1
|
Ito T, Suzuki T, Sakai Y, Nishioka K, Itoh Y, Sakamoto K, Ikemura N, Matoba S, Kanda Y, Takagi J, Okamoto T, Tahara K, Hoshino A. Engineered ACE2 decoy in dry powder form for inhalation: A novel therapy for SARS-CoV-2 variants. Mol Ther Methods Clin Dev 2025; 33:101459. [PMID: 40276779 PMCID: PMC12019485 DOI: 10.1016/j.omtm.2025.101459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 03/27/2025] [Indexed: 04/26/2025]
Abstract
The persistent threat of SARS-CoV-2 and the emergence of new variants has prompted the development of a novel, easily administered modality that can overcome viral mutations. The engineered ACE2 decoy shows neutralizing activity comparable to monoclonal antibodies and is broadly effective against SARS-CoV-2 variants and ACE2-utilizing sarbecoviruses. In addition to intravenous administration, this decoy has shown antiviral efficacy through nebulized aerosol inhalation in murine and primate models, offering a dose-sparing advantage. Clinically, dry powder formulation is ideal for convenience and storage but poses challenges for protein biologics. This study developed a freeze-dried spray formulation of the ACE2 decoy for inhalation. The trehalose and leucine-based excipient maintained neutralizing activity and prevented aggregate formation. The dry powder showed aerodynamic distribution from bronchi to alveoli, aiding protection against SARS-CoV-2 infections. Neutralizing activity, structural stability, and powder dispersibility were preserved after 6 months of storage. In a mouse model of SARS-CoV-2 infection, significant reductions in viral replication and lung pathology were observed with intratracheal administration 24 h post-infection. The ACE2 decoy retained activity against recent JN.1 and current KP.3 strains, confirming its robust efficacy against viral mutations. This ACE2 decoy powder inhalant is a self-administered, next-generation treatment addressing the ongoing immune-evading evolution of SARS-CoV-2.
Collapse
Affiliation(s)
- Takaaki Ito
- Laboratory of Pharmaceutical Engineering, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Tatsuya Suzuki
- Department of Microbiology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Yusuke Sakai
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 208-0011, Japan
| | - Keisuke Nishioka
- Department of Infectious Diseases, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yumi Itoh
- Department of Microbiology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Kentarou Sakamoto
- Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| | - Nariko Ikemura
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, Kanagawa 210-9501, Japan
| | - Junichi Takagi
- Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka 565-0871, Japan
| | - Toru Okamoto
- Department of Microbiology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Kohei Tahara
- Laboratory of Pharmaceutical Engineering, Gifu Pharmaceutical University, Gifu 501-1196, Japan
- Laboratory of Nanofiber Technology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Atsushi Hoshino
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| |
Collapse
|
2
|
Bassett J, Balasubramanian B, Clouse H, Trepakova E. High content imaging of relative ATP levels for mitochondrial toxicity prediction in human induced pluripotent stem cell derived cardiomyocytes. Toxicology 2025; 514:154088. [PMID: 39971086 DOI: 10.1016/j.tox.2025.154088] [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: 10/23/2024] [Revised: 02/05/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025]
Abstract
Human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) are increasingly being evaluated in assays aimed at better understanding potential cardiotoxic liability of newly developed therapeutic compounds. Disruption of mitochondria has been implicated in the mechanism of drug-induced cardiotoxicity of some compounds. Therefore, we have developed a high content imaging assay for the investigation of mitochondrial toxicity in hiPSC-CMs using ATP-Red, a fluorescent dye capable of detecting subcellular localization of relative ATP levels in living cells. We demonstrated time-dependent decreases in ATP-Red signal over 6 h treatment with known mitochondrial toxicants antimycin (0.03, 0.1 µM) or oligomycin (3, 10 µM). Concentration-dependent decreases in ATP-Red signal with antimycin (0.001-0.3 µM) and oligomycin (0.003-1 µM) were rescued by restoring glycolysis through glucose supplementation. Decreased ATP levels were also identified in a selection of clinically available drugs with reported association with mitochondrial toxicity but absent in compounds with no known association with mitochondrial dysfunction. ATP measurements using the ATP-Red imaging assay were consistent with orthogonal measurements of whole cell ATP levels in lysed hiPSC-CMs following compound treatment. Similar findings were also obtained with measurement of mitochondrial membrane potential, except amiodarone which had no change despite decreased ATP levels. The developed high throughput imaging assay, assessing subcellular ATP dynamics, could provide mechanistic insights for tested compounds.
Collapse
|
3
|
Kort EJ, Sayed N, Liu C, Mondéjar-Parreño G, Forsberg J, Eugster E, Wu SM, Wu JC, Jovinge S. Olmesartan Restores LMNA Function in Haploinsufficient Cardiomyocytes. Circulation 2025; 151:1436-1448. [PMID: 40166828 PMCID: PMC12084018 DOI: 10.1161/circulationaha.121.058621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/17/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Gene mutations are responsible for a sizeable proportion of cases of heart failure. However, the number of patients with any specific mutation is small. Repositioning of existing US Food and Drug Administration-approved compounds to target specific mutations is a promising approach to efficient identification of new therapies for these patients. METHODS The National Institutes of Health Library of Integrated Network-Based Cellular Signatures database was interrogated to identify US Food and Drug Administration-approved compounds that demonstrated the ability to reverse the transcriptional effects of LMNA knockdown. Top hits from this screening were validated in vitro with patient-specific induced pluripotent stem cell-derived cardiomyocytes combined with force measurement, gene expression profiling, electrophysiology, and protein expression analysis. RESULTS Several angiotensin receptor blockers were identified from our in silico screen. Of these, olmesartan significantly elevated the expression of sarcomeric genes and rate and force of contraction and ameliorated arrhythmogenic potential. In addition, olmesartan exhibited the ability to reduce phosphorylation of extracellular signal-regulated kinase 1 in LMNA-mutant induced pluripotent stem cell-derived cardiomyocytes. CONCLUSIONS In silico screening followed by in vitro validation with induced pluripotent stem cell-derived models can be an efficient approach to identifying repositionable therapies for monogenic cardiomyopathies.
Collapse
Affiliation(s)
- Eric J. Kort
- DeVos Cardiovascular Research Program, Fredrik Meijer Heart and Vascular Institute, Spectrum Health and Van Andel Institute, Grand Rapids, MI (E.J.K., J.F., E.E., S.J.)
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids (E.J.K.)
- Helen DeVos Children’s Hospital, Corewell Health, Grand Rapids, MI (E.J.K.)
| | - Nazish Sayed
- Cardiovascular Institute, Stanford University, CA (N.S., C.L., G.M.-P., S.M.W., J.C.W., S.J.)
- Division of Vascular Surgery, Department of Surgery, Stanford University, CA (N.S.)
| | - Chun Liu
- Cardiovascular Institute, Stanford University, CA (N.S., C.L., G.M.-P., S.M.W., J.C.W., S.J.)
| | - Gema Mondéjar-Parreño
- Cardiovascular Institute, Stanford University, CA (N.S., C.L., G.M.-P., S.M.W., J.C.W., S.J.)
| | - Jens Forsberg
- DeVos Cardiovascular Research Program, Fredrik Meijer Heart and Vascular Institute, Spectrum Health and Van Andel Institute, Grand Rapids, MI (E.J.K., J.F., E.E., S.J.)
| | - Emily Eugster
- DeVos Cardiovascular Research Program, Fredrik Meijer Heart and Vascular Institute, Spectrum Health and Van Andel Institute, Grand Rapids, MI (E.J.K., J.F., E.E., S.J.)
| | - Sean M. Wu
- Cardiovascular Institute, Stanford University, CA (N.S., C.L., G.M.-P., S.M.W., J.C.W., S.J.)
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, CA (S.M.W., J.C.W.)
| | - Joseph C. Wu
- Cardiovascular Institute, Stanford University, CA (N.S., C.L., G.M.-P., S.M.W., J.C.W., S.J.)
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, CA (S.M.W., J.C.W.)
- Department of Radiology, Stanford University, CA (J.C.W.)
| | - Stefan Jovinge
- DeVos Cardiovascular Research Program, Fredrik Meijer Heart and Vascular Institute, Spectrum Health and Van Andel Institute, Grand Rapids, MI (E.J.K., J.F., E.E., S.J.)
- Cardiovascular Institute, Stanford University, CA (N.S., C.L., G.M.-P., S.M.W., J.C.W., S.J.)
- Skåne University Hospital, Lund University, Lund, Sweden (S.J.)
| |
Collapse
|
4
|
Lee SG, Kim Y, Park SW, Kim MW, Oh JS, Park S, Lee S, Lee YH, Jeong Y, Park JH, Lee M, Shin H, Kim S, Bae YM, Kim CY, Chung HM. Evaluation of phthalates induced cardiotoxicity using human iPSCs-derived cardiomyocyte and dual-cardiotoxicity evaluation methods. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 296:118196. [PMID: 40262243 DOI: 10.1016/j.ecoenv.2025.118196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 03/26/2025] [Accepted: 04/12/2025] [Indexed: 04/24/2025]
Abstract
Phthalates, known as plasticizers, are endocrine disruptor, and their risks are being highlighted as their use increases worldwide. Di-2-ethylhexyl phthalate (DEHP), the most prevalent of the phthalates, is known to be toxic to humans, and it has recently been reported to be linked to cardiotoxicity. Although many other phthalates are also widely used, data on their cardiotoxic effects are yet to be well established. In this study, we assessed the cardiotoxic potential of various phthalates using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and a microelectrode array-based dual-cardiotoxicity evaluation method previously reported. Cytotoxicity results showed that acute exposure to DEHP, dibutyl phthalate (DBP), benzyl butyl phthalate (BBP), and di-n-octyl phthalate (DnOP) did not affect the viability of hiPSC-CMs. Before examining the functional changes in hiPSC-CMs caused by exposure to these four phthalates, we present changes in field potential (FP) and contractility based on the blocking of major ions for reference. Contrary to concerns, FP results showed a dramatic decrease in spike amplitude, beat period, and FP duration (FPD) at high doses of DBP and BBP rather than DEHP. Interestingly, DnOP resulted in a prolonged FPD, unlike the others. Furthermore, contractility results indicated that, unlike DEHP and DnOP, high doses of DBP and BBP caused beating arrest along with decreased beat amplitude. Overall, this study demonstrated that phthalates other than DEHP can also induce cardiotoxicity, even with acute exposure. It is expected that the application of the established evaluation method will facilitate the development of safe alternatives.
Collapse
Affiliation(s)
- Seul-Gi Lee
- College of Veterinary Medicine, Konkuk University, Seoul 05029, South Korea
| | - Yoonseo Kim
- College of Veterinary Medicine, Konkuk University, Seoul 05029, South Korea
| | - Sang Woong Park
- Department of Emergency Medical Services, Eulji University, 553 Sanseong-daero, Seongnam-si, Gyeonggi-do 13135, South Korea
| | - Min Woo Kim
- College of Veterinary Medicine, Konkuk University, Seoul 05029, South Korea
| | - Jeong-Seop Oh
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, 1, Gwanak-Ro, Gwanak-Gu, Seoul 08826, South Korea
| | - Shinhye Park
- College of Veterinary Medicine, Konkuk University, Seoul 05029, South Korea
| | - Suemin Lee
- College of Veterinary Medicine, Konkuk University, Seoul 05029, South Korea
| | - Yun Hyeong Lee
- College of Veterinary Medicine, Konkuk University, Seoul 05029, South Korea
| | - Youngin Jeong
- College of Veterinary Medicine, Konkuk University, Seoul 05029, South Korea
| | - Jeong Hwan Park
- College of Veterinary Medicine, Konkuk University, Seoul 05029, South Korea
| | - Myeonghee Lee
- College of Veterinary Medicine, Konkuk University, Seoul 05029, South Korea
| | - Hyewon Shin
- College of Veterinary Medicine, Konkuk University, Seoul 05029, South Korea
| | - Seeun Kim
- College of Veterinary Medicine, Konkuk University, Seoul 05029, South Korea
| | - Young Min Bae
- Department of Physiology, Institute of Functional Genomics and Research Institute of Medical Science, Konkuk University School of Medicine, 268 Chungwon-daero, Chungju 27478, South Korea
| | - C-Yoon Kim
- College of Veterinary Medicine, Konkuk University, Seoul 05029, South Korea.
| | - Hyung Min Chung
- Department of Stem Cell Biology, Konkuk University School of Medicine, 268 Chungwon-daero, Chungju 27478, South Korea.
| |
Collapse
|
5
|
Yoshinaga D, Craven I, Feng R, Prondzynski M, Shani K, Tharani Y, Mayourian J, Joseph M, Walker D, Bortolin RH, Carreon CK, Boss B, Upton S, Parker KK, Pu WT, Bezzerides VJ. Dysregulation of N-terminal acetylation causes cardiac arrhythmia and cardiomyopathy. Nat Commun 2025; 16:3604. [PMID: 40234403 PMCID: PMC12000442 DOI: 10.1038/s41467-025-58539-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 03/21/2025] [Indexed: 04/17/2025] Open
Abstract
N-terminal acetyltransferases including NAA10 catalyze N-terminal acetylation, an evolutionarily conserved co- and post-translational modification. However, little is known about the role of N-terminal acetylation in cardiac homeostasis. To gain insight into cardiac-dependent NAA10 function, we studied a previously unidentified NAA10 variant p.(Arg4Ser) segregating with QT-prolongation, cardiomyopathy, and developmental delay in a large kindred. Here, we show that the NAA10R4S variant reduced enzymatic activity, decreased NAA10-NAA15 complex formation, and destabilized the enzymatic complex N-terminal acetyltransferase A. In NAA10R4S/Y-induced pluripotent stem-cell-derived cardiomyocytes (iPSC-CMs), dysregulation of the late sodium and slow delayed rectifier potassium currents caused severe repolarization abnormalities, consistent with clinical QT prolongation. Engineered heart tissues generated from NAA10R4S/Y-iPSC-CMs had significantly decreased contractile force and sarcomeric disorganization, consistent with the pedigree's cardiomyopathic phenotype. Proteomic studies revealed dysregulation of metabolic pathways and cardiac structural proteins. We identified small molecule and genetic therapies that normalized the phenotype of NAA10R4S/Y-iPSC-CMs. Our study defines the roles of N-terminal acetylation in cardiac regulation and delineates mechanisms underlying QT prolongation, arrhythmia, and cardiomyopathy caused by NAA10 dysfunction.
Collapse
Affiliation(s)
- Daisuke Yoshinaga
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Isabel Craven
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rui Feng
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maksymilian Prondzynski
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kevin Shani
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Allston, MA, USA
| | - Yashasvi Tharani
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joshua Mayourian
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Milosh Joseph
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - David Walker
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Raul H Bortolin
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Bridget Boss
- Department of Pediatric Cardiology, Dartmouth Hitchcock Medical Center, Manchester, NH, USA
| | - Sheila Upton
- Department of Medical Genetics, Dartmouth Hitchcock Medical Center, Manchester, NH, USA
| | - Kevin Kit Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Allston, MA, USA
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Vassilios J Bezzerides
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Pediatric Cardiology, Dartmouth Hitchcock Medical Center, Manchester, NH, USA.
| |
Collapse
|
6
|
Li W, Luo X, Strano A, Arun S, Gamm O, Poetsch MS, Hasse M, Steiner RP, Fischer K, Pöche J, Ulbricht Y, Lesche M, Trimaglio G, El-Armouche A, Dahl A, Mirtschink P, Guan K, Schubert M. Comprehensive promotion of iPSC-CM maturation by integrating metabolic medium with nanopatterning and electrostimulation. Nat Commun 2025; 16:2785. [PMID: 40118846 PMCID: PMC11928738 DOI: 10.1038/s41467-025-58044-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 03/03/2025] [Indexed: 03/24/2025] Open
Abstract
The immaturity of human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) is a major limitation for their use in drug screening to identify pro-arrhythmogenic or cardiotoxic molecules. Here, we demonstrate an approach that combines lipid-enriched maturation medium with a high concentration of calcium, nanopatterning of culture surfaces and electrostimulation to generate iPSC-CMs with advanced electrophysiological, structural and metabolic phenotypes. Systematic testing reveals that electrostimulation is the key driver of enhanced mitochondrial development and metabolic maturation and improved electrophysiological properties of iPSC-CMs. Increased calcium concentration strongly promotes electrophysiological maturation, while nanopatterning primarily facilitates sarcomere organisation with minor effect on electrophysiological properties. Transcriptome analysis reveals that activation of HMCES and TFAM targets contributes to mitochondrial development, whereas downregulation of MAPK/PI3K and SRF targets is associated with iPSC-CM polyploidy. These findings provide mechanistic insights into iPSC-CM maturation, paving the way for pharmacological responses that more closely resemble those of adult CMs.
Collapse
Affiliation(s)
- Wener Li
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Xiaojing Luo
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Anna Strano
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Shakthi Arun
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Oliver Gamm
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Mareike S Poetsch
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Marcel Hasse
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Robert-Patrick Steiner
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Konstanze Fischer
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Jessie Pöche
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Ying Ulbricht
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Mathias Lesche
- DRESDEN-concept Genome Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Giulia Trimaglio
- Institute of Clinical Chemistry and Laboratory Medicine, Department of Clinical Pathobiochemistry, University Hospital Dresden, Dresden, Germany
- National Center for Tumor Diseases, Partner Site Dresden, 01307 Dresden, and German Cancer Research Center, Heidelberg, Germany
| | - Ali El-Armouche
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Andreas Dahl
- DRESDEN-concept Genome Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Peter Mirtschink
- Institute of Clinical Chemistry and Laboratory Medicine, Department of Clinical Pathobiochemistry, University Hospital Dresden, Dresden, Germany
| | - Kaomei Guan
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany.
| | - Mario Schubert
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
7
|
Kim B, Choi JS, Zhu Y, Kim J, Kim YS, Parra A, Locke PA, Kim JH, Herron T, Kim DH. Effect of electrochemical topology on detection sensitivity in MEA assay for drug-induced cardiotoxicity screening. Biosens Bioelectron 2025; 272:117082. [PMID: 39778241 PMCID: PMC11773427 DOI: 10.1016/j.bios.2024.117082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 12/15/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025]
Abstract
Cardiotoxicity remains a major challenge in drug development, accounting for 45% of medication withdrawals due to cardiac ischemia and arrhythmogenicity. To overcome the limitations of traditional multielectrode array (MEA)-based cardiotoxicity assays, we developed a Nafion-coated NanoMEA platform with decoupled reference electrodes, offering enhanced sensitivity for electrophysiological measurements. The 'Decoupled' configuration significantly reduced polarization resistance (Rp) from 12.77 MΩ to 3.41 MΩ, improving charge transfer efficiency as demonstrated by electrochemical impedance spectroscopy and cyclic voltammetry. Additionally, the limit of detection significantly decreased from 0.175 MΩ (Coupled) to 0.040 MΩ (Decoupled), underscoring the system's enhanced sensitivity. Using human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), we evaluated the effects of three proarrhythmic drugs: Ranolazine, Domperidone, and Sotalol. Under the decoupled condition, the platform exhibited reductions in IC50 values for Domperidone (0.71 μM-0.29 μM), Sotalol (7.61 μM-0.27 μM), and Ranolazine (53.08 μM-5.89 μM), demonstrating significantly improved drug detection sensitivity. Longitudinal analysis revealed significant alterations in key electrophysiological parameters, including beating period (BP), field potential duration (FPD), spike slope, and amplitude, which were consistent with the known pharmacological actions of these drugs. Further validation through action potential (AP) waveform analysis showed enhanced repolarization dynamics, confirming the platform's predictive capabilities. Our findings highlight the critical role of electrochemical topology in optimizing MEA performance. The NanoMEA system, featuring decoupled Nafion-coated electrodes, represents a robust and sensitive platform for cardiotoxicity screening, setting a new standard for preclinical drug safety assessment and advancing bioelectronic device design for cardiac research.
Collapse
Affiliation(s)
- Byunggik Kim
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, United States
| | - Jong Seob Choi
- Division of Advanced Materials Engineering, Kongju National University, Budaedong 275, Seobuk-gu, Cheonan-si, Chungnam, 31080, Republic of Korea; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, United States
| | - Yiguang Zhu
- Department of Environmental Health and Engineering, Johns Hopkins University, Baltimore, MD, 21205, United States
| | - Juhyun Kim
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute (KBRI), Daegu, 41062, Republic of Korea
| | - Ye Seul Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Andres Parra
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, United States
| | - Paul A Locke
- Department of Environmental Health and Engineering, Johns Hopkins University, Baltimore, MD, 21205, United States
| | - Jae Ho Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Todd Herron
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, United States; Department of Medicine, Johns Hopkins University, Baltimore, MD, 21205, United States; Institute for NanoBio Technology, Johns Hopkins University, Baltimore, MD, 21218, United States; Center for Microphysiological Systems, Johns Hopkins University, Baltimore, MD, 21205, United States.
| |
Collapse
|
8
|
Park NK, Park YG, Choi JH, Choi HK, Moon SH, Park SJ, Choi SW. Human induced pluripotent stem cell-cardiomyocytes for cardiotoxicity assessment: a comparative study of arrhythmiainducing drugs with multi-electrode array analysis. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2025; 29:257-269. [PMID: 39972675 PMCID: PMC11842287 DOI: 10.4196/kjpp.24.413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/03/2025] [Accepted: 01/03/2025] [Indexed: 02/21/2025]
Abstract
Reliable preclinical models for assessing drug-induced cardiotoxicity are essential to reduce the high rate of drug withdrawals during development. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have emerged as a promising platform for such assessments due to their expression of cardiacspecific ion channels and electrophysiological properties. In this study, we investigated the effects of eight arrhythmogenic drugs-E4031, nifedipine, mexiletine, JNJ303, flecainide, moxifloxacin, quinidine, and ranolazine-on hiPSC-CMs derived from both healthy individuals and a long QT syndrome (LQTS) patient using multielectrode array systems. The results demonstrated dose-dependent changes in field potential duration and arrhythmogenic risk, with LQTS-derived hiPSC-CMs showing increased sensitivity to hERG channel blockers such as E4031. Furthermore, the study highlights the potential of hiPSC-CMs to model disease-specific cardiac responses, providing insights into genetic predispositions and personalized drug responses. Despite challenges related to the immaturity of hiPSC-CMs, their ability to recapitulate human cardiac electrophysiology makes them a valuable tool for preclinical cardiotoxicity assessments. This study underscores the utility of integrating patientderived hiPSC-CMs with advanced analytical platforms, such as multi-electrode array systems, to evaluate drug-induced electrophysiological changes. These findings reinforce the role of hiPSC-CMs in drug development, facilitating safer and more efficient screening methods while supporting precision medicine applications.
Collapse
Affiliation(s)
| | - Yun-Gwi Park
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Korea
| | - Ji-Hee Choi
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Korea
| | - Hyung Kyu Choi
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Korea
| | - Sung-Hwan Moon
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Korea
| | | | - Seong Woo Choi
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Korea
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Korea
| |
Collapse
|
9
|
Kuo CW, Gök C, Fulton H, Dickson-Murray E, Adu S, Gallen EK, Mary S, Robertson AD, Jordan F, Dunning E, Mullen W, Smith GL, Fuller W. Nanobody-thioesterase chimeras to specifically target protein palmitoylation. Nat Commun 2025; 16:1445. [PMID: 39920166 PMCID: PMC11805987 DOI: 10.1038/s41467-025-56716-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 01/27/2025] [Indexed: 02/09/2025] Open
Abstract
The complexity of the cellular proteome is massively expanded by a repertoire of chemically distinct reversible post-translational modifications (PTMs) that control protein localisation, interactions, and function. The temporal and spatial control of these PTMs is central to organism physiology, and mis-regulation of PTMs is a hallmark of many diseases. Here we present an approach to manipulate PTMs on target proteins using nanobodies fused to enzymes that control these PTMs. Anti-GFP nanobodies fused to thioesterases (which depalmitoylate protein cysteines) depalmitoylate GFP tagged substrates. A chemogenetic approach to enhance nanobody affinity for its target enables temporal control of target depalmitoylation. Using a thioesterase fused to a nanobody directed against the Ca(v)1.2 beta subunit we reduce palmitoylation of the Ca(v)1.2 alpha subunit, modifying the channel's voltage dependence and arrhythmia susceptibility in stem cell derived cardiac myocytes. We conclude that nanobody enzyme chimeras represent an approach to specifically manipulate PTMs, with applications in both the laboratory and the clinic.
Collapse
Affiliation(s)
- Chien-Wen Kuo
- School of Cardiovascular & Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Caglar Gök
- School of Cardiovascular & Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- School of Natural Sciences, College of Health and Science, University of Lincoln, Lincoln, UK
| | - Hannah Fulton
- School of Cardiovascular & Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Eleanor Dickson-Murray
- School of Cardiovascular & Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Samuel Adu
- School of Cardiovascular & Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Emily K Gallen
- School of Cardiovascular & Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Harvard Medical School, Boston, MA, USA
| | - Sheon Mary
- School of Cardiovascular & Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Alan D Robertson
- School of Cardiovascular & Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Fiona Jordan
- School of Cardiovascular & Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Emma Dunning
- School of Cardiovascular & Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - William Mullen
- School of Cardiovascular & Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Godfrey L Smith
- School of Cardiovascular & Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - William Fuller
- School of Cardiovascular & Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
10
|
Parekh P, Sherfey J, Alaybeyoglu B, Cirit M. Pathway-Based Similarity Measurement to Quantify Transcriptomics Similarity Between Human Tissues and Preclinical Models. Clin Pharmacol Ther 2025; 117:485-494. [PMID: 39377352 PMCID: PMC11747893 DOI: 10.1002/cpt.3465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 09/14/2024] [Indexed: 10/09/2024]
Abstract
Accurate clinical translation of preclinical research remains challenging, primarily due to species-specific differences and disease and patient heterogeneity. An important recent advancement has been development of microphysiological systems that consist of multiple human cell types that recapitulate key characteristics of their respective human systems, allowing essential physiologic processes to be accurately assessed during drug development. However, an unmet need remains regarding a quantitative method to evaluate the similarity between diverse sample types for various contexts of use (CoU)-specific pathways. To address this gap, this study describes the development of pathway-based similarity measurement (PBSM), which leverages RNA-seq data and pathway-based information to assess the human relevance of preclinical models for specific CoU. PBSM offers a quantitative method to compare the transcriptomic similarity of preclinical models to human tissues, shown here as proof of concept for liver and cardiac tissues, enabling improved model selection and validation. Thus, PBSM can successfully support CoU selection for preclinical models, assess the impact of different gene sets on similarity calculations, and differentiate among various in vitro and in vivo models. PBSM has the potential to reduce the translational gap in drug development by allowing quantitative evaluation of the similarity of preclinical models to human tissues, facilitating model selection, and improving understanding of context-specific applications. PBSM can serve as a foundation for enhancing the physiological relevance of in vitro models and supporting the development of more effective therapeutic interventions.
Collapse
Affiliation(s)
- Paarth Parekh
- Javelin Biotech, Inc., 299 Washington Street, Woburn, Massachusetts 01801, USA
| | - Jason Sherfey
- Javelin Biotech, Inc., 299 Washington Street, Woburn, Massachusetts 01801, USA
| | - Begum Alaybeyoglu
- Javelin Biotech, Inc., 299 Washington Street, Woburn, Massachusetts 01801, USA
| | - Murat Cirit
- Javelin Biotech, Inc., 299 Washington Street, Woburn, Massachusetts 01801, USA
| |
Collapse
|
11
|
Reynolds TS, Mishra SJ, Blagg BSJ. Assessment of Hsp90β-selective inhibitor safety and on-target effects. Sci Rep 2025; 15:3692. [PMID: 39880847 PMCID: PMC11779861 DOI: 10.1038/s41598-025-86647-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/13/2025] [Indexed: 01/31/2025] Open
Abstract
The heat shock protein 90 (Hsp90) family of molecular chaperones mediates the folding and activation of ~ 400 client proteins, many of which contribute to oncogenesis. As a result, Hsp90 pan-inhibitors, which inhibit all four Hsp90 isoforms, have been investigated in the clinic for the treatment of cancer. Unfortunately, detrimental side effects were observed and hindered the clinical development of pan-Hsp90 inhibitors. The two most common on-target toxicities, cardio-toxicity and ocular-toxicity, have been attributed to inhibition of the Hsp90α isoform. As an alternative strategy, Hsp90β-selective inhibitors have been developed, which have shown promising anti-cancer activity in vitro and in vivo in combination with immune-checkpoint blockade therapy. This study aims to assess the potential risks of cardio-toxicity and ocular-toxicity exhibited by Hsp90β-selective inhibitors in vitro. In summary, the Hsp90β-selective NDNB1182 was found to avoid the cardio- and ocular-toxicity typical of Hsp90 pan-inhibitors (e.g. 17-AAG), providing a promising path toward the generation of isoform-selective Hsp90 inhibitors.
Collapse
Affiliation(s)
- Tyelor S Reynolds
- Department of Chemistry and Biochemistry, The University of Notre Dame, 305 McCourtney Hall, Notre Dame, IN, 46556, USA
| | - Sanket J Mishra
- Department of Chemistry and Biochemistry, The University of Notre Dame, 305 McCourtney Hall, Notre Dame, IN, 46556, USA
- Grannus Therapeutics Inc., 1400 E Angela Blvd, South Bend, IN, 46617, USA
| | - Brian S J Blagg
- Department of Chemistry and Biochemistry, The University of Notre Dame, 305 McCourtney Hall, Notre Dame, IN, 46556, USA.
| |
Collapse
|
12
|
Paredes-Espinosa MB, Paluh JL. Synthetic embryology of the human heart. Front Cell Dev Biol 2025; 12:1478549. [PMID: 39935786 PMCID: PMC11810959 DOI: 10.3389/fcell.2024.1478549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 12/30/2024] [Indexed: 02/13/2025] Open
Abstract
The evolution of stem cell-based heart models from cells and tissues to organoids and assembloids and recently synthetic embryology gastruloids, is poised to revolutionize our understanding of cardiac development, congenital to adult diseases, and patient customized therapies. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have already been integrated into transplantable patches and are in preclinical efforts to reverse fibrotic scarring from myocardial infarctions. To inform on the complexity of heart diseases, multi-tissue morphogenic heart models are needed that replicate fundamental components of heart function to heart organogenesis in vitro and which require a deep understanding of heart development. Organoid and assembloid models capture selected multicellular cardiac processes, such as chamber formation and priming events for vascularization. Gastruloid heart models offer deeper insights as synthetic embryology to mimic multi-staged developmental events of in vivo heart organogenesis including established heart fields, crescent formation and heart tube development along with vascular systemic foundation and even further steps. The human Elongating Multi-Lineage Organized Cardiac (EMLOC) gastruloid model captures these stages and additional events including chamber genesis, patterned vascularization, and extrinsic central and intrinsic cardiac nervous system (CNS-ICNS) integration guided by spatiotemporal and morphogenic processes with neural crest cells. Gastruloid synthetic embryology heart models offer new insights into previously hidden processes of development and provide powerful platforms for addressing heart disease that extends beyond cardiomyocytes, such as arrhythmogenic diseases, congenital defects, and systemic injury interactions, as in spinal cord injuries. The holistic view that is emerging will reveal heart development and disease in unprecedented detail to drive transformative state-of-the-art innovative applications for heart health.
Collapse
Affiliation(s)
| | - Janet L. Paluh
- Department of Nanoscale Science and Engineering, College of Nanotechnology, Science and Engineering, University at Albany, Albany, NY, United States
| |
Collapse
|
13
|
Kawagishi H, Kanda Y. [Safety and efficacy assessments using human iPS cell-derived cardiomyocytes]. Nihon Yakurigaku Zasshi 2025; 160:4-8. [PMID: 39756904 DOI: 10.1254/fpj.24043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
The delay and loss of drugs are serious problems in Japan. To overcome this issue, it is important to strengthen drug development capabilities. For drug development, the establishment and advancement of non-clinical testing methods are necessary for safe and effective clinical trials. Recently, the movement toward alternatives to animal testing has accelerated internationally. New Approach Methodologies (NAMs), such as human inducible pluripotent stem cell (hiPSC) technology and in silico modeling & simulation, are considered valuable for drug development. It has been demonstrated that hiPSC-derived cardiomyocytes (hiPSC-CMs) are useful tools to assess drug-induced cardiotoxicity, including arrhythmia and cardiac contractile dysfunction, leading to the use of hiPSC-CMs in the drug review process. Advancing hiPSC technologies have enabled the generation of mature hiPSC-CMs and engineered heart tissues, which are expected to provide novel information in drug safety and efficacy evaluation. Furthermore, it would be possible to establish the non-clinical evaluation that takes into account individual differences by developing hiPSCs bearing characteristics specific to certain populations, such as pediatrics or rare disease patients. Here, we present the recent findings and future perspectives on non-clinical evaluation using hiPSC technology.
Collapse
|
14
|
Kawagishi H, Nakajima T, Ramadhiani R, Nakada T, Tomita T, Kanda Y, Emoto N, Yamada M. [Development of a Novel Therapeutic for Pediatric Heart Failure Targeting Angiotensin II Receptor]. YAKUGAKU ZASSHI 2025; 145:275-280. [PMID: 40175146 DOI: 10.1248/yakushi.24-00171-1] [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] [Indexed: 04/04/2025]
Abstract
Pediatric drug development is a global challenge. Children undergo organ growth and exhibit different drug reactions than adults, resulting in different pharmacological responses. Therefore, it is necessary to consider children's physiological characteristics when evaluating drug efficacy and safety in pediatric patients. We conducted drug discovery research for the treatment of pediatric heart failure, based on neonatal-specific physiological functions of angiotensin II. Pediatric heart failure is one of the most important causes of death in children, particularly neonates and infants. However, only a few therapeutic agents are available to treat pediatric heart failure. Angiotensin II binds to its receptors (angiotensin II receptor type 1: AT1R) and regulates cellular functions through G protein and β-arrestin pathways. We previously found that the β-arrestin-biased AT1R agonist, TRV027, induced a positive inotropic effect in the hearts of neonatal mice. Acute administration of TRV027 did not affect heart rate, oxygen consumption, or production of reactive oxygen species. The inotropic effect of TRV027 was also observed in human iPS cell-derived cardiomyocytes and a neonatal mouse model of dilated cardiomyopathy. Furthermore, chronic administration of TRV027 improved the survival rate of mice with dilated cardiomyopathy during the preweaning period. These results demonstrate that TRV027 has the potential to be a safe and effective drug for treating pediatric heart failure.
Collapse
Affiliation(s)
- Hiroyuki Kawagishi
- National Institute of Health Sciences
- Shinshu University School of Medicine
| | | | | | - Tsutomu Nakada
- Shinshu University Research Center for Advanced Science and Technology
| | | | | | | | | |
Collapse
|
15
|
Kraushaar U. Compound Testing of Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes Using Multi-Well Microelectrode Arrays. Methods Mol Biol 2025; 2924:189-204. [PMID: 40307643 DOI: 10.1007/978-1-0716-4530-7_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
The cardiac action potential is a highly orchestrated process that depends on the precise timing of activation and inactivation of various ion channel subtypes. Any deviation from this carefully regulated sequence, such as the blockage of specific voltage-gated potassium channels, can disrupt the electrical balance and lead to life-threatening arrhythmias. Standard cellular assays, which typically rely on cells engineered to express only a single ion channel subtype, have significant limitations in accurately predicting a compound's effects on the heart. These limitations often result in prematurely rejecting many potentially promising compounds during the early stages of drug development.To address this, there is an increasing demand for more physiologically relevant and predictive functional assays. The emergence of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) has revolutionized this field by offering assays that are more representative of human cardiac physiology. These cells not only provide a more accurate model for assessing drug effects but also support the development of patient-derived disease models, offering personalized insights into cardiac health.Microelectrode array (MEA) systems have become a crucial tool in this context, enabling the non-invasive recording and analysis of cardiac field action potentials from cultured hiPSC-CMs. These systems provide a platform to monitor the electrical activity of cardiomyocytes in real time, offering valuable data on the electrophysiological properties of cardiac cells under various conditions. This chapter presents a comprehensive protocol for cultivating hiPSC-CMs on two parallelized MEA systems. We also outline a strategic approach for conducting compound testing, designed to maximize the predictive power of these assays and enhance their application in safety pharmacology.
Collapse
Affiliation(s)
- Udo Kraushaar
- Department of Pharma & Biotech, Electrophysiology, NMI Natural and Medical Sciences Institute at the University of Tuebingen, Reutlingen, Germany.
| |
Collapse
|
16
|
Guichardaz M, Bottini S, Balmas E, Bertero A. Overcoming the Silencing of Doxycycline-Inducible Promoters in hiPSC-derived Cardiomyocytes. OPEN RESEARCH EUROPE 2024; 4:266. [PMID: 39926351 PMCID: PMC11803382 DOI: 10.12688/openreseurope.19024.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 12/09/2024] [Indexed: 02/11/2025]
Abstract
Background Human induced pluripotent stem cells (hiPSCs) are pivotal for studying human development, modeling diseases, and advancing regenerative medicine. Effective control of transgene expression is crucial to achieve temporal and quantitative precision in all of these contexts. The doxycycline (dox)-inducible OPTi-OX system, which integrates the Tet-On 3G transactivator and dox-responsive transgene at the hROSA26 and AAVS1 genomic safe harbors (GSHs), respectively, offers a promising solution. Yet, transgene silencing, particularly in hiPSC-derived cardiomyocytes (hiPSC-CMs), limits its utility. Methods To address this, we evaluated strategies to enhance dox-inducible transgene expression. We compared two promoters, TRE3VG and T11, for activity and stability, and investigated the addition of a Ubiquitous Chromatin Opening Element (UCOE) to reduce silencing. We also tested relocating the transgene cassette to the CLYBL GSH, and employed sodium butyrate (SB), a histone deacetylase inhibitor, to restore promoter activity. Transgene expression was assessed via flow cytometry and real-time quantitative PCR. Results TRE3VG exhibited higher activity than T11, but both were prone to silencing. UCOE did not enhance promoter activity in hiPSCs, but modestly reduced silencing in hiPSC-CMs. Targeting the CLYBL locus improved promoter activity compared to AAVS1 in both hiPSCs and hiPSC-CMs. SB restored activity in silenced inducible promoters within hiPSC-CMs, but compromised hiPSC viability. Unexpectedly, Tet-On 3G was silenced in some clones and could not be reactivated by SB. Conclusions These findings underscore the need for integrating multiple strategies, including careful GSH selection, improved cassette design, epigenetic modulation, and clone screening, to develop robust dox-inducible systems that retain functionality during hiPSC differentiation.
Collapse
Affiliation(s)
- Michelle Guichardaz
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Turin, Torino, 10126, Italy
| | - Sveva Bottini
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Turin, Torino, 10126, Italy
| | - Elisa Balmas
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Turin, Torino, 10126, Italy
| | - Alessandro Bertero
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Turin, Torino, 10126, Italy
| |
Collapse
|
17
|
Lee J, Duperrex E, El-Battrawy I, Hohn A, Saguner AM, Duru F, Emmenegger V, Cyganek L, Hierlemann A, Ulusan H. CardioMEA: comprehensive data analysis platform for studying cardiac diseases and drug responses. Front Physiol 2024; 15:1472126. [PMID: 39539954 PMCID: PMC11557525 DOI: 10.3389/fphys.2024.1472126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction In recent years, high-density microelectrode arrays (HD-MEAs) have emerged as a valuable tool in preclinical research for characterizing the electrophysiology of human induced pluripotent stem-cell-derived cardiomyocytes (iPSC-CMs). HD-MEAs enable the capturing of both extracellular and intracellular signals on a large scale, while minimizing potential damage to the cell. However, despite technological advancements of HD-MEAs, there is a lack of effective data-analysis platforms that are capable of processing and analyzing the data, particularly in the context of cardiac arrhythmias and drug testing. Methods To address this need, we introduce CardioMEA, a comprehensive data-analysis platform designed specifically for HD-MEA data that have been obtained from iPSCCMs. CardioMEA features scalable data processing pipelines and an interactive web-based dashboard for advanced visualization and analysis. In addition to its core functionalities, CardioMEA incorporates modules designed to discern crucial electrophysiological features between diseased and healthy iPSC-CMs. Notably, CardioMEA has the unique capability to analyze both extracellular and intracellular signals, thereby facilitating customized analyses for specific research tasks. Results and discussion We demonstrate the practical application of CardioMEA by analyzing electrophysiological signals from iPSC-CM cultures exposed to seven antiarrhythmic drugs. CardioMEA holds great potential as an intuitive, userfriendly platform for studying cardiac diseases and assessing drug effects.
Collapse
Affiliation(s)
- Jihyun Lee
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Eliane Duperrex
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Ibrahim El-Battrawy
- St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
- German Center for Cardiovascular Research, Partner Site Heidelberg-Mannheim and Göttingen, Heidelberg, Germany
- Department of Molecular and Experimental Cardiology, Institut für Forschung und Lehre (IFL), Ruhr-University Bochum, Bochum, Germany
- Institute of Physiology, Ruhr-University Bochum, Bochum, Germany
| | - Alyssa Hohn
- German Center for Cardiovascular Research, Partner Site Heidelberg-Mannheim and Göttingen, Heidelberg, Germany
| | - Ardan M. Saguner
- Department of Cardiology, Electrophysiology Division, University Heart Center Zurich, Zurich, Switzerland
- Center for Translational and Experimental Cardiology (CTEC), University of Zürich, Zurich, Switzerland
| | - Firat Duru
- Department of Cardiology, Electrophysiology Division, University Heart Center Zurich, Zurich, Switzerland
- Center for Translational and Experimental Cardiology (CTEC), University of Zürich, Zurich, Switzerland
| | - Vishalini Emmenegger
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Lukas Cyganek
- German Center for Cardiovascular Research, Partner Site Heidelberg-Mannheim and Göttingen, Heidelberg, Germany
- Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Andreas Hierlemann
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Hasan Ulusan
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| |
Collapse
|
18
|
Ge N, Li R, Liu M, Xia W, O'Brien ST, McInerney V, Galvin J, Ward D, McGorrian C, O'Brien T, Shen S, Prendiville TW. Defining Cardiomyocyte Repolarization Response to Pharmacotherapy in Long-QT Syndrome Type 3. J Am Heart Assoc 2024; 13:e034690. [PMID: 39377211 PMCID: PMC11935574 DOI: 10.1161/jaha.124.034690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 08/01/2024] [Indexed: 10/09/2024]
Abstract
BACKGROUND Long-QT syndrome is a primary cardiac ion channelopathy predisposing a patient to ventricular arrhythmia through delayed repolarization on the resting ECG. We aimed to establish a patient-specific, human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes model of long-QT syndrome type 3 (LQT3) using clustered regularly interspaced palindromic repeats (CRISPR/Cas9), for disease modeling and drug challenge. METHODS AND RESULTS HiPSCs were generated from a patient with LQT3 harboring an SCN5A pathogenic variant (c.1231G>A; p.Val411Met), and an unrelated healthy control. The same SCN5A pathogenic variant was engineered into the background healthy control hiPSCs via CRISPR/Cas9 gene editing to generate a second disease model of LQT3 for comparison with an isogenic control. All 3 hiPSC lines were differentiated into cardiomyocytes. Both the patient-derived LQT3 (SCN5A+/-) and genetically engineered LQT3 (SCN5A+/-) hiPSC-derived cardiomyocytes showed significantly prolonged cardiomyocyte repolarization compared with the healthy control. Mexiletine, a cardiac voltage-gated sodium channel (NaV1.5) blocker, shortened repolarization in both patient-derived LQT3 and genetically engineered LQT3 hiPSC-derived cardiomyocytes, but had no effect in the control. Notably, calcium channel blockers nifedipine and verapamil showed a dose-dependent shortening of repolarization, rescuing the phenotype. Additionally, therapeutic drugs known to prolong the corrected QT in humans (ondansetron, clarithromycin, and sotalol) demonstrated this effect in vitro, but the LQT3 clones were not more disproportionately affected compared with the control. CONCLUSIONS We demonstrated that patient-derived and genetically engineered LQT3 hiPSC-derived cardiomyocytes faithfully recapitulate pathologic characteristics of LQT3. The clinical significance of such an in vitro model is in the exploration of novel therapeutic strategies, stratifying drug adverse reaction risk and potentially facilitating a more targeted, patient-specific approach in high-risk patients with LQT3.
Collapse
Affiliation(s)
- Ning Ge
- Regenerative Medicine Institute, School of MedicineUniversity of GalwayGalwayIreland
- Department of PharmacologyNorthwestern University Feinberg School of MedicineChicagoILUSA
| | - Rui Li
- Lambe Institute for Translational ResearchUniversity of GalwayGalwayIreland
| | - Min Liu
- Department of Physiology, College of Life ScienceHebei Normal UniversityShijiazhuangChina
| | - Wenxin Xia
- Regenerative Medicine Institute, School of MedicineUniversity of GalwayGalwayIreland
| | - Stephen T. O'Brien
- Department of PaediatricsChildren’s Health Ireland at CrumlinDublinIreland
| | | | - Joseph Galvin
- Mater Misericordiae University HospitalDublinIreland
| | - Deirdre Ward
- Tallaght University HospitalTallaght, DublinIreland
| | | | - Timothy O'Brien
- Regenerative Medicine Institute, School of MedicineUniversity of GalwayGalwayIreland
| | - Sanbing Shen
- Regenerative Medicine Institute, School of MedicineUniversity of GalwayGalwayIreland
| | - Terence W. Prendiville
- Regenerative Medicine Institute, School of MedicineUniversity of GalwayGalwayIreland
- National Children’s Research CentreChildren’s Health Ireland at CrumlinDublinIreland
| |
Collapse
|
19
|
Shead KD, Huethorst E, Burton F, Lang NN, Myles RC, Smith GL. Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Preclinical Cardiotoxicity Screening in Cardio-Oncology. JACC CardioOncol 2024; 6:678-683. [PMID: 39479327 PMCID: PMC11520213 DOI: 10.1016/j.jaccao.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/27/2024] [Accepted: 07/25/2024] [Indexed: 11/02/2024] Open
Abstract
•hiPSC-CM offer an alternative to in vivo models for predicting cardiotoxicity. •hiPSC-CM monolayers detect pro-arrhythmic effects; inotropic detection is less established. •Cardiac spheroids and engineered tissue may suit chronic cardiotoxicity studies (>2 weeks). •Cardiac assays with non-myocyte cells may be key to identifying some cardiotoxicity forms. •hiPSC-CM technologies are well placed to develop patient-specific assays in the future.
Collapse
Affiliation(s)
- Kyle D. Shead
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Eline Huethorst
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Francis Burton
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Ninian N. Lang
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Rachel C. Myles
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Godfrey L. Smith
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
20
|
Clark AP, Krogh-Madsen T, Christini DJ. Stem cell-derived cardiomyocyte heterogeneity confounds electrophysiological insights. J Physiol 2024; 602:5155-5162. [PMID: 38723234 PMCID: PMC11493526 DOI: 10.1113/jp284618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/24/2024] [Indexed: 08/21/2024] Open
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) offer potential as an in vitro model for studying drug cardiotoxicity and patient-specific cardiovascular disease. The inherent electrophysiological heterogeneity of these cells limits the depth of insights that can be drawn from well-designed experiments. In this review, we provide our perspective on some sources and the consequences of iPSC-CM heterogeneity. We demonstrate the extent of heterogeneity in the literature and explain how such heterogeneity is exacerbated by patch-clamp experimental artifacts in the manual and automated set-up. Finally, we discuss how this heterogeneity, caused by both intrinsic and extrinsic factors, limits our ability to build digital twins of patient-derived cardiomyocytes.
Collapse
Affiliation(s)
- Alexander P Clark
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Trine Krogh-Madsen
- Department of Physiology & Biophysics, Weill Cornell Medicine, New York, NY, USA
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - David J Christini
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| |
Collapse
|
21
|
Li J, Li M, Nawa Y, Liu Y, Bando K, Hua Y, Sun L, Fujita S, Sawa Y, Fujita K, Liu L. Label-Free Raman Spectroscopy for Assessing Purity and Maturation of hiPSC-Derived Cardiac Tissue. Anal Chem 2024; 96:15765-15772. [PMID: 39291743 PMCID: PMC11447663 DOI: 10.1021/acs.analchem.4c03871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/05/2024] [Accepted: 09/06/2024] [Indexed: 09/19/2024]
Abstract
I. BACKGROUND Human induced pluripotent stem cell (hiPSC) derived cardiomyocytes (CMs) have been utilized in drug toxicity evaluation, drug discovery, and treating heart failure patients, showing substantial effects. Ensuring the quality, purity, and maturation of hiPSC-CMs during large-scale production is crucial. There is a growing demand for a novel method to characterize cell molecular profiles without labels and without causing damage. II. METHODS In this study, we employed label-free Raman microscopy to evaluate hiPSC-derived CMs. The study involved the characterization of cell molecular profiles without labels and without causing damage. The correlation between Raman spectroscopy of specific components, such as cytochrome c and myoglobin, and CM purity and maturation following hiPSC differentiation was investigated. Additionally, the validation of this correlation was performed by assessing mixtures of commercially available CMs (iCell cardiomyocytes2) and fibroblasts at various ratios as well as hiPSC-derived CMs with different efficiencies. Furthermore, CMs were matured using rapid pacing of traveling waves, and the Raman profiles of matured CMs were compared to those of immature ones. III. RESULTS Raman spectroscopy indicated that the cytochrome c and myoglobin showed correlation with the purity and maturation of CMs following differentiation of hiPSCs. This correlation was validated through experiments involving different CM-fibroblast mixtures and hiPSC-derived CMs with varying efficiencies. Moreover, matured CMs exhibited markedly different Raman profiles compared to immature ones, indicating the potential of Raman imaging as a tool for assessing CM maturation. IV. CONCLUSIONS We discovered that Raman spectroscopy of certain components, such as cytochrome c and myoglobin, correlates with the CM purity and maturation following hiPSC differentiation. The findings of this study highlight the potential of label-free Raman microscopy as a nondestructive, high-content, and time-efficient method for quality control of hiPSC-derived CMs. This approach could significantly contribute to ensuring the quality and maturity of hiPSC-CMs for various applications in drug discovery and regenerative medicine.
Collapse
Affiliation(s)
- Junjun Li
- Laboratory
of Nanophotonics, Department of Applied Physics, Osaka University, Suita, Osaka 565-0871, Japan
- Frontier
of Regenerative Medicine, Osaka University
Graduate School of Medicine, 2-2 Yamada-Oka, Osaka, Suita 565-0871, Japan
| | - Menglu Li
- Laboratory
of Nanophotonics, Department of Applied Physics, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yasunori Nawa
- Laboratory
of Nanophotonics, Department of Applied Physics, Osaka University, Suita, Osaka 565-0871, Japan
- Advanced
Photonics and Biosensing Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology
(AIST), Suita, Osaka 565-0871, Japan
| | - Yuting Liu
- Department
of Cardiovascular Surgery, Osaka University
Graduate School of Medicine, Osaka, Suita 565-0871, Japan
| | - Kazuki Bando
- Advanced
Photonics and Biosensing Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology
(AIST), Suita, Osaka 565-0871, Japan
| | - Ying Hua
- Department
of Cardiovascular Surgery, Osaka University
Graduate School of Medicine, Osaka, Suita 565-0871, Japan
| | - Lifu Sun
- Department
of Cardiovascular Surgery, Osaka University
Graduate School of Medicine, Osaka, Suita 565-0871, Japan
| | - Satoshi Fujita
- Advanced
Photonics and Biosensing Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology
(AIST), Suita, Osaka 565-0871, Japan
| | - Yoshiki Sawa
- Department
of Cardiovascular Surgery, Osaka Police
Hospital, Osaka 543-0035, Japan
- Cuorips
Inc., Nihonbashihoncho
3, Chome-11-5, Chuo City, Tokyo 103-0023, Japan
| | - Katsumasa Fujita
- Laboratory
of Nanophotonics, Department of Applied Physics, Osaka University, Suita, Osaka 565-0871, Japan
- Advanced
Photonics and Biosensing Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology
(AIST), Suita, Osaka 565-0871, Japan
| | - Li Liu
- Laboratory
of Nanophotonics, Department of Applied Physics, Osaka University, Suita, Osaka 565-0871, Japan
- Frontier
of Regenerative Medicine, Osaka University
Graduate School of Medicine, 2-2 Yamada-Oka, Osaka, Suita 565-0871, Japan
| |
Collapse
|
22
|
Kistamás K, Lamberto F, Vaiciuleviciute R, Leal F, Muenthaisong S, Marte L, Subías-Beltrán P, Alaburda A, Arvanitis DN, Zana M, Costa PF, Bernotiene E, Bergaud C, Dinnyés A. The Current State of Realistic Heart Models for Disease Modelling and Cardiotoxicity. Int J Mol Sci 2024; 25:9186. [PMID: 39273136 PMCID: PMC11394806 DOI: 10.3390/ijms25179186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/18/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
One of the many unresolved obstacles in the field of cardiovascular research is an uncompromising in vitro cardiac model. While primary cell sources from animal models offer both advantages and disadvantages, efforts over the past half-century have aimed to reduce their use. Additionally, obtaining a sufficient quantity of human primary cardiomyocytes faces ethical and legal challenges. As the practically unlimited source of human cardiomyocytes from induced pluripotent stem cells (hiPSC-CM) is now mostly resolved, there are great efforts to improve their quality and applicability by overcoming their intrinsic limitations. The greatest bottleneck in the field is the in vitro ageing of hiPSC-CMs to reach a maturity status that closely resembles that of the adult heart, thereby allowing for more appropriate drug developmental procedures as there is a clear correlation between ageing and developing cardiovascular diseases. Here, we review the current state-of-the-art techniques in the most realistic heart models used in disease modelling and toxicity evaluations from hiPSC-CM maturation through heart-on-a-chip platforms and in silico models to the in vitro models of certain cardiovascular diseases.
Collapse
Affiliation(s)
- Kornél Kistamás
- BioTalentum Ltd., Aulich Lajos Str 26, H-2100 Gödöllő, Hungary
| | - Federica Lamberto
- BioTalentum Ltd., Aulich Lajos Str 26, H-2100 Gödöllő, Hungary
- Department of Physiology and Animal Health, Institute of Physiology and Animal Nutrition, Hungarian University of Agriculture and Life Sciences, Páter Károly Str 1, H-2100 Gödöllő, Hungary
| | - Raminta Vaiciuleviciute
- Department of Regenerative Medicine, State Research Institute Innovative Medicine Centre, Santariskiu g. 5, LT-08406 Vilnius, Lithuania
| | - Filipa Leal
- Biofabics Lda, Rua Alfredo Allen 455, 4200-135 Porto, Portugal
| | | | - Luis Marte
- Digital Health Unit, Eurecat-Centre Tecnològic de Catalunya, 08005 Barcelona, Spain
| | - Paula Subías-Beltrán
- Digital Health Unit, Eurecat-Centre Tecnològic de Catalunya, 08005 Barcelona, Spain
| | - Aidas Alaburda
- Department of Regenerative Medicine, State Research Institute Innovative Medicine Centre, Santariskiu g. 5, LT-08406 Vilnius, Lithuania
- Institute of Biosciences, Life Sciences Center, Vilnius University, Sauletekio al. 7, LT-10257 Vilnius, Lithuania
| | - Dina N Arvanitis
- Laboratory for Analysis and Architecture of Systems-French National Centre for Scientific Research (LAAS-CNRS), 7 Avenue du Colonel Roche, F-31400 Toulouse, France
| | - Melinda Zana
- BioTalentum Ltd., Aulich Lajos Str 26, H-2100 Gödöllő, Hungary
| | - Pedro F Costa
- Biofabics Lda, Rua Alfredo Allen 455, 4200-135 Porto, Portugal
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Innovative Medicine Centre, Santariskiu g. 5, LT-08406 Vilnius, Lithuania
- Faculty of Fundamental Sciences, Vilnius Tech, Sauletekio al. 11, LT-10223 Vilnius, Lithuania
| | - Christian Bergaud
- Laboratory for Analysis and Architecture of Systems-French National Centre for Scientific Research (LAAS-CNRS), 7 Avenue du Colonel Roche, F-31400 Toulouse, France
| | - András Dinnyés
- BioTalentum Ltd., Aulich Lajos Str 26, H-2100 Gödöllő, Hungary
- Department of Physiology and Animal Health, Institute of Physiology and Animal Nutrition, Hungarian University of Agriculture and Life Sciences, Páter Károly Str 1, H-2100 Gödöllő, Hungary
| |
Collapse
|
23
|
Doris Tsai HH, Ford LC, Burnett SD, Dickey AN, Wright FA, Chiu WA, Rusyn I. Informing Hazard Identification and Risk Characterization of Environmental Chemicals by Combining Transcriptomic and Functional Data from Human-Induced Pluripotent Stem-Cell-Derived Cardiomyocytes. Chem Res Toxicol 2024; 37:1428-1444. [PMID: 39046974 PMCID: PMC11691792 DOI: 10.1021/acs.chemrestox.4c00193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Environmental chemicals may contribute to the global burden of cardiovascular disease, but experimental data are lacking to determine which substances pose the greatest risk. Human-induced pluripotent stem cell (iPSC)-derived cardiomyocytes are a high-throughput cardiotoxicity model that is widely used to test drugs and chemicals; however, most studies focus on exploring electro-physiological readouts. Gene expression data may provide additional molecular insights to be used for both mechanistic interpretation and dose-response analyses. Therefore, we hypothesized that both transcriptomic and functional data in human iPSC-derived cardiomyocytes may be used as a comprehensive screening tool to identify potential cardiotoxicity hazards and risks of the chemicals. To test this hypothesis, we performed concentration-response analysis of 464 chemicals from 12 classes, including both pharmaceuticals and nonpharmaceutical substances. Functional effects (beat frequency, QT prolongation, and asystole), cytotoxicity, and whole transcriptome response were evaluated. Points of departure were derived from phenotypic and transcriptomic data, and risk characterization was performed. Overall, 244 (53%) substances were active in at least one phenotype; as expected, pharmaceuticals with known cardiac liabilities were the most active. Positive chronotropy was the functional phenotype activated by the largest number of tested chemicals. No chemical class was particularly prone to pose a potential hazard to cardiomyocytes; a varying proportion (10-44%) of substances in each class had effects on cardiomyocytes. Transcriptomic data showed that 69 (15%) substances elicited significant gene expression changes; most perturbed pathways were highly relevant to known key characteristics of human cardiotoxicants. The bioactivity-to-exposure ratios showed that phenotypic- and transcriptomic-based POD led to similar results for risk characterization. Overall, our findings demonstrate how the integrative use of in vitro transcriptomic and phenotypic data from iPSC-derived cardiomyocytes not only offers a complementary approach for hazard and risk prioritization, but also enables mechanistic interpretation of the in vitro test results to increase confidence in decision-making.
Collapse
Affiliation(s)
- Han-Hsuan Doris Tsai
- Interdisciplinary Faculty of Toxicology, College Station, Texas 77843, USA
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, USA
| | - Lucie C. Ford
- Interdisciplinary Faculty of Toxicology, College Station, Texas 77843, USA
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, USA
| | - Sarah D. Burnett
- Interdisciplinary Faculty of Toxicology, College Station, Texas 77843, USA
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, USA
| | - Allison N. Dickey
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, USA
| | - Fred A. Wright
- Interdisciplinary Faculty of Toxicology, College Station, Texas 77843, USA
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, USA
- Department of Statistics and Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina 27603, USA
| | - Weihsueh A. Chiu
- Interdisciplinary Faculty of Toxicology, College Station, Texas 77843, USA
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, USA
| | - Ivan Rusyn
- Interdisciplinary Faculty of Toxicology, College Station, Texas 77843, USA
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, USA
| |
Collapse
|
24
|
Pang PD, Ahmed SM, Nishiga M, Stockbridge NL, Wu JC. Tackling the challenges of new approach methods for predicting drug effects from model systems. Nat Rev Drug Discov 2024; 23:565-566. [PMID: 38750208 PMCID: PMC11482555 DOI: 10.1038/d41573-024-00081-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
The passage of the FDA Modernization Act 2.0 in 2022 has provided additional impetus to develop new approach methods for predicting the effects of drug candidates in humans from models such as microphysiological systems based on human-derived induced pluripotent stem cells. Here, we highlight progress in the field and strategies to address various challenges, including the application of artificial intelligence tools.
Collapse
|
25
|
Bezzerides V, Yoshinaga D, Feng R, Prondzynski M, Shani K, Tharani Y, Mayourian J, Joseph M, Walker D, Bortolin R, Carreon C, Boss B, Upton S, Parker K, Pu W. Dysregulation of N-terminal acetylation causes cardiac arrhythmia and cardiomyopathy. RESEARCH SQUARE 2024:rs.3.rs-3398860. [PMID: 39070617 PMCID: PMC11275982 DOI: 10.21203/rs.3.rs-3398860/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
N-terminal-acetyltransferases including NAA10 catalyze N-terminal acetylation (Nt-acetylation), an evolutionarily conserved co-translational modification. Little is known about the role of Nt-acetylation in cardiac homeostasis. To gain insights, we studied a novel NAA10 variant (p.R4S) segregating with QT-prolongation, cardiomyopathy and developmental delay in a large kindred. Here we show that the NAA10-R4S mutation reduced enzymatic activity, decreased expression levels of NAA10/NAA15 proteins, and destabilized the enzymatic complex NatA. In NAA10R4S/Y-iPSC-CMs, dysregulation of the late sodium and slow rectifying potassium currents caused severe repolarization abnormalities, consistent with clinical QT prolongation. Engineered heart tissues generated from NAA10R4S/Y-iPSC-CMs had significantly decreased contractile force and sarcomeric disorganization, consistent with the pedigree's cardiomyopathic phenotype. We identified small molecule and genetic therapies that normalized the phenotype of NAA10R4S/Y-iPSC-CMs. Our study defines novel roles of Nt-acetylation in cardiac regulation and delineates mechanisms underlying QT prolongation, arrhythmia, and cardiomyopathy caused by NAA10 dysfunction.
Collapse
Affiliation(s)
| | | | | | | | - Kevin Shani
- Harvard School of Engineering and Applied Sciences
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Satsuka A, Ribeiro AJS, Kawagishi H, Yanagida S, Hirata N, Yoshinaga T, Kurokawa J, Sugiyama A, Strauss DG, Kanda Y. Contractility assessment using aligned human iPSC-derived cardiomyocytes. J Pharmacol Toxicol Methods 2024; 128:107530. [PMID: 38917571 DOI: 10.1016/j.vascn.2024.107530] [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: 02/08/2024] [Revised: 05/17/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024]
Abstract
INTRODUCTION Cardiac safety assessment, such as lethal arrhythmias and contractility dysfunction, is critical during drug development. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have been shown to be useful in predicting drug-induced proarrhythmic risk through international validation studies. Although cardiac contractility is another key function, fit-for-purpose hiPSC-CMs in evaluating drug-induced contractile dysfunction remain poorly understood. In this study, we investigated whether alignment of hiPSC-CMs on nanopatterned culture plates can assess drug-induced contractile changes more efficiently than non-aligned monolayer culture. METHODS Aligned hiPSC-CMs were obtained by culturing on 96-well culture plates with a ridge-groove-ridge nanopattern on the bottom surface, while non-aligned hiPSC-CMs were cultured on regular 96-well plates. Next-generation sequencing and qPCR experiments were performed for gene expression analysis. Contractility of the hiPSC-CMs was assessed using an imaging-based motion analysis system. RESULTS When cultured on nanopatterned plates, hiPSC-CMs exhibited an aligned morphology and enhanced expression of genes encoding proteins that regulate contractility, including myosin heavy chain, calcium channel, and ryanodine receptor. Compared to cultures on regular plates, the aligned hiPSC-CMs also showed both enhanced contraction and relaxation velocity. In addition, the aligned hiPSC-CMs showed a more physiological response to positive and negative inotropic agents, such as isoproterenol and verapamil. DISCUSSION Taken together, the aligned hiPSC-CMs exhibited enhanced structural and functional properties, leading to an improved capacity for contractility assessment compared to the non-aligned cells. These findings suggest that the aligned hiPSC-CMs can be used to evaluate drug-induced cardiac contractile changes.
Collapse
Affiliation(s)
- Ayano Satsuka
- Division of Pharmacology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki-shi, Kanagawa 210-9501, Japan
| | - Alexandre J S Ribeiro
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Silver Spring, MD 20903, USA
| | - Hiroyuki Kawagishi
- Division of Pharmacology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki-shi, Kanagawa 210-9501, Japan
| | - Shota Yanagida
- Division of Pharmacology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki-shi, Kanagawa 210-9501, Japan
| | - Naoya Hirata
- Division of Pharmacology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki-shi, Kanagawa 210-9501, Japan
| | - Takashi Yoshinaga
- Advanced Biosignal Safety Assessment, Eisai Co., Ltd, 5-1-3 Tokodai, Tsukuba, Ibaraki 300-2635, Japan
| | - Junko Kurokawa
- Department of Bio-Informational Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka-shi, Shizuoka 422-8526, Japan
| | - Atsushi Sugiyama
- Department of Pharmacology, Faculty of Medicine, Toho University, 5-21-16 Omori-nishi, Ota-ku, Tokyo 143-8540, Japan
| | - David G Strauss
- Division of Applied Regulatory Science, Office of Translational Science, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20903, USA
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki-shi, Kanagawa 210-9501, Japan.
| |
Collapse
|
27
|
Wakatsuki T, Daily N, Hisada S, Nunomura K, Lin B, Zushida K, Honda Y, Asyama M, Takasuna K. Bayesian approach enabled objective comparison of multiple human iPSC-derived Cardiomyocytes' Proarrhythmia sensitivities. J Pharmacol Toxicol Methods 2024; 128:107531. [PMID: 38852688 DOI: 10.1016/j.vascn.2024.107531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 05/07/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024]
Abstract
The one-size-fits-all approach has been the mainstream in medicine, and the well-defined standards support the development of safe and effective therapies for many years. Advancing technologies, however, enabled precision medicine to treat a targeted patient population (e.g., HER2+ cancer). In safety pharmacology, computational population modeling has been successfully applied in virtual clinical trials to predict drug-induced proarrhythmia risks against a wide range of pseudo cohorts. In the meantime, population modeling in safety pharmacology experiments has been challenging. Here, we used five commercially available human iPSC-derived cardiomyocytes growing in 384-well plates and analyzed the effects of ten potential proarrhythmic compounds with four concentrations on their calcium transients (CaTs). All the cell lines exhibited an expected elongation or shortening of calcium transient duration with various degrees. Depending on compounds inhibiting several ion channels, such as hERG, peak and late sodium and L-type calcium or IKs channels, some of the cell lines exhibited irregular, discontinuous beating that was not predicted by computational simulations. To analyze the shapes of CaTs and irregularities of beat patterns comprehensively, we defined six parameters to characterize compound-induced CaT waveform changes, successfully visualizing the similarities and differences in compound-induced proarrhythmic sensitivities of different cell lines. We applied Bayesian statistics to predict sample populations based on experimental data to overcome the limited number of experimental replicates in high-throughput assays. This process facilitated the principal component analysis to classify compound-induced sensitivities of cell lines objectively. Finally, the association of sensitivities in compound-induced changes between phenotypic parameters and ion channel inhibitions measured using patch clamp recording was analyzed. Successful ranking of compound-induced sensitivity of cell lines was in lined with visual inspection of raw data.
Collapse
Affiliation(s)
- Tetsuro Wakatsuki
- Consortium for Safety Assessment Using Human iPS Cells (CSAHi), HEART Team, Tokyo, Japan; InvivoSciences, Inc., Madison, WI 53719, USA.
| | - Neil Daily
- InvivoSciences, Inc., Madison, WI 53719, USA
| | - Sunao Hisada
- Consortium for Safety Assessment Using Human iPS Cells (CSAHi), HEART Team, Tokyo, Japan; Hamamatsu Photonics, K.K. Systems Division, Shizuoka 431-3196, Japan
| | - Kazuto Nunomura
- Consortium for Safety Assessment Using Human iPS Cells (CSAHi), HEART Team, Tokyo, Japan; Center for Supporting Drug Discovery and Life Science Research, Graduate School of Pharmaceutical Science, Osaka University, Osaka 565-0871, Japan
| | - Bangzhong Lin
- Consortium for Safety Assessment Using Human iPS Cells (CSAHi), HEART Team, Tokyo, Japan; Center for Supporting Drug Discovery and Life Science Research, Graduate School of Pharmaceutical Science, Osaka University, Osaka 565-0871, Japan
| | - Ko Zushida
- Consortium for Safety Assessment Using Human iPS Cells (CSAHi), HEART Team, Tokyo, Japan; FUJIFILM Wako Pure Chemical Corporation, Osaka 540-8605, Japan
| | - Yayoi Honda
- Consortium for Safety Assessment Using Human iPS Cells (CSAHi), HEART Team, Tokyo, Japan; Sumika Chemical Analysis Service, Ltd. (SCAS), Osaka 554-0022, Japan
| | - Mahoko Asyama
- Consortium for Safety Assessment Using Human iPS Cells (CSAHi), HEART Team, Tokyo, Japan; Mitsubishi Tanabe Pharma Corporation, Kanagawa 251-8555, Japan
| | - Kiyoshi Takasuna
- Consortium for Safety Assessment Using Human iPS Cells (CSAHi), HEART Team, Tokyo, Japan; Daiichi Sankyo RD Novare Co., Ltd., Tokyo 134-8630, Japan; Axcelead Drug Discovery Partners, Inc., Kanagawa 251-0012, Japan
| |
Collapse
|
28
|
Guerrelli D, Pressman J, Salameh S, Posnack N. hiPSC-CM electrophysiology: impact of temporal changes and study parameters on experimental reproducibility. Am J Physiol Heart Circ Physiol 2024; 327:H12-H27. [PMID: 38727253 PMCID: PMC11390151 DOI: 10.1152/ajpheart.00631.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 04/30/2024] [Accepted: 04/30/2024] [Indexed: 05/21/2024]
Abstract
Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are frequently used for preclinical cardiotoxicity testing and remain an important tool for confirming model-based predictions of drug effects in accordance with the comprehensive in vitro proarrhythmia assay (CiPA). Despite the considerable benefits hiPSC-CMs provide, concerns surrounding experimental reproducibility have emerged. We investigated the effects of temporal changes and experimental parameters on hiPSC-CM electrophysiology. iCell cardiomyocytes2 were cultured and biosignals were acquired using a microelectrode array (MEA) system (2-14 days). Continuous recordings revealed a 22.6% increase in the beating rate and 7.7% decrease in the field potential duration (FPD) during a 20-min equilibration period. Location-specific differences across a multiwell plate were also observed, with iCell cardiomyocytes2 in the outer rows beating 8.8 beats/min faster than the inner rows. Cardiac endpoints were also impacted by cell culture duration; from 2 to 14 days, the beating rate decreased (-12.7 beats/min), FPD lengthened (+257 ms), and spike amplitude increased (+3.3 mV). Cell culture duration (4-10 days) also impacted cardiomyocyte drug responsiveness (E-4031, nifedipine, isoproterenol). qRT-PCR results suggest that daily variations in cardiac metrics may be linked to the continued maturation of hiPSC-CMs in culture (2-30 days). Daily experiments were also repeated using a second cell line (Cor.4U). Collectively, our study highlights multiple sources of variability to consider and address when performing hiPSC-CM MEA studies. To improve reproducibility and data interpretation, MEA-based studies should establish a standardized protocol and report key experimental conditions (e.g., cell line, culture time, equilibration time, electrical stimulation settings, and raw data values).NEW & NOTEWORTHY We demonstrate that iCell cardiomyocytes2 electrophysiology measurements are impacted by deviations in experimental techniques including electrical stimulation protocols, equilibration time, well-to-well variability, and length of hiPSC-CM culture. Furthermore, our results indicate that hiPSC-CM drug responsiveness changes within the first 2 wk following defrost.
Collapse
Affiliation(s)
- Devon Guerrelli
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Hospital, Washington, District of Columbia, United States
- Department of Biomedical Engineering, The George Washington University School of Engineering and Applied Science, Washington, District of Columbia, United States
- Children's National Heart Institute, Children's National Hospital, Washington, District of Columbia, United States
| | - Jenna Pressman
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Hospital, Washington, District of Columbia, United States
- Department of Biomedical Engineering, The George Washington University School of Engineering and Applied Science, Washington, District of Columbia, United States
| | - Shatha Salameh
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Hospital, Washington, District of Columbia, United States
- Children's National Heart Institute, Children's National Hospital, Washington, District of Columbia, United States
| | - Nikki Posnack
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Hospital, Washington, District of Columbia, United States
- Children's National Heart Institute, Children's National Hospital, Washington, District of Columbia, United States
- Department of Pediatrics, Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, United States
| |
Collapse
|
29
|
Pugsley MK, Winters BR, Koshman YE, Authier S, Foley CM, Hayes ES, Curtis MJ. Innovative approaches to cardiovascular safety pharmacology assessment. J Pharmacol Toxicol Methods 2024; 128:107533. [PMID: 38945308 DOI: 10.1016/j.vascn.2024.107533] [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: 06/09/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024]
Abstract
This editorial prefaces the annual themed issue on safety pharmacology (SP) methods which has been published since 2004 in the Journal of Pharmacological and Toxicological Methods (JPTM). Here we highlight content derived from the 2023 Safety Pharmacology Society (SPS) meeting held in Brussels, Belgium. The meeting generated 138 abstracts, reproduced in the current volume of JPTM. As in prior years, the manuscripts reflect various areas of innovation in SP including in silico modeling of stroke volume, cardiac output and systemic vascular resistance, computational approaches that compare drug-induced proarrhythmic sensitivity of human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), an evaluation of the utility of the corrected J-Tpeak and Tpeak-to-Tend parameters from the ECG as potential proarrhythmia biomarkers, and the applicability of nonclinical concentration-QTc (C-QTc) modeling of data derived from the conduct of the in vivo QTc study as a component of the core battery of safety pharmacology studies.
Collapse
Affiliation(s)
- Michael K Pugsley
- Toxicology & Safety Pharmacology, Cytokinetics, South San Francisco, CA 94080, United States of America.
| | - Brett R Winters
- Toxicology & Safety Pharmacology, Cytokinetics, South San Francisco, CA 94080, United States of America
| | - Yevgeniya E Koshman
- Safety Pharmacology, Abbvie, North Chicago, IL 60064, United States of America
| | - Simon Authier
- Charles River Laboratories, Laval, QC H7V 4B3, Canada
| | - C Michael Foley
- Safety Pharmacology, Abbvie, North Chicago, IL 60064, United States of America
| | - Eric S Hayes
- BioCurate Pty Ltd, Carlton, Victoria 3053, Australia
| | - Michael J Curtis
- Cardiovascular Division, King's College London, Rayne Institute, St Thomas' Hospital, London SE17EH, UK
| |
Collapse
|
30
|
Lickiss B, Hunker J, Bhagwan J, Linder P, Thomas U, Lotay H, Broadbent S, Dragicevic E, Stoelzle-Feix S, Turner J, Gossmann M. Chamber-specific contractile responses of atrial and ventricular hiPSC-cardiomyocytes to GPCR and ion channel targeting compounds: A microphysiological system for cardiac drug development. J Pharmacol Toxicol Methods 2024; 128:107529. [PMID: 38857637 DOI: 10.1016/j.vascn.2024.107529] [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: 02/26/2024] [Revised: 05/15/2024] [Accepted: 06/05/2024] [Indexed: 06/12/2024]
Abstract
Human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs) have found utility for conducting in vitro drug screening and disease modelling to gain crucial insights into pharmacology or disease phenotype. However, diseases such as atrial fibrillation, affecting >33 M people worldwide, demonstrate the need for cardiac subtype-specific cells. Here, we sought to investigate the base characteristics and pharmacological differences between commercially available chamber-specific atrial or ventricular hiPSC-CMs seeded onto ultra-thin, flexible PDMS membranes to simultaneously measure contractility in a 96 multi-well format. We investigated the effects of GPCR agonists (acetylcholine and carbachol), a Ca2+ channel agonist (S-Bay K8644), an HCN channel antagonist (ivabradine) and K+ channel antagonists (4-AP and vernakalant). We observed differential effects between atrial and ventricular hiPSC-CMs on contractile properties including beat rate, beat duration, contractile force and evidence of arrhythmias at a range of concentrations. As an excerpt of the compound analysis, S-Bay K8644 treatment showed an induced concentration-dependent transient increase in beat duration of atrial hiPSC-CMs, whereas ventricular cells showed a physiological increase in beat rate over time. Carbachol treatment produced marked effects on atrial cells, such as increased beat duration alongside a decrease in beat rate over time, but only minimal effects on ventricular cardiomyocytes. In the context of this chamber-specific pharmacology, we not only add to contractile characterization of hiPSC-CMs but propose a multi-well platform for medium-throughput early compound screening. Overall, these insights illustrate the key pharmacological differences between chamber-specific cardiomyocytes and their application on a multi-well contractility platform to gain insights for in vitro cardiac liability studies and disease modelling.
Collapse
Affiliation(s)
| | - Jan Hunker
- innoVitro GmbH, Artilleriestr 2, 52428 Jülich, Germany
| | - Jamie Bhagwan
- Axol Bioscience Ltd, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Peter Linder
- innoVitro GmbH, Artilleriestr 2, 52428 Jülich, Germany
| | - Ulrich Thomas
- Nanion Technologies GmbH, Ganghoferstr 70A, 80339 Munich, Germany
| | - Hardeep Lotay
- Axol Bioscience Ltd, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Steven Broadbent
- Axol Bioscience Ltd, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Elena Dragicevic
- Nanion Technologies GmbH, Ganghoferstr 70A, 80339 Munich, Germany
| | | | - Jan Turner
- Axol Bioscience Ltd, Babraham Research Campus, Cambridge CB22 3AT, UK
| | | |
Collapse
|
31
|
Xi NM, Huang DP. Drug safety assessment by machine learning models. J Biopharm Stat 2024:1-12. [PMID: 38888177 DOI: 10.1080/10543406.2024.2365976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 06/05/2024] [Indexed: 06/20/2024]
Abstract
The evaluation of drug-induced Torsades de pointes (TdP) risks is crucial in drug safety assessment. In this study, we discuss machine learning approaches in the prediction of drug-induced TdP risks using preclinical data. Specifically, a random forest model was trained on the dataset generated by the rabbit ventricular wedge assay. The model prediction performance was measured on 28 drugs from the Comprehensive In Vitro Proarrhythmia Assay initiative. Leave-one-drug-out cross-validation provided an unbiased estimation of model performance. Stratified bootstrap revealed the uncertainty in the asymptotic model prediction. Our study validated the utility of machine learning approaches in predicting drug-induced TdP risks from preclinical data. Our methods can be extended to other preclinical protocols and serve as a supplementary evaluation in drug safety assessment.
Collapse
Affiliation(s)
- Nan Miles Xi
- Department of Mathematics and Statistics, Loyola University Chicago, Chicago, IL, USA
| | - Dalong Patrick Huang
- Office of Biostatistics, Office of Translational Science, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
32
|
Leow JWH, Chan ECY. CYP2J2-mediated metabolism of arachidonic acid in heart: A review of its kinetics, inhibition and role in heart rhythm control. Pharmacol Ther 2024; 258:108637. [PMID: 38521247 DOI: 10.1016/j.pharmthera.2024.108637] [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: 06/18/2023] [Revised: 02/06/2024] [Accepted: 03/11/2024] [Indexed: 03/25/2024]
Abstract
Cytochrome P450 2 J2 (CYP2J2) is primarily expressed extrahepatically and is the predominant epoxygenase in human cardiac tissues. This highlights its key role in the metabolism of endogenous substrates. Significant scientific interest lies in cardiac CYP2J2 metabolism of arachidonic acid (AA), an omega-6 polyunsaturated fatty acid, to regioisomeric bioactive epoxyeicosatrienoic acid (EET) metabolites that show cardioprotective effects including regulation of cardiac electrophysiology. From an in vitro perspective, the accurate characterization of the kinetics of CYP2J2 metabolism of AA including its inhibition and inactivation by drugs could be useful in facilitating in vitro-in vivo extrapolations to predict drug-AA interactions in drug discovery and development. In this review, background information on the structure, regulation and expression of CYP2J2 in human heart is presented alongside AA and EETs as its endogenous substrate and metabolites. The in vitro and in vivo implications of the kinetics of this endogenous metabolic pathway as well as its perturbation via inhibition and inactivation by drugs are elaborated. Additionally, the role of CYP2J2-mediated metabolism of AA to EETs in cardiac electrophysiology will be expounded.
Collapse
Affiliation(s)
- Jacqueline Wen Hui Leow
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore
| | - Eric Chun Yong Chan
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore.
| |
Collapse
|
33
|
Wu W, Sun J, Zhang J, Zhao H, Qiu S, Li C, Shi C, Xu Y. Phosphoproteomics reveals a novel mechanism underlying the proarrhythmic effects of nilotinib, vandetanib, and mobocertinib. Toxicology 2024; 505:153830. [PMID: 38754619 DOI: 10.1016/j.tox.2024.153830] [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: 03/11/2024] [Revised: 04/30/2024] [Accepted: 05/13/2024] [Indexed: 05/18/2024]
Abstract
The use of tyrosine kinase inhibitors (TKIs) has resulted in significant occurrence of arrhythmias. However, the precise mechanism of the proarrhythmic effect is not fully understood. In this study, we found that nilotinib (NIL), vandetanib (VAN), and mobocertinib (MOB) induced the development of "cellrhythmia" (arrhythmia-like events) in a concentration-dependent manner in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Continuous administration of NIL, VAN, or MOB in animals significantly prolonged the action potential durations (APD) and increased susceptibility to arrhythmias. Using phosphoproteomic analysis, we identified proteins with altered phosphorylation levels after treatment with 3 μM NIL, VAN, and MOB for 1.5 h. Using these identified proteins as substrates, we performed kinase-substrate enrichment analysis to identify the kinases driving the changes in phosphorylation levels of these proteins. MAPK and WNK were both inhibited by NIL, VAN, and MOB. A selective inhibitor of WNK1, WNK-IN-11, induced concentration- and time-dependent cellrhythmias and prolonged field potential duration (FPD) in hiPSC-CMs in vitro; furthermore, administration in guinea pigs confirmed that WNK-IN-11 prolonged ventricular repolarization and increased susceptibility to arrhythmias. Fingding indicated that WNK1 inhibition had an in vivo and in vitro arrhythmogenic phenotype similar to TKIs. Additionally,three of TKIs reduced hERG and KCNQ1 expression at protein level, not at transcription level. Similarly, the knockdown of WNK1 decreased hERG and KCNQ1 protein expression in hiPSC-CMs. Collectively, our data suggest that the proarrhythmic effects of NIL, VAN, and MOB occur through a kinase inhibition mechanism. NIL, VAN, and MOB inhibit WNK1 kinase, leading to a decrease in hERG and KCNQ1 protein expression, thereby prolonging action potential repolarization and consequently cause arrhythmias.
Collapse
Affiliation(s)
- Wenting Wu
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang 050017, China
| | - Jinglei Sun
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang 050017, China
| | - Jiali Zhang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang 050017, China
| | - Haining Zhao
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang 050017, China
| | - Suhua Qiu
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang 050017, China
| | - Congxin Li
- Department of Pharmacy, Third Hospital of Hebei Medical University, Shijiazhuang 050051, China
| | - Chenxia Shi
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang 050017, China
| | - Yanfang Xu
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang 050017, China.
| |
Collapse
|
34
|
Lee HA, Woo DH, Lim DS, Oh J, Kim CY, Bae ON, Ahn SJ. Guidelines for Manufacturing and Application of Organoids: Heart. Int J Stem Cells 2024; 17:130-140. [PMID: 38777829 PMCID: PMC11170119 DOI: 10.15283/ijsc24046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
Cardiac organoids have emerged as invaluable tools for assessing the impact of diverse substances on heart function. This report introduces guidelines for general requirements for manufacturing cardiac organoids and conducting cardiac organoid-based assays, encompassing protocols, analytical methodologies, and ethical considerations. In the quest to employ recently developed three-dimensional cardiac organoid models as substitutes for animal testing, it becomes imperative to establish robust criteria for evaluating organoid quality and conducting toxicity assessments. This guideline addresses this need, catering to regulatory requirements, and describes common standards for organoid quality and toxicity assessment methodologies, commensurate with current technological capabilities. While acknowledging the dynamic nature of technological progress and the potential for future comparative studies, this guideline serves as a foundational framework. It offers a comprehensive approach to standardized cardiac organoid testing, ensuring scientific rigor, reproducibility, and ethical integrity in investigations of cardiotoxicity, particularly through the utilization of human pluripotent stem cell-derived cardiac organoids.
Collapse
Affiliation(s)
- Hyang-Ae Lee
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, Korea
- Organoid Standards Initiative
| | - Dong-Hun Woo
- Organoid Standards Initiative
- Department of Commercializing Organoid Technology, NEXEL Co., Ltd., Seoul, Korea
| | - Do-Sun Lim
- Organoid Standards Initiative
- Department of Cardiology, Korea University Anam Hospital, Seoul, Korea
| | - Jisun Oh
- Organoid Standards Initiative
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - C-Yoon Kim
- Organoid Standards Initiative
- College of Veterinary Medicine, Konkuk University, Seoul, Korea
| | - Ok-Nam Bae
- Organoid Standards Initiative
- College of Pharmacy, Hanyang University, Ansan, Korea
| | - Sun-Ju Ahn
- Organoid Standards Initiative
- Department of Biophysics, Sungkyunkwan University, Suwon, Korea
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Korea
| |
Collapse
|
35
|
Yin DE, Palin AC, Lombo TB, Mahon RN, Poon B, Wu DY, Atala A, Brooks KM, Chen S, Coyne CB, D’Souza MP, Fackler OT, Furler O’Brien RL, Garcia-de-Alba C, Jean-Philippe P, Karn J, Majji S, Muotri AR, Ozulumba T, Sakatis MZ, Schlesinger LS, Singh A, Spiegel HM, Struble E, Sung K, Tagle DA, Thacker VV, Tidball AM, Varthakavi V, Vunjak-Novakovic G, Wagar LE, Yeung CK, Ndhlovu LC, Ott M. 3D human tissue models and microphysiological systems for HIV and related comorbidities. Trends Biotechnol 2024; 42:526-543. [PMID: 38071144 PMCID: PMC11065605 DOI: 10.1016/j.tibtech.2023.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/22/2023] [Accepted: 10/24/2023] [Indexed: 03/03/2024]
Abstract
Three-dimensional (3D) human tissue models/microphysiological systems (e.g., organs-on-chips, organoids, and tissue explants) model HIV and related comorbidities and have potential to address critical questions, including characterization of viral reservoirs, insufficient innate and adaptive immune responses, biomarker discovery and evaluation, medical complexity with comorbidities (e.g., tuberculosis and SARS-CoV-2), and protection and transmission during pregnancy and birth. Composed of multiple primary or stem cell-derived cell types organized in a dedicated 3D space, these systems hold unique promise for better reproducing human physiology, advancing therapeutic development, and bridging the human-animal model translational gap. Here, we discuss the promises and achievements with 3D human tissue models in HIV and comorbidity research, along with remaining barriers with respect to cell biology, virology, immunology, and regulatory issues.
Collapse
|
36
|
Dow R, DeLong C, Jiang G, Attili D, Creech J, Kraan R, Campbell K, Saraithong P, O’Shea S, Monteiro da Rocha A, McInnis MG, Herron TJ. Bipolar Patient-Specific In Vitro Diagnostic Test Reveals Underlying Cardiac Arrhythmia Phenotype Caused by Calcium Channel Genetic Risk Factor. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:100296. [PMID: 38560725 PMCID: PMC10978474 DOI: 10.1016/j.bpsgos.2024.100296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/12/2024] [Accepted: 02/02/2024] [Indexed: 04/04/2024] Open
Abstract
A common genetic risk factor for bipolar disorder is CACNA1C, a gene that is also critical for cardiac rhythm. The impact of CACNA1C mutations on bipolar patient cardiac rhythm is unknown. Here, we report the cardiac electrophysiological implications of a bipolar disorder-associated genetic risk factor in CACNA1C using patient induced pluripotent stem cell-derived cardiomyocytes. Results indicate that the CACNA1C bipolar disorder-related mutation causes cardiac electrical impulse conduction slowing mediated by impaired intercellular coupling via connexin 43 gap junctions. In vitro gene therapy to restore connexin 43 expression increased cardiac electrical impulse conduction velocity and protected against thioridazine-induced QT prolongation. Patients positive for bipolar disorder CACNA1C genetic risk factors may have elevated proarrhythmic risk for adverse events in response to psychiatric medications that slow conduction or prolong the QT interval. This in vitro diagnostic tool enables cardiac testing specific to patients with psychiatric disorders to determine their sensitivity to off-target effects of psychiatric medications.
Collapse
Affiliation(s)
- Rachel Dow
- Frankel Cardiovascular Regeneration Core Laboratory, University of Michigan, Ann Arbor, Michigan
| | - Cindy DeLong
- Michigan Medicine, Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - Guihua Jiang
- Michigan Medicine, Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - Durga Attili
- Michigan Medicine, Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - Jeffery Creech
- Frankel Cardiovascular Regeneration Core Laboratory, University of Michigan, Ann Arbor, Michigan
| | - Rachel Kraan
- Frankel Cardiovascular Regeneration Core Laboratory, University of Michigan, Ann Arbor, Michigan
| | - Katherine Campbell
- Michigan Medicine, Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - Prakaimuk Saraithong
- Frankel Cardiovascular Regeneration Core Laboratory, University of Michigan, Ann Arbor, Michigan
- Michigan Medicine, Internal Medicine-Cardiology, University of Michigan, Ann Arbor, Michigan
| | - Sue O’Shea
- Michigan Medicine, Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan
- Michigan Medicine, Psychiatry Department, University of Michigan, Ann Arbor, Michigan
| | - Andre Monteiro da Rocha
- Frankel Cardiovascular Regeneration Core Laboratory, University of Michigan, Ann Arbor, Michigan
- Michigan Medicine, Internal Medicine-Cardiology, University of Michigan, Ann Arbor, Michigan
| | - Melvin G. McInnis
- Michigan Medicine, Psychiatry Department, University of Michigan, Ann Arbor, Michigan
| | - Todd J. Herron
- Frankel Cardiovascular Regeneration Core Laboratory, University of Michigan, Ann Arbor, Michigan
- Michigan Medicine, Internal Medicine-Cardiology, University of Michigan, Ann Arbor, Michigan
- Michigan Medicine, Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
37
|
Feaster TK, Ewoldt JK, Avila A, Casciola M, Narkar A, Chen CS, Blinova K. Nonclinical evaluation of chronic cardiac contractility modulation on 3D human engineered cardiac tissues. J Cardiovasc Electrophysiol 2024; 35:895-905. [PMID: 38433304 DOI: 10.1111/jce.16222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/29/2024] [Accepted: 02/09/2024] [Indexed: 03/05/2024]
Abstract
INTRODUCTION Cardiac contractility modulation (CCM) is a medical device-based therapy delivering non-excitatory electrical stimulations to the heart to enhance cardiac function in heart failure (HF) patients. The lack of human in vitro tools to assess CCM hinders our understanding of CCM mechanisms of action. Here, we introduce a novel chronic (i.e., 2-day) in vitro CCM assay to evaluate the effects of CCM in a human 3D microphysiological system consisting of engineered cardiac tissues (ECTs). METHODS Cryopreserved human induced pluripotent stem cell-derived cardiomyocytes were used to generate 3D ECTs. The ECTs were cultured, incorporating human primary ventricular cardiac fibroblasts and a fibrin-based gel. Electrical stimulation was applied using two separate pulse generators for the CCM group and control group. Contractile properties and intracellular calcium were measured, and a cardiac gene quantitative PCR screen was conducted. RESULTS Chronic CCM increased contraction amplitude and duration, enhanced intracellular calcium transient amplitude, and altered gene expression related to HF (i.e., natriuretic peptide B, NPPB) and excitation-contraction coupling (i.e., sodium-calcium exchanger, SLC8). CONCLUSION These data represent the first study of chronic CCM in a 3D ECT model, providing a nonclinical tool to assess the effects of cardiac electrophysiology medical device signals complementing in vivo animal studies. The methodology established a standardized 3D ECT-based in vitro testbed for chronic CCM, allowing evaluation of physiological and molecular effects on human cardiac tissues.
Collapse
Affiliation(s)
- Tromondae K Feaster
- Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jourdan K Ewoldt
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
| | - Anna Avila
- Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Maura Casciola
- Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Akshay Narkar
- Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Christopher S Chen
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA
| | - Ksenia Blinova
- Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
38
|
Pierson JB, Berridge B, Blinova K, Brooks MB, Eldridge S, O'Brien CE, Pugsley MK, Schultze AE, Smith G, Stockbridge N, Valentin JP, Vicente J. Collaborative science in action: A 20 year perspective from the Health and Environmental Sciences Institute (HESI) Cardiac Safety Committee. J Pharmacol Toxicol Methods 2024; 127:107511. [PMID: 38710237 PMCID: PMC11318526 DOI: 10.1016/j.vascn.2024.107511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/02/2024] [Accepted: 05/02/2024] [Indexed: 05/08/2024]
Abstract
The Health and Environmental Sciences Institute (HESI) is a nonprofit organization dedicated to resolving global health challenges through collaborative scientific efforts across academia, regulatory authorities and the private sector. Collaborative science across non-clinical disciplines offers an important keystone to accelerate the development of safer and more effective medicines. HESI works to address complex challenges by leveraging diverse subject-matter expertise across sectors offering access to resources, data and shared knowledge. In 2008, the HESI Cardiac Safety Committee (CSC) was established to improve public health by reducing unanticipated cardiovascular (CV)-related adverse effects from pharmaceuticals or chemicals. The committee continues to significantly impact the field of CV safety by bringing together experts from across sectors to address challenges of detecting and predicting adverse cardiac outcomes. Committee members have collaborated on the organization, management and publication of prospective studies, retrospective analyses, workshops, and symposia resulting in 38 peer reviewed manuscripts. Without this collaboration these manuscripts would not have been published. Through their work, the CSC is actively addressing challenges and opportunities in detecting potential cardiac failure modes using in vivo, in vitro and in silico models, with the aim of facilitating drug development and improving study design. By examining past successes and future prospects of the CSC, this manuscript sheds light on how the consortium's multifaceted approach not only addresses current challenges in detecting potential cardiac failure modes but also paves the way for enhanced drug development and study design methodologies. Further, exploring future opportunities and challenges will focus on improving the translational predictability of nonclinical evaluations and reducing reliance on animal research in CV safety assessments.
Collapse
Affiliation(s)
| | | | | | - Marjory B Brooks
- Comparative Coagulation Section, Department of Population Medicine and Diagnostic Sciences, Cornell University, Ithaca, NY, USA
| | - Sandy Eldridge
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Claire E O'Brien
- Health and Environmental Sciences Institute, Washington, DC, USA.
| | - Michael K Pugsley
- Toxicology & Safety Pharmacology, Cytokinetics, South San Francisco, CA, USA
| | - A Eric Schultze
- Pathology, Lilly Research Laboratories, Indianapolis, IN, USA
| | - Godfrey Smith
- Clyde Biosciences Ltd, Newhouse, UK; University of Glasgow, Scotland, UK
| | | | - Jean-Pierre Valentin
- UCB Biopharma SRL, Development Science, Non-Clinical Safety Evaluation, Braine l'Alleud, Belgium
| | - Jose Vicente
- Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
39
|
Clark AP, Wei S, Fullerton K, Krogh-Madsen T, Christini DJ. Single-cell ionic current phenotyping explains stem cell-derived cardiomyocyte action potential morphology. Am J Physiol Heart Circ Physiol 2024; 326:H1146-H1154. [PMID: 38488520 PMCID: PMC11380975 DOI: 10.1152/ajpheart.00063.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 02/26/2024] [Accepted: 03/01/2024] [Indexed: 04/14/2024]
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) are a promising tool to study arrhythmia-related factors, but the variability of action potential (AP) recordings from these cells limits their use as an in vitro model. In this study, we use recently published brief (10 s), dynamic voltage-clamp (VC) data to provide mechanistic insights into the ionic currents contributing to AP heterogeneity; we call this approach rapid ionic current phenotyping (RICP). Features of this VC data were correlated to AP recordings from the same cells, and we used computational models to generate mechanistic insights into cellular heterogeneity. This analysis uncovered several interesting links between AP morphology and ionic current density: both L-type calcium and sodium currents contribute to upstroke velocity, rapid delayed rectifier K+ current is the main determinant of the maximal diastolic potential, and an outward current in the activation range of slow delayed rectifier K+ is the main determinant of AP duration. Our analysis also identified an outward current in several cells at 6 mV that is not reproduced by iPSC-CM mathematical models but contributes to determining AP duration. RICP can be used to explain how cell-to-cell variability in ionic currents gives rise to AP heterogeneity. Because of its brief duration (10 s) and ease of data interpretation, we recommend the use of RICP for single-cell patch-clamp experiments that include the acquisition of APs.NEW & NOTEWORTHY We present rapid ionic current phenotyping (RICP), a current quantification approach based on an optimized voltage-clamp protocol. The method captures a rich snapshot of the ionic current dynamics, providing quantitative information about multiple currents (e.g., ICa,L, IKr) in the same cell. The protocol helped to identify key ionic determinants of cellular action potential heterogeneity in iPSC-CMs. This included unexpected results, such as the critical role of IKr in establishing the maximum diastolic potential.
Collapse
Affiliation(s)
- Alexander P Clark
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, United States
| | - Siyu Wei
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, New York, United States
| | - Kristin Fullerton
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York, United States
| | - Trine Krogh-Madsen
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York, United States
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, New York, United States
| | - David J Christini
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, United States
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, New York, United States
| |
Collapse
|
40
|
Grandits T, Augustin CM, Haase G, Jost N, Mirams GR, Niederer SA, Plank G, Varró A, Virág L, Jung A. Neural network emulation of the human ventricular cardiomyocyte action potential for more efficient computations in pharmacological studies. eLife 2024; 12:RP91911. [PMID: 38598284 PMCID: PMC11006416 DOI: 10.7554/elife.91911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024] Open
Abstract
Computer models of the human ventricular cardiomyocyte action potential (AP) have reached a level of detail and maturity that has led to an increasing number of applications in the pharmaceutical sector. However, interfacing the models with experimental data can become a significant computational burden. To mitigate the computational burden, the present study introduces a neural network (NN) that emulates the AP for given maximum conductances of selected ion channels, pumps, and exchangers. Its applicability in pharmacological studies was tested on synthetic and experimental data. The NN emulator potentially enables massive speed-ups compared to regular simulations and the forward problem (find drugged AP for pharmacological parameters defined as scaling factors of control maximum conductances) on synthetic data could be solved with average root-mean-square errors (RMSE) of 0.47 mV in normal APs and of 14.5 mV in abnormal APs exhibiting early afterdepolarizations (72.5% of the emulated APs were alining with the abnormality, and the substantial majority of the remaining APs demonstrated pronounced proximity). This demonstrates not only very fast and mostly very accurate AP emulations but also the capability of accounting for discontinuities, a major advantage over existing emulation strategies. Furthermore, the inverse problem (find pharmacological parameters for control and drugged APs through optimization) on synthetic data could be solved with high accuracy shown by a maximum RMSE of 0.22 in the estimated pharmacological parameters. However, notable mismatches were observed between pharmacological parameters estimated from experimental data and distributions obtained from the Comprehensive in vitro Proarrhythmia Assay initiative. This reveals larger inaccuracies which can be attributed particularly to the fact that small tissue preparations were studied while the emulator was trained on single cardiomyocyte data. Overall, our study highlights the potential of NN emulators as powerful tool for an increased efficiency in future quantitative systems pharmacology studies.
Collapse
Affiliation(s)
- Thomas Grandits
- Department of Mathematics and Scientific Computing, University of GrazGrazAustria
- NAWI Graz, University of GrazGrazAustria
| | - Christoph M Augustin
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging - Division of Medical Physics and Biophysics, Medical University of GrazGrazAustria
- BioTechMed-GrazGrazAustria
| | - Gundolf Haase
- Department of Mathematics and Scientific Computing, University of GrazGrazAustria
| | - Norbert Jost
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
- HUN-REN-TKI, Research Group of PharmacologyBudapestHungary
| | - Gary R Mirams
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of NottinghamNottinghamUnited Kingdom
| | - Steven A Niederer
- Division of Imaging Sciences & Biomedical Engineering, King’s College LondonLondonUnited Kingdom
| | - Gernot Plank
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging - Division of Medical Physics and Biophysics, Medical University of GrazGrazAustria
- BioTechMed-GrazGrazAustria
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
- HUN-REN-TKI, Research Group of PharmacologyBudapestHungary
| | - László Virág
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | - Alexander Jung
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging - Division of Medical Physics and Biophysics, Medical University of GrazGrazAustria
| |
Collapse
|
41
|
Min S, Kim S, Sim WS, Choi YS, Joo H, Park JH, Lee SJ, Kim H, Lee MJ, Jeong I, Cui B, Jo SH, Kim JJ, Hong SB, Choi YJ, Ban K, Kim YG, Park JU, Lee HA, Park HJ, Cho SW. Versatile human cardiac tissues engineered with perfusable heart extracellular microenvironment for biomedical applications. Nat Commun 2024; 15:2564. [PMID: 38519491 PMCID: PMC10960018 DOI: 10.1038/s41467-024-46928-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 03/13/2024] [Indexed: 03/25/2024] Open
Abstract
Engineered human cardiac tissues have been utilized for various biomedical applications, including drug testing, disease modeling, and regenerative medicine. However, the applications of cardiac tissues derived from human pluripotent stem cells are often limited due to their immaturity and lack of functionality. Therefore, in this study, we establish a perfusable culture system based on in vivo-like heart microenvironments to improve human cardiac tissue fabrication. The integrated culture platform of a microfluidic chip and a three-dimensional heart extracellular matrix enhances human cardiac tissue development and their structural and functional maturation. These tissues are comprised of cardiovascular lineage cells, including cardiomyocytes and cardiac fibroblasts derived from human induced pluripotent stem cells, as well as vascular endothelial cells. The resultant macroscale human cardiac tissues exhibit improved efficacy in drug testing (small molecules with various levels of arrhythmia risk), disease modeling (Long QT Syndrome and cardiac fibrosis), and regenerative therapy (myocardial infarction treatment). Therefore, our culture system can serve as a highly effective tissue-engineering platform to provide human cardiac tissues for versatile biomedical applications.
Collapse
Affiliation(s)
- Sungjin Min
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Suran Kim
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
- Cellartgen, Seoul, 03722, Republic of Korea
| | - Woo-Sup Sim
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Yi Sun Choi
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyebin Joo
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jae-Hyun Park
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Su-Jin Lee
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Hyeok Kim
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Mi Jeong Lee
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Inhea Jeong
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Baofang Cui
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Sung-Hyun Jo
- Department of Chemical Engineering, Soongsil University, Seoul, 06978, Republic of Korea
| | - Jin-Ju Kim
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Seok Beom Hong
- Department of Thoracic and Cardiovascular Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Yeon-Jik Choi
- Division of Cardiology, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 03312, Republic of Korea
| | - Kiwon Ban
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, 999077, Hong Kong
| | - Yun-Gon Kim
- Department of Chemical Engineering, Soongsil University, Seoul, 06978, Republic of Korea
| | - Jang-Ung Park
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyang-Ae Lee
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Hun-Jun Park
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
- Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
- Cellartgen, Seoul, 03722, Republic of Korea.
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, Republic of Korea.
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
42
|
Raniga K, Nasir A, Vo NTN, Vaidyanathan R, Dickerson S, Hilcove S, Mosqueira D, Mirams GR, Clements P, Hicks R, Pointon A, Stebbeds W, Francis J, Denning C. Strengthening cardiac therapy pipelines using human pluripotent stem cell-derived cardiomyocytes. Cell Stem Cell 2024; 31:292-311. [PMID: 38366587 DOI: 10.1016/j.stem.2024.01.007] [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: 09/14/2023] [Revised: 11/27/2023] [Accepted: 01/19/2024] [Indexed: 02/18/2024]
Abstract
Advances in hiPSC isolation and reprogramming and hPSC-CM differentiation have prompted their therapeutic application and utilization for evaluating potential cardiovascular safety liabilities. In this perspective, we showcase key efforts toward the large-scale production of hiPSC-CMs, implementation of hiPSC-CMs in industry settings, and recent clinical applications of this technology. The key observations are a need for traceable gender and ethnically diverse hiPSC lines, approaches to reduce cost of scale-up, accessible clinical trial datasets, and transparent guidelines surrounding the safety and efficacy of hiPSC-based therapies.
Collapse
Affiliation(s)
- Kavita Raniga
- The Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK; Pathology, Non-Clinical Safety, GlaxoSmithKline R&D, Stevenage SG1 2NY, UK.
| | - Aishah Nasir
- The Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Nguyen T N Vo
- The Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | | | | | | | - Diogo Mosqueira
- The Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Gary R Mirams
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | - Peter Clements
- Pathology, Non-Clinical Safety, GlaxoSmithKline R&D, Stevenage SG1 2NY, UK
| | - Ryan Hicks
- BioPharmaceuticals R&D Cell Therapy Department, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London WC2R 2LS, UK
| | - Amy Pointon
- Safety Sciences, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB2 0AA, UK
| | | | - Jo Francis
- Mechanstic Biology and Profiling, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB2 0AA, UK
| | - Chris Denning
- The Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK.
| |
Collapse
|
43
|
Lee SG, Song GE, Seok J, Kim J, Kim MW, Rhee J, Park S, Jeong KS, Lee S, Lee YH, Jeong Y, Chung HM, Kim CY. Evaluation of the cardiotoxicity potential of bisphenol analogues in human induced pluripotent stem cells derived cardiomyocytes. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 272:116108. [PMID: 38364764 DOI: 10.1016/j.ecoenv.2024.116108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 01/31/2024] [Accepted: 02/09/2024] [Indexed: 02/18/2024]
Abstract
The importance of evaluating the cardiotoxicity potential of common chemicals as well as new drugs is increasing as a result of the development of animal alternative test methods using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM). Bisphenol A (BPA), which is used as a main material in plastics, is known as an endocrine-disrupting chemical, and recently reported to cause cardiotoxicity through inhibition of ion channels in CMs even with acute exposure. Accordingly, the need for the development of alternatives to BPA has been highlighted, and structural analogues including bisphenol AF, C, E, F, and S have been developed. However, cardiotoxicity data for analogues of bisphenol are not well known. In this study, in order to evaluate the cardiotoxicity potential of analogues, including BPA, a survival test of hiPSC-CMs and a dual-cardiotoxicity evaluation based on a multi-electrode array were performed. Acute exposure to all bisphenol analogues did not affect survival rate, but spike amplitude, beat period, and field potential duration were decreased in a dose-dependent manner in most of the bisphenols except bisphenol S. In addition, bisphenols, except for bisphenol S, reduced the contractile force of hiPSC-CMs and resulted in beating arrest at high doses. Taken together, it can be suggested that the developed bisphenol analogues could cause cardiotoxicity even with acute exposure, and it is considered that the application of the MEA-based dual-cardiotoxicity evaluation method can be an effective help in the development of safe alternatives.
Collapse
Affiliation(s)
- Seul-Gi Lee
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, Seoul 05029, Republic of Korea; College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Gyeong-Eun Song
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Jin Seok
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Jin Kim
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Min Woo Kim
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Jooeon Rhee
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Shinhye Park
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Kyu Sik Jeong
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Suemin Lee
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Yun Hyeong Lee
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Youngin Jeong
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Hyung Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, Seoul 05029, Republic of Korea; Miraecell Bio Co. Ltd., Seoul 04795, Republic of Korea
| | - C-Yoon Kim
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea.
| |
Collapse
|
44
|
Kussauer S, Dilk P, Elleisy M, Michaelis C, Lichtwark S, Rimmbach C, David R, Jung J. Heart rhythm in vitro: measuring stem cell-derived pacemaker cells on microelectrode arrays. Front Cardiovasc Med 2024; 11:1200786. [PMID: 38450366 PMCID: PMC10915086 DOI: 10.3389/fcvm.2024.1200786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 02/05/2024] [Indexed: 03/08/2024] Open
Abstract
Background Cardiac arrhythmias have markedly increased in recent decades, highlighting the urgent need for appropriate test systems to evaluate the efficacy and safety of new pharmaceuticals and the potential side effects of established drugs. Methods The Microelectrode Array (MEA) system may be a suitable option, as it provides both real-time and non-invasive monitoring of cellular networks of spontaneously active cells. However, there is currently no commercially available cell source to apply this technology in the context of the cardiac conduction system (CCS). In response to this problem, our group has previously developed a protocol for the generation of pure functional cardiac pacemaker cells from mouse embryonic stem cells (ESCs). In addition, we compared the hanging drop method, which was previously utilized, with spherical plate-derived embryoid bodies (EBs) and the pacemaker cells that are differentiated from these. Results We described the application of these pacemaker cells on the MEA platform, which required a number of crucial optimization steps in terms of coating, dissociation, and cell density. As a result, we were able to generate a monolayer of pure pacemaker cells on an MEA surface that is viable and electromechanically active for weeks. Furthermore, we introduced spherical plates as a convenient and scalable method to be applied for the production of induced sinoatrial bodies. Conclusion We provide a tool to transfer modeling and analysis of cardiac rhythm diseases to the cell culture dish. Our system allows answering CCS-related queries within a cellular network, both under baseline conditions and post-drug exposure in a reliable and affordable manner. Ultimately, our approach may provide valuable guidance not only for cardiac pacemaker cells but also for the generation of an MEA test platform using other sensitive non-proliferating cell types.
Collapse
Affiliation(s)
- Sophie Kussauer
- Department of Cardiac Surgery, Rostock University Medical Centre, Rostock, Germany
- Department of Life, Light, & Matter, University of Rostock, Rostock, Germany
| | - Patrick Dilk
- Department of Cardiac Surgery, Rostock University Medical Centre, Rostock, Germany
- Department of Life, Light, & Matter, University of Rostock, Rostock, Germany
| | - Moustafa Elleisy
- Department of Cardiac Surgery, Rostock University Medical Centre, Rostock, Germany
- Department of Life, Light, & Matter, University of Rostock, Rostock, Germany
| | - Claudia Michaelis
- Department of Cardiac Surgery, Rostock University Medical Centre, Rostock, Germany
- Department of Life, Light, & Matter, University of Rostock, Rostock, Germany
| | - Sarina Lichtwark
- Department of Cardiac Surgery, Rostock University Medical Centre, Rostock, Germany
- Department of Life, Light, & Matter, University of Rostock, Rostock, Germany
| | - Christian Rimmbach
- Department of Cardiac Surgery, Rostock University Medical Centre, Rostock, Germany
- Department of Life, Light, & Matter, University of Rostock, Rostock, Germany
| | - Robert David
- Department of Cardiac Surgery, Rostock University Medical Centre, Rostock, Germany
- Department of Life, Light, & Matter, University of Rostock, Rostock, Germany
| | - Julia Jung
- Department of Cardiac Surgery, Rostock University Medical Centre, Rostock, Germany
- Department of Life, Light, & Matter, University of Rostock, Rostock, Germany
| |
Collapse
|
45
|
Li J, Hua Y, Liu Y, Qu X, Zhang J, Ishida M, Yoshida N, Tabata A, Miyoshi H, Shiba M, Higo S, Sougawa N, Takeda M, Kawamura T, Matsuura R, Okuzaki D, Toyofuku T, Sawa Y, Liu L, Miyagawa S. Human induced pluripotent stem cell-derived closed-loop cardiac tissue for drug assessment. iScience 2024; 27:108992. [PMID: 38333703 PMCID: PMC10850789 DOI: 10.1016/j.isci.2024.108992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/16/2023] [Accepted: 01/18/2024] [Indexed: 02/10/2024] Open
Abstract
Human iPSC-derived cardiomyocytes (hiPSC-CMs) exhibit functional immaturity, potentially impacting their suitability for assessing drug proarrhythmic potential. We previously devised a traveling wave (TW) system to promote maturation in 3D cardiac tissue. To align with current drug assessment paradigms (CiPA and JiCSA), necessitating a 2D monolayer cardiac tissue, we integrated the TW system with a multi-electrode array. This gave rise to a hiPSC-derived closed-loop cardiac tissue (iCT), enabling spontaneous TW initiation and swift pacing of cardiomyocytes from various cell lines. The TW-paced cardiomyocytes demonstrated heightened sarcomeric and functional maturation, exhibiting enhanced response to isoproterenol. Moreover, these cells showcased diminished sensitivity to verapamil and maintained low arrhythmia rates with ranolazine-two drugs associated with a low risk of torsades de pointes (TdP). Notably, the TW group displayed increased arrhythmia rates with high and intermediate risk TdP drugs (quinidine and pimozide), underscoring the potential utility of this system in drug assessment applications.
Collapse
Affiliation(s)
- Junjun Li
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Ying Hua
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yuting Liu
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Xiang Qu
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Jingbo Zhang
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Masako Ishida
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Noriko Yoshida
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Akiko Tabata
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Hayato Miyoshi
- Fujifilm Corporation, Ashigarakami 258-8577, Kanagawa, Japan
| | - Mikio Shiba
- Cardiovascular Division, Osaka Police Hospital, Tennoji 543-0035, Osaka, Japan
| | - Shuichiro Higo
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita 565-0871, Osaka, Japan
- Department of Medical Therapeutics for Heart Failure, Osaka University Graduate School of Medicine, Suita 565-0871, Osaka, Japan
| | - Nagako Sougawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
- Department of Physiology, Osaka Dental University, 8-1 Kuzuha Hanazono-cho, Hirakata 573-1121, Osaka, Japan
| | - Maki Takeda
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Takuji Kawamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Ryohei Matsuura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Daisuke Okuzaki
- Laboratory of Human Immunology (Single Cell Genomics), WPI Immunology Research Center, Osaka University, Osaka, Japan
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Toshihiko Toyofuku
- Department of Immunology and Molecular Medicine, Graduate School of Medicine, Osaka University, Suita 565-0871, Osaka, Japan
| | - Yoshiki Sawa
- Department of Future Medicine, Division of Health Science, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Li Liu
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| |
Collapse
|
46
|
Lin HC, Rusyn I, Chiu WA. Assessing proarrhythmic potential of environmental chemicals using a high throughput in vitro-in silico model with human induced pluripotent stem cell-derived cardiomyocytes. ALTEX 2024; 41:37-49. [PMID: 37921411 PMCID: PMC10898275 DOI: 10.14573/altex.2306231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 10/19/2023] [Indexed: 11/04/2023]
Abstract
QT prolongation and the potentially fatal arrhythmia Torsades de Pointes are common causes for withdrawing or restricting drugs; however, little is known about similar liabilities of environmental chemicals. Current in vitro-in silico models for testing proarrhythmic liabilities, using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM), provide an opportunity to address this data gap. These methods are still low- to medium-throughput and not suitable for testing the tens of thousands of chemicals in commerce. We hypothesized that combining high-throughput population- based in vitro testing in hiPSC-CMs with a fully in silico data analysis workflow can offer sensitive and specific predictions of proarrhythmic potential. We calibrated the model with a published hiPSC-CM dataset of drugs known to be positive or negative for proarrhythmia and tested its performance using internal cross-validation and external validation. Additionally, we used computational down-sampling to examine three study designs for hiPSC-CM data: one replicate of one donor, five replicates of one donor, and one replicate of a population of five donors. We found that the population of five donors had the best performance for predicting proarrhythmic potential. The resulting model was then applied to predict the proarrhythmic potential of environmental chemicals, additionally characterizing risk through margin of exposure (MOE) calculations. Out of over 900 environmental chemicals tested, over 150 were predicted to have proarrhythmic potential, but only seven chemicals had a MOE < 1. We conclude that a high-throughput in vitro-in silico approach using population-based hiPSC-CM testing provides a reasonable strategy to screen environmental chemicals for proarrhythmic potential.
Collapse
Affiliation(s)
- Hsing-Chieh Lin
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Ivan Rusyn
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Weihsueh A. Chiu
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| |
Collapse
|
47
|
Lee SG, Kim J, Seok J, Kim MW, Rhee J, Song GE, Park S, Lee S, Jeong Y, Chung HM, Kim CY. Development of heart organoid cryopreservation method through Fe 3 O 4 nanoparticles based nanowarming system. Biotechnol J 2024; 19:e2300311. [PMID: 37953523 DOI: 10.1002/biot.202300311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/25/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023]
Abstract
Beyond single cell two-dimensional (2D) culture, research on organoids that can mimic human organs is rapidly developing. However, there are still problems in commercialization and joint research using organoids due to the lack of technology to safely store organoids. Since organoids are 3D complex structures with a certain size (0.1-5 mm) beyond the size of cells, the conventional cell-level cryopreservation method using cryoprotectant (CPA) cannot overcome the damage caused by volume change due to osmotic pressure difference and ice nucleation. Herein, we attempted to solve such limitations by applying a nanowarming system using CPA with high cell permeability and Fe3 O4 nanoparticles. By performing beat rate measurement, histological analysis, contractility analysis, and multi-electrode array, it was verified that the developed method could significantly improve functional recovery and survival of heart organoids after freezing and thawing. In this study, we demonstrated a successful organoid cryopreservation method based on a Fe3 O4 nanowarming system. The developed technology will provide clues to the field of tissue cryopreservation and spur the application of organoids.
Collapse
Affiliation(s)
- Seul-Gi Lee
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Gwangjin-Gu, Seoul, Republic of Korea
| | - Jin Kim
- College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Jin Seok
- College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Min Woo Kim
- College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Jooeon Rhee
- College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Gyeong-Eun Song
- College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Shinhye Park
- College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Suemin Lee
- College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Youngin Jeong
- College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Hyung Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Gwangjin-Gu, Seoul, Republic of Korea
- Miraecell Bio Co. Ltd., Seoul, Republic of Korea
| | - C-Yoon Kim
- College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
48
|
Yanagida S, Kawagishi H, Kanda Y. [Cardiotoxicity risk assessment of anti-cancer drugs and future perspectives]. Nihon Yakurigaku Zasshi 2024; 159:83-89. [PMID: 38432924 DOI: 10.1254/fpj.23094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
Cardiotoxicity is a serious adverse effect of anti-cancer drugs. Anti-cancer drug-induced cardiotoxicity are arrhythmia, cardiac contractile dysfunction, coronary artery disease, and hypertension, which affect to the quality of life in patients with cancer. In particular, cardiac contractile dysfunction is a life-threatening symptom leading to heart failure, suggesting that it is very important to predict the risk of developing the contractile dysfunction by anti-cancer drugs. Recently, human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) can be used to assess the risk of drug-induced arrhythmias. This prompts us to evaluate other cardiotoxic effects such as contractility dysfunction and structural toxicity with hiPSC-CMs. Since anti-cancer drug-induced contractility dysfunction are considered to be induced by chronic exposure, we have developed a method to assess chronic contractility dysfunction by imaging analysis of hiPSC-CMs. BMS-986094, which failed in clinical trials due to the occurrence of heart failure, was used as a positive compound. We found that chronic exposure to BMS-986094 decreased the contraction and relaxation velocity in hiPSC-CMs. Doxorubicin was observed to decrease cytotoxicity and both contraction and relaxation velocities in hiPSC-CMs. We are currently further evaluating other anti-cancer drugs with different mode-of-actions using hiPSC-CMs and assess the predictivity and utility of contractile assessment using hiPSC-CMs by comparing with real-world data. Here, we introduce our novel method to assess the chronic contractility of hiPSC-CMs by imaging analysis and discuss the future perspectives for assessing the anti-cancer drug-induced cardiotoxicity.
Collapse
Affiliation(s)
- Shota Yanagida
- Division of Pharmacology, National Institute of Health Sciences
| | | | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences
| |
Collapse
|
49
|
Yanagida S, Kanda Y. [Prediction of Cardiac Toxicity by Anti-cancer Drugs Using iPSC Cardiomyocytes]. YAKUGAKU ZASSHI 2024; 144:265-271. [PMID: 38432935 DOI: 10.1248/yakushi.23-00164-3] [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] [Indexed: 03/05/2024]
Abstract
Recent advances in cancer therapy have significantly improved the survival rate of patients with cancer. In contrast, anti-cancer drug-induced adverse effects, especially cardiotoxicity, have come to affect patients' prognosis and quality of life. Therefore, there is a growing need to understand the anti-cancer drug-induced cardiotoxicity. Human induced pluripotent stem (iPS) cell-derived cardiomyocytes (hiPSC-CMs) have been used to assess drug-induced cardiotoxicity by improving the predictability of clinical cardiotoxicity and the principles of the 3Rs (replacement, reduction and refinement). To predict the anti-cancer drug-induced cardiotoxicity, we developed a novel method to assess drug-induced proarrhythmia risk using hiPSC-CMs by participating in the international validation. In addition, we established the chronic contractility toxicity assessment by image-based motion analysis. The compound BMS-986094, which was withdrawn from clinical trials, inhibited contractility velocity and relaxation velocity in hiPSC-CMs. Currently, we are trying to investigate the predictability of the contractility assay by comparing the hiPSC-CM data with adverse events reports from real-world database. In this review, we would like to introduce the novel imaging-based contractility method using hiPSC-CMs and future perspectives in anti-cancer drug-induced cardiotoxicity.
Collapse
Affiliation(s)
- Shota Yanagida
- Division of Pharmacology, National Institute of Health Sciences
- Graduate School of Biomedical and Health Sciences (Pharmaceutical Sciences), Hiroshima University
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences
| |
Collapse
|
50
|
Grandits T, Augustin CM, Haase G, Jost N, Mirams GR, Niederer SA, Plank G, Varró A, Virág L, Jung A. Neural network emulation of the human ventricular cardiomyocyte action potential: a tool for more efficient computation in pharmacological studies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.16.553497. [PMID: 38234850 PMCID: PMC10793461 DOI: 10.1101/2023.08.16.553497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Computer models of the human ventricular cardiomyocyte action potential (AP) have reached a level of detail and maturity that has led to an increasing number of applications in the pharmaceutical sector. However, interfacing the models with experimental data can become a significant computational burden. To mitigate the computational burden, the present study introduces a neural network (NN) that emulates the AP for given maximum conductances of selected ion channels, pumps, and exchangers. Its applicability in pharmacological studies was tested on synthetic and experimental data. The NN emulator potentially enables massive speed-ups compared to regular simulations and the forward problem (find drugged AP for pharmacological parameters defined as scaling factors of control maximum conductances) on synthetic data could be solved with average root-mean-square errors (RMSE) of 0.47mV in normal APs and of 14.5mV in abnormal APs exhibiting early afterdepolarizations (72.5% of the emulated APs were alining with the abnormality, and the substantial majority of the remaining APs demonstrated pronounced proximity). This demonstrates not only very fast and mostly very accurate AP emulations but also the capability of accounting for discontinuities, a major advantage over existing emulation strategies. Furthermore, the inverse problem (find pharmacological parameters for control and drugged APs through optimization) on synthetic data could be solved with high accuracy shown by a maximum RMSE of 0.21 in the estimated pharmacological parameters. However, notable mismatches were observed between pharmacological parameters estimated from experimental data and distributions obtained from the Comprehensive in vitro Proarrhythmia Assay initiative. This reveals larger inaccuracies which can be attributed particularly to the fact that small tissue preparations were studied while the emulator was trained on single cardiomyocyte data. Overall, our study highlights the potential of NN emulators as powerful tool for an increased efficiency in future quantitative systems pharmacology studies.
Collapse
Affiliation(s)
- Thomas Grandits
- Department of Mathematics and Scientific Computing, University of Graz
- NAWI Graz, University of Graz
| | - Christoph M Augustin
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging - Division of Medical Physics and Biophysics, Medical University of Graz
- BioTechMed-Graz
| | - Gundolf Haase
- Department of Mathematics and Scientific Computing, University of Graz
| | - Norbert Jost
- Department of Pharmacology and Pharmacotherapy, University of Szeged
- HUN-REN-TKI, Research Group of Pharmacology
| | - Gary R Mirams
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham
| | - Steven A Niederer
- Division of Imaging Sciences & Biomedical Engineering, King's College London
| | - Gernot Plank
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging - Division of Medical Physics and Biophysics, Medical University of Graz
- BioTechMed-Graz
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, University of Szeged
- HUN-REN-TKI, Research Group of Pharmacology
| | - László Virág
- Department of Pharmacology and Pharmacotherapy, University of Szeged
| | - Alexander Jung
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging - Division of Medical Physics and Biophysics, Medical University of Graz
| |
Collapse
|