed Abdelaziz
- eADMET GmbH, Lichtenbergstraße 8, 85748, Garching, Munich, Germany
| | - Jesper G Wiklander
- Bioorganic and Biophysical Chemistry Laboratory, Department of Chemistry and Biomedical Sciences, Linnaeus University Centre for Biomaterials Chemistry, Linnaeus University, 391 82, Kalmar, Sweden
| | - Ian A Nicholls
- Bioorganic and Biophysical Chemistry Laboratory, Department of Chemistry and Biomedical Sciences, Linnaeus University Centre for Biomaterials Chemistry, Linnaeus University, 391 82, Kalmar, Sweden. .,Department of Chemistry-BMC, Uppsala University, Box 576, 751 23, Uppsala, Sweden.
| |
Collapse
|
34
|
Maysinger D, Ji J, Hutter E, Cooper E. Nanoparticle-Based and Bioengineered Probes and Sensors to Detect Physiological and Pathological Biomarkers in Neural Cells. Front Neurosci 2015; 9:480. [PMID: 26733793 PMCID: PMC4683200 DOI: 10.3389/fnins.2015.00480] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 11/30/2015] [Indexed: 01/11/2023] Open
Abstract
Nanotechnology, a rapidly evolving field, provides simple and practical tools to investigate the nervous system in health and disease. Among these tools are nanoparticle-based probes and sensors that detect biochemical and physiological properties of neurons and glia, and generate signals proportionate to physical, chemical, and/or electrical changes in these cells. In this context, quantum dots (QDs), carbon-based structures (C-dots, grapheme, and nanodiamonds) and gold nanoparticles are the most commonly used nanostructures. They can detect and measure enzymatic activities of proteases (metalloproteinases, caspases), ions, metabolites, and other biomolecules under physiological or pathological conditions in neural cells. Here, we provide some examples of nanoparticle-based and genetically engineered probes and sensors that are used to reveal changes in protease activities and calcium ion concentrations. Although significant progress in developing these tools has been made for probing neural cells, several challenges remain. We review many common hurdles in sensor development, while highlighting certain advances. In the end, we propose some future directions and ideas for developing practical tools for neural cell investigations, based on the maxim "Measure what is measurable, and make measurable what is not so" (Galileo Galilei).
Collapse
Affiliation(s)
- Dusica Maysinger
- Department of Pharmacology and Therapeutics, McGill University Montreal, QC, Canada
| | - Jeff Ji
- Department of Pharmacology and Therapeutics, McGill University Montreal, QC, Canada
| | - Eliza Hutter
- Department of Pharmacology and Therapeutics, McGill University Montreal, QC, Canada
| | - Elis Cooper
- Department of Physiology, McGill University Montreal, QC, Canada
| |
Collapse
|
35
|
Cardiotoxicity screening: a review of rapid-throughput in vitro approaches. Arch Toxicol 2015; 90:1803-16. [PMID: 26676948 DOI: 10.1007/s00204-015-1651-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 11/18/2015] [Indexed: 01/07/2023]
Abstract
Cardiac toxicity represents one of the leading causes of drug failure along different stages of drug development. Multiple very successful pharmaceuticals had to be pulled from the market or labeled with strict usage warnings due to adverse cardiac effects. In order to protect clinical trial participants and patients, the International Conference on Harmonization published guidelines to recommend that all new drugs to be tested preclinically for hERG (Kv11.1) channel sensitivity before submitting for regulatory reviews. However, extensive studies have demonstrated that measurement of hERG activity has limitations due to the multiple molecular targets of drug compound through which it may mitigate or abolish a potential arrhythmia, and therefore, a model measuring multiple ion channel effects is likely to be more predictive. Several phenotypic rapid-throughput methods have been developed to predict the potential cardiac toxic compounds in the early stages of drug development using embryonic stem cells- or human induced pluripotent stem cell-derived cardiomyocytes. These rapid-throughput methods include microelectrode array-based field potential assay, impedance-based or Ca(2+) dynamics-based cardiomyocytes contractility assays. This review aims to discuss advantages and limitations of these phenotypic assays for cardiac toxicity assessment.
Collapse
|
36
|
Harris K. A Human Induced Pluripotent Stem Cell−Derived Cardiomyocyte (hiPSC‐CM) Multielectrode Array Assay for Preclinical Cardiac Electrophysiology Safety Screening. ACTA ACUST UNITED AC 2015; 71:11.18.1-11.18.15. [DOI: 10.1002/0471141755.ph1118s71] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Kate Harris
- Safety Pharmacology, GlaxoSmithKline, David Jack Centre for R&D, Ware Hertfordshire United Kingdom
| |
Collapse
|
37
|
Abstract
Attrition due to nonclinical safety represents a major issue for the productivity of pharmaceutical research and development (R&D) organizations, especially during the compound optimization stages of drug discovery and the early stages of clinical development. Focusing on decreasing nonclinical safety-related attrition is not a new concept, and various approaches have been experimented with over the last two decades. Front-loading testing funnels in Discovery with in vitro toxicity assays designed to rapidly identify unfavorable molecules was the approach adopted by most pharmaceutical R&D organizations a few years ago. However, this approach has also a non-negligible opportunity cost. Hence, significant refinements to the "fail early, fail often" paradigm have been proposed recently to reflect the complexity of accurately categorizing compounds with early data points without taking into account other important contextual aspects, in particular efficacious systemic and tissue exposures. This review provides an overview of toxicology approaches and models that can be used in pharmaceutical Discovery at the series/lead identification and lead optimization stages to guide and inform chemistry efforts, as well as a personal view on how to best use them to meet nonclinical safety-related attrition objectives consistent with a sustainable pharmaceutical R&D model. The scope of this review is limited to small molecules, as large molecules are associated with challenges that are quite different. Finally, a perspective on how several emerging technologies may impact toxicity evaluation is also provided.
Collapse
Affiliation(s)
- Eric A G Blomme
- Global Preclinical Safety, AbbVie Inc. , 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Yvonne Will
- Drug Safety Research and Development, Pfizer , Eastern Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|
38
|
An improved functional assay for rapid detection of marine toxins, saxitoxin and brevetoxin using a portable cardiomyocyte-based potential biosensor. Biosens Bioelectron 2015; 72:10-7. [DOI: 10.1016/j.bios.2015.04.028] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 03/31/2015] [Accepted: 04/12/2015] [Indexed: 11/21/2022]
|
39
|
Asakura K, Hayashi S, Ojima A, Taniguchi T, Miyamoto N, Nakamori C, Nagasawa C, Kitamura T, Osada T, Honda Y, Kasai C, Ando H, Kanda Y, Sekino Y, Sawada K. Improvement of acquisition and analysis methods in multi-electrode array experiments with iPS cell-derived cardiomyocytes. J Pharmacol Toxicol Methods 2015; 75:17-26. [DOI: 10.1016/j.vascn.2015.04.002] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 03/30/2015] [Accepted: 04/15/2015] [Indexed: 10/23/2022]
|
40
|
Frieß JL, Heselich A, Ritter S, Haber A, Kaiser N, Layer PG, Thielemann C. Electrophysiologic and cellular characteristics of cardiomyocytes after X-ray irradiation. Mutat Res 2015; 777:1-10. [PMID: 25912077 DOI: 10.1016/j.mrfmmm.2015.03.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 03/24/2015] [Accepted: 03/26/2015] [Indexed: 06/04/2023]
Abstract
The aim of this study was to investigate possible effects of ionizing irradiation on the electrophysiological functionality of cardiac myocytes in vitro. Primary chicken cardiomyocytes with spontaneous beating activity were irradiated with X-rays (dose range of 0.5-7 Gy). Functional alterations of cardiac cell cultures were evaluated up to 7 days after irradiation using microelectrode arrays. As examined endpoints, cell proliferation, apoptosis, reactive oxygen species (ROS) and DNA damage were evaluated. The beat rate of the cardiac networks increased in a dose-dependent manner over one week. The duration of single action potentials was slightly shortened. Additionally, we observed lower numbers of mitotic and S-phase cells at certain time points after irradiation. Also, the number of cells with γH2AX foci increased as a function of the dose. No significant changes in the level of ROS were detected. Induction of apoptosis was generally negligibly low. This is the first report to directly show alterations in cardiac electrophysiology caused by ionizing radiation, which were detectable up to one week after irradiation.
Collapse
Affiliation(s)
- Johannes L Frieß
- University for Applied Sciences Aschaffenburg, biomems lab, Würzburger Straße 45, 63743 Aschaffenburg, Germany.
| | - Anja Heselich
- Technische Universität Darmstadt, Developmental Biology and Neurogenetics, Schnittspahnstraße 13, 64287 Darmstadt, Germany
| | - Sylvia Ritter
- Helmholtz Institute for Heavy Ion Research (GSI), Biophysics Department, Planckstraße 1, 64291 Darmstadt, Germany
| | - Angelina Haber
- Technische Universität Darmstadt, Developmental Biology and Neurogenetics, Schnittspahnstraße 13, 64287 Darmstadt, Germany
| | - Nicole Kaiser
- Technische Universität Darmstadt, Developmental Biology and Neurogenetics, Schnittspahnstraße 13, 64287 Darmstadt, Germany
| | - Paul G Layer
- Technische Universität Darmstadt, Developmental Biology and Neurogenetics, Schnittspahnstraße 13, 64287 Darmstadt, Germany
| | - Christiane Thielemann
- University for Applied Sciences Aschaffenburg, biomems lab, Würzburger Straße 45, 63743 Aschaffenburg, Germany
| |
Collapse
|
41
|
32-Channel System to Measure the Electrophysiological Properties of Bioengineered Cardiac Muscle. IEEE Trans Biomed Eng 2015; 62:1614-22. [DOI: 10.1109/tbme.2015.2399437] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
42
|
Stancescu M, Molnar P, McAleer CW, McLamb W, Long CJ, Oleaga C, Prot JM, Hickman JJ. A phenotypic in vitro model for the main determinants of human whole heart function. Biomaterials 2015; 60:20-30. [PMID: 25978005 DOI: 10.1016/j.biomaterials.2015.04.035] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 04/09/2015] [Accepted: 04/14/2015] [Indexed: 11/24/2022]
Abstract
This article details the construction and testing of a phenotypic assay system that models in vivo cardiac function in a parallel in vitro environment with human stem cell derived cardiomyocytes. The major determinants of human whole-heart function were experimentally modeled by integrating separate 2D cellular systems with BioMicroelectromechanical Systems (BioMEMS) constructs. The model features a serum-free defined medium to enable both acute and chronic evaluation of drugs and toxins. The integration of data from both systems produced biologically relevant predictions of cardiac function in response to varying concentrations of selected drugs. Sotalol, norepinephrine and verapamil were shown to affect the measured parameters according to their specific mechanism of action, in agreement with clinical data. This system is applicable for cardiac side effect assessment, general toxicology, efficacy studies, and evaluation of in vitro cellular disease models in body-on-a-chip systems.
Collapse
Affiliation(s)
- Maria Stancescu
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Peter Molnar
- Department of Zoology, University of West Hungary, Szombathely H-9700, Hungary; Biomedical Engineering, Cornell University, Department of Biomedical Engineering, 115 Weill Hall, Ithaca, NY 14853, USA
| | - Christopher W McAleer
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - William McLamb
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Christopher J Long
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Carlota Oleaga
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Jean-Matthieu Prot
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; Biomedical Engineering, Cornell University, Department of Biomedical Engineering, 115 Weill Hall, Ithaca, NY 14853, USA
| | - James J Hickman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA.
| |
Collapse
|
43
|
Kunze A, Steel D, Dahlenborg K, Sartipy P, Svedhem S. Non-Invasive Acoustical sensing of Drug-Induced Effects on the Contractile Machinery of Human Cardiomyocyte Clusters. PLoS One 2015; 10:e0125540. [PMID: 25961711 PMCID: PMC4427273 DOI: 10.1371/journal.pone.0125540] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 03/25/2015] [Indexed: 11/19/2022] Open
Abstract
There is an urgent need for improved models for cardiotoxicity testing. Here we propose acoustic sensing applied to beating human cardiomyocyte clusters for non-invasive, surrogate measuring of the QT interval and other characteristics of the contractile machinery. In experiments with the acoustic method quartz crystal microbalance with dissipation monitoring (QCM-D), the shape of the recorded signals was very similar to the extracellular field potential detected in electrochemical experiments, and the expected changes of the QT interval in response to addition of conventional drugs (E-4031 or nifedipine) were observed. Additionally, changes in the dissipation signal upon addition of cytochalasin D were in good agreement with the known, corresponding shortening of the contraction-relaxation time. These findings suggest that QCM-D has great potential as a tool for cardiotoxicological screening, where effects of compounds on the cardiomyocyte contractile machinery can be detected independently of whether the extracellular field potential is altered or not.
Collapse
Affiliation(s)
- Angelika Kunze
- Department of Applied Physics, Chalmers University of Technology, Göteborg, Sweden
| | | | | | - Peter Sartipy
- Cellectis AB, Göteborg, Sweden
- Systems Biology Research Center, School of Bioscience, University of Skövde, Skövde, Sweden
| | - Sofia Svedhem
- Department of Applied Physics, Chalmers University of Technology, Göteborg, Sweden
- * E-mail:
| |
Collapse
|
44
|
Molecularly engineered graphene surfaces for sensing applications: A review. Anal Chim Acta 2015; 859:1-19. [DOI: 10.1016/j.aca.2014.07.031] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 07/09/2014] [Accepted: 07/20/2014] [Indexed: 11/23/2022]
|
45
|
Shim W, Yu H, Ja KPMM, Parasuram M, Lim KP, Wong P. Electrotonic coupled metabolic purification of chick cardiomyocytes. Methods Mol Biol 2015; 1299:167-175. [PMID: 25836584 DOI: 10.1007/978-1-4939-2572-8_14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Cardiomyocytes isolated from chick and rodent are widely used in studying cardiac physiology. However, contaminating non-cardiomyocytes are an inherent problem that hinders downstream analysis. Here, we report a novel electrical stimulation coupled with metabolic selection method using cytosine arabinoside (AraC) to efficiently eliminate contaminating cells in isolating chick embryonic cardiomyocytes. Compared with conventional methods of pre-plating or AraC alone, electrical stimulation coupled with AraC increased the percentage purity of cardiomyocytes by 2-6-fold with added effect of improved contractile function and maturation. This simple method could be useful in isolating and maintaining purified cardiomyocytes for long-term studies of cardiac physiology.
Collapse
Affiliation(s)
- Winston Shim
- Stem Cell Laboratory, National Heart Research Institute Singapore (NHRIS), National Heart Centre Singapore (NHCS), 5 Hospital Drive, 9th Floor, Singapore, 169609, Singapore,
| | | | | | | | | | | |
Collapse
|
46
|
Image-based evaluation of contraction–relaxation kinetics of human-induced pluripotent stem cell-derived cardiomyocytes: Correlation and complementarity with extracellular electrophysiology. J Mol Cell Cardiol 2014; 77:178-91. [DOI: 10.1016/j.yjmcc.2014.09.010] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Accepted: 09/10/2014] [Indexed: 01/05/2023]
|
47
|
Kienast R, Stöger M, Handler M, Hanser F, Baumgartner C. Alterations of field potentials in isotropic cardiomyocyte cell layers induced by multiple endogenous pacemakers under normal and hypothermal conditions. Am J Physiol Heart Circ Physiol 2014; 307:H1013-23. [DOI: 10.1152/ajpheart.00097.2014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The use of autonomous contracting randomly grown cardiomyocyte monolayers cultivated on microelectrode arrays (MEAs) represents an accepted experimental setting for preclinical experimental research in the field of cardiac electrophysiology. A dominant pacemaker forces a monolayer to adhere to a regular and synchronized contraction. Randomly distributed multiple pacemakers interfere with this dominant center, resulting in more or less frequent changes of propagation direction. This study aims to characterize the impact of changing propagation directions at single electrodes of the MEA on the four intrinsic parameters of registered field potentials (FPs) FPrise, FPMIN, FPpre, and FPdur and conduction velocity (CV) under normal and hypothermal conditions. Primary cultures of chicken cardiomyocytes ( n = 18) were plated directly onto MEAs and FPs were recorded in a temperature range between 37 and 29°C. The number and spatiotemporal distribution of biological and artificial pacemakers of each cell layer inside and outside of the MEA registration area were evaluated using an algorithm developed in-house. In almost every second myocardial cell layer, interfering autonomous pacemakers were detected at stable temperatures, showing random spatial distributions with similar beating rates. Additionally, a temperature-dependent change of the dominant pacemaker center was observed in n = 16 experiments. A significant spread-direction-dependent variation of CV, FPrise, FPMIN, and FPpre up to 14% could be measured between different endogenous pacemakers. In conclusion, based on our results, disregarding the spatial origin of excitation may lead to misinterpretations and erroneous conclusions of FP parameters in the verification of research hypotheses in cellular electrocardiology.
Collapse
Affiliation(s)
- R. Kienast
- Institute of Electrical and Biomedical Engineering, University for Health Sciences, Medical Informatics and Technology, Hall in Tyrol, Austria; and
| | - M. Stöger
- Institute of Electrical and Biomedical Engineering, University for Health Sciences, Medical Informatics and Technology, Hall in Tyrol, Austria; and
- Division of Internal Medicine III/Cardiology, Medical University Innsbruck, Innsbruck, Austria
| | - M. Handler
- Institute of Electrical and Biomedical Engineering, University for Health Sciences, Medical Informatics and Technology, Hall in Tyrol, Austria; and
| | - F. Hanser
- Institute of Electrical and Biomedical Engineering, University for Health Sciences, Medical Informatics and Technology, Hall in Tyrol, Austria; and
| | - C. Baumgartner
- Institute of Electrical and Biomedical Engineering, University for Health Sciences, Medical Informatics and Technology, Hall in Tyrol, Austria; and
| |
Collapse
|
48
|
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
|
49
|
Ting S, Liew SJ, Japson F, Shang F, Chong WK, Reuveny S, Tham JY, Li X, Oh S. Time‐resolved video analysis and management system for monitoring cardiomyocyte differentiation processes and toxicology assays. Biotechnol J 2014; 9:675-83. [DOI: 10.1002/biot.201300262] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 02/12/2014] [Accepted: 03/07/2014] [Indexed: 12/22/2022]
Affiliation(s)
- Sherwin Ting
- Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), Singapore
| | - Seaw Jia Liew
- Singapore Institute of Manufacturing Technology, A*STAR (Agency for Science, Technology and Research), Singapore
| | - Francis Japson
- Institute of Infocomm Research, A*STAR (Agency for Science, Technology and Research), Singapore
| | - Fuchun Shang
- Institute of Infocomm Research, A*STAR (Agency for Science, Technology and Research), Singapore
| | - Wee Keat Chong
- Singapore Institute of Manufacturing Technology, A*STAR (Agency for Science, Technology and Research), Singapore
| | - Shaul Reuveny
- Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), Singapore
| | - Jo Yew Tham
- Institute of Infocomm Research, A*STAR (Agency for Science, Technology and Research), Singapore
| | - Xiang Li
- Singapore Institute of Manufacturing Technology, A*STAR (Agency for Science, Technology and Research), Singapore
| | - Steve Oh
- Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), Singapore
| |
Collapse
|
50
|
Fu TM, Duan X, Jiang Z, Dai X, Xie P, Cheng Z, Lieber CM. Sub-10-nm intracellular bioelectronic probes from nanowire-nanotube heterostructures. Proc Natl Acad Sci U S A 2014; 111:1259-64. [PMID: 24474745 PMCID: PMC3910633 DOI: 10.1073/pnas.1323389111] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The miniaturization of bioelectronic intracellular probes with a wide dynamic frequency range can open up opportunities to study biological structures inaccessible by existing methods in a minimally invasive manner. Here, we report the design, fabrication, and demonstration of intracellular bioelectronic devices with probe sizes less than 10 nm. The devices are based on a nanowire-nanotube heterostructure in which a nanowire field-effect transistor detector is synthetically integrated with a nanotube cellular probe. Sub-10-nm nanotube probes were realized by a two-step selective etching approach that reduces the diameter of the nanotube free-end while maintaining a larger diameter at the nanowire detector necessary for mechanical strength and electrical sensitivity. Quasi-static water-gate measurements demonstrated selective device response to solution inside the nanotube, and pulsed measurements together with numerical simulations confirmed the capability to record fast electrophysiological signals. Systematic studies of the probe bandwidth in different ionic concentration solutions revealed the underlying mechanism governing the time response. In addition, the bandwidth effect of phospholipid coatings, which are important for intracellular recording, was investigated and modeled. The robustness of these sub-10-nm bioelectronics probes for intracellular interrogation was verified by optical imaging and recording the transmembrane resting potential of HL-1 cells. These ultrasmall bioelectronic probes enable direct detection of cellular electrical activity with highest spatial resolution achieved to date, and with further integration into larger chip arrays could provide a unique platform for ultra-high-resolution mapping of activity in neural networks and other systems.
Collapse
Affiliation(s)
- Tian-Ming Fu
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
| | - Xiaojie Duan
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China; and
| | - Zhe Jiang
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
| | - Xiaochuan Dai
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
| | - Ping Xie
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
| | - Zengguang Cheng
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
| | - Charles M. Lieber
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138
| |
Collapse
|