1
|
Mir A, Zhu A, Lau R, Barr N, Sheikh Z, Acuna D, Dayal A, Hibino N. Applications, Limitations, and Considerations of Clinical Trials in a Dish. Bioengineering (Basel) 2024; 11:1096. [PMID: 39593756 PMCID: PMC11591410 DOI: 10.3390/bioengineering11111096] [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: 10/07/2024] [Revised: 10/26/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Recent advancements in biotechnology forged the path for clinical trials in dish (CTiDs) to advance as a popular method of experimentation in biomedicine. CTiDs play a fundamental role in translational research through technologies such as induced pluripotent stem cells, whole genome sequencing, and organs-on-a-chip. In this review, we explore advancements that enable these CTiD biotechnologies and their applications in animal testing, disease modeling, and space radiation technologies. Furthermore, this review dissects the advantages and disadvantages of CTiDs, as well as their regulatory considerations. Lastly, we evaluate the challenges that CTiDs pose and the role of CTiDs in future experimentation.
Collapse
Affiliation(s)
- Amatullah Mir
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (A.M.); (A.Z.); (R.L.); (N.B.); (Z.S.); (D.A.); (A.D.)
| | - Angie Zhu
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (A.M.); (A.Z.); (R.L.); (N.B.); (Z.S.); (D.A.); (A.D.)
| | - Rico Lau
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (A.M.); (A.Z.); (R.L.); (N.B.); (Z.S.); (D.A.); (A.D.)
| | - Nicolás Barr
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (A.M.); (A.Z.); (R.L.); (N.B.); (Z.S.); (D.A.); (A.D.)
| | - Zyva Sheikh
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (A.M.); (A.Z.); (R.L.); (N.B.); (Z.S.); (D.A.); (A.D.)
| | - Diana Acuna
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (A.M.); (A.Z.); (R.L.); (N.B.); (Z.S.); (D.A.); (A.D.)
| | - Anuhya Dayal
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (A.M.); (A.Z.); (R.L.); (N.B.); (Z.S.); (D.A.); (A.D.)
| | - Narutoshi Hibino
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (A.M.); (A.Z.); (R.L.); (N.B.); (Z.S.); (D.A.); (A.D.)
- Pediatric Cardiac Surgery, Advocate Children’s Hospital, 4440 W 95th St., Oak Lawn, IL 60453, USA
| |
Collapse
|
2
|
Zitzmann FD, Schmidt S, Frank R, Weigel W, Meier M, Jahnke HG. Microcavity well-plate for automated parallel bioelectronic analysis of 3D cell cultures. Biosens Bioelectron 2024; 250:116042. [PMID: 38266619 DOI: 10.1016/j.bios.2024.116042] [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: 10/31/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/26/2024]
Abstract
Three-dimensional (3D) in vitro cell culture models serve as valuable tools for accurately replicating cellular microenvironments found in vivo. While cell culture technologies are rapidly advancing, the availability of non-invasive, real-time, and label-free analysis methods for 3D cultures remains limited. To meet the demand for higher-throughput drug screening, there is a demanding need for analytical methods that can operate in parallel. Microelectrode systems in combination with microcavity arrays (MCAs), offer the capability of spatially resolved electrochemical impedance analysis and field potential monitoring of 3D cultures. However, the fabrication and handling of small-scale MCAs have been labour-intensive, limiting their broader application. To overcome this challenge, we have established a process for creating MCAs in a standard 96-well plate format using high-precision selective laser etching. In addition, to automate and ensure the accurate placement of 3D cultures on the MCA, we have designed and characterized a plug-in tool using SLA-3D-printing. To characterize our new 96-well plate MCA-based platform, we conducted parallel analyses of human melanoma 3D cultures and monitored the effect of cisplatin in real-time by impedance spectroscopy. In the following we demonstrate the capabilities of the MCA approach by analysing contraction rates of human pluripotent stem cell-derived cardiomyocyte aggregates in response to cardioactive compounds. In summary, our MCA system significantly expands the possibilities for label-free analysis of 3D cell and tissue cultures, offering an order of magnitude higher parallelization capacity than previous devices. This advancement greatly enhances its applicability in real-world settings, such as drug development or clinical diagnostics.
Collapse
Affiliation(s)
- Franziska D Zitzmann
- Centre for Biotechnology and Biomedicine, Biochemical Cell Technology, Leipzig University, Deutscher Platz 5, D-04103, Leipzig, Germany; b-ACT Matter, Research and Transfer Centre for bioactive Matter, Leipzig University, Deutscher Platz 5, D-04103, Leipzig, Germany
| | - Sabine Schmidt
- Centre for Biotechnology and Biomedicine, Biochemical Cell Technology, Leipzig University, Deutscher Platz 5, D-04103, Leipzig, Germany
| | - Ronny Frank
- Centre for Biotechnology and Biomedicine, Biochemical Cell Technology, Leipzig University, Deutscher Platz 5, D-04103, Leipzig, Germany
| | - Winnie Weigel
- Centre for Biotechnology and Biomedicine, Biochemical Cell Technology, Leipzig University, Deutscher Platz 5, D-04103, Leipzig, Germany
| | - Matthias Meier
- Centre for Biotechnology and Biomedicine, Biochemical Cell Technology, Leipzig University, Deutscher Platz 5, D-04103, Leipzig, Germany; Helmholtz Pioneer Campus, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Heinz-Georg Jahnke
- Centre for Biotechnology and Biomedicine, Biochemical Cell Technology, Leipzig University, Deutscher Platz 5, D-04103, Leipzig, Germany.
| |
Collapse
|
3
|
Deir S, Mozhdehbakhsh Mofrad Y, Mashayekhan S, Shamloo A, Mansoori-Kermani A. Step-by-step fabrication of heart-on-chip systems as models for cardiac disease modeling and drug screening. Talanta 2024; 266:124901. [PMID: 37459786 DOI: 10.1016/j.talanta.2023.124901] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/23/2023] [Accepted: 07/01/2023] [Indexed: 09/20/2023]
Abstract
Cardiovascular diseases are caused by hereditary factors, environmental conditions, and medication-related issues. On the other hand, the cardiotoxicity of drugs should be thoroughly examined before entering the market. In this regard, heart-on-chip (HOC) systems have been developed as a more efficient and cost-effective solution than traditional methods, such as 2D cell culture and animal models. HOCs must replicate the biology, physiology, and pathology of human heart tissue to be considered a reliable platform for heart disease modeling and drug testing. Therefore, many efforts have been made to find the best methods to fabricate different parts of HOCs and to improve the bio-mimicry of the systems in the last decade. Beating HOCs with different platforms have been developed and techniques, such as fabricating pumpless HOCs, have been used to make HOCs more user-friendly systems. Recent HOC platforms have the ability to simultaneously induce and record electrophysiological stimuli. Additionally, systems including both heart and cancer tissue have been developed to investigate tissue-tissue interactions' effect on cardiac tissue response to cancer drugs. In this review, all steps needed to be considered to fabricate a HOC were introduced, including the choice of cellular resources, biomaterials, fabrication techniques, biomarkers, and corresponding biosensors. Moreover, the current HOCs used for modeling cardiac diseases and testing the drugs are discussed. We finally introduced some suggestions for fabricating relatively more user-friendly HOCs and facilitating the commercialization process.
Collapse
Affiliation(s)
- Sara Deir
- School of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Yasaman Mozhdehbakhsh Mofrad
- Nano-Bioengineering Lab, School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran; Stem Cell and Regenerative Medicine Center, Sharif University of Technology, Tehran, Iran
| | - Shohreh Mashayekhan
- School of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran.
| | - Amir Shamloo
- Nano-Bioengineering Lab, School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran; Stem Cell and Regenerative Medicine Center, Sharif University of Technology, Tehran, Iran.
| | | |
Collapse
|
4
|
Finkel S, Sweet S, Locke T, Smith S, Wang Z, Sandini C, Imredy J, He Y, Durante M, Lagrutta A, Feinberg A, Lee A. FRESH™ 3D bioprinted cardiac tissue, a bioengineered platform for in vitro pharmacology. APL Bioeng 2023; 7:046113. [PMID: 38046544 PMCID: PMC10693443 DOI: 10.1063/5.0163363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 10/30/2023] [Indexed: 12/05/2023] Open
Abstract
There is critical need for a predictive model of human cardiac physiology in drug development to assess compound effects on human tissues. In vitro two-dimensional monolayer cultures of cardiomyocytes provide biochemical and cellular readouts, and in vivo animal models provide information on systemic cardiovascular response. However, there remains a significant gap in these models due to their incomplete recapitulation of adult human cardiovascular physiology. Recent efforts in developing in vitro models from engineered heart tissues have demonstrated potential for bridging this gap using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) in three-dimensional tissue structure. Here, we advance this paradigm by implementing FRESH™ 3D bioprinting to build human cardiac tissues in a medium throughput, well-plate format with controlled tissue architecture, tailored cellular composition, and native-like physiological function, specifically in its drug response. We combined hiPSC-CMs, endothelial cells, and fibroblasts in a cellular bioink and FRESH™ 3D bioprinted this mixture in the format of a thin tissue strip stabilized on a tissue fixture. We show that cardiac tissues could be fabricated directly in a 24-well plate format were composed of dense and highly aligned hiPSC-CMs at >600 million cells/mL and, within 14 days, demonstrated reproducible calcium transients and a fast conduction velocity of ∼16 cm/s. Interrogation of these cardiac tissues with the β-adrenergic receptor agonist isoproterenol showed responses consistent with positive chronotropy and inotropy. Treatment with calcium channel blocker verapamil demonstrated responses expected of hiPSC-CM derived cardiac tissues. These results confirm that FRESH™ 3D bioprinted cardiac tissues represent an in vitro platform that provides data on human physiological response.
Collapse
Affiliation(s)
| | | | - Tyler Locke
- FluidForm, Inc., Waltham, Massachusetts 02451, USA
| | - Sydney Smith
- FluidForm, Inc., Waltham, Massachusetts 02451, USA
| | - Zhefan Wang
- FluidForm, Inc., Waltham, Massachusetts 02451, USA
| | | | - John Imredy
- In Vitro Safety Pharmacology, Genetic and Cellular Toxicology, Merck & Co. Inc., Rahway, New Jersey 07065, USA
| | - Yufang He
- Division of Technology, Infrastructure, Operations and Experience, Merck & Co. Inc., Rahway, New Jersey 07065, USA
| | - Marc Durante
- Division of Technology, Infrastructure, Operations and Experience, Merck & Co. Inc., Rahway, New Jersey 07065, USA
| | - Armando Lagrutta
- In Vitro Safety Pharmacology, Genetic and Cellular Toxicology, Merck & Co. Inc., Rahway, New Jersey 07065, USA
| | | | - Andrew Lee
- FluidForm, Inc., Waltham, Massachusetts 02451, USA
| |
Collapse
|
5
|
Zhang X, Aggarwal P, Broeckel U, Abassi YA. Enhancing the functional maturity of hiPSC-derived cardiomyocytes to assess inotropic compounds. J Pharmacol Toxicol Methods 2023; 123:107282. [PMID: 37419294 DOI: 10.1016/j.vascn.2023.107282] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/19/2023] [Accepted: 07/04/2023] [Indexed: 07/09/2023]
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) present an attractive in vitro platform to model safety and toxicity assessments-notably screening pro-arrhythmic compounds. The utility of the platform is stymied by a hiPSC-CM contractile apparatus and calcium handling mechanism akin to fetal phenotypes, evidenced by a negative force-frequency relationship. As such, hiPSC-CMs are limited in their ability to assess compounds that modulate contraction mediated by ionotropic compounds (Robertson, Tran, & George, 2013). To address this limitation, we utilize Agilent's xCELLigence Real-Time Cell Analyzer ePacer (RTCA ePacer) to enhance hiPSC-CM functional maturity. A continuous, progressive increase of electrical pacing is applied to hiPSC-CMs for up to 15 days. Contraction and viability are recorded by measurement of impedance using the RTCA ePacer. Our data confirms hiPSC-CMs inherently demonstrate a negative impedance amplitude frequency that is reversed after long-term electrical pacing. The data also indicate positive inotropic compounds increase the contractility of paced cardiomyocytes and calcium handling machinery is improved. Increased expression of genes critical to cardiomyocyte maturation further underscores the maturity of paced cells. In summary, our data suggest the application of continuous electrical pacing can functionally mature hiPSC-CMs, enhancing cellular response to positive inotropic compounds and improving calcium handling. SUMMARY: Long-term electrical stimulation of hiPSC-CM leads to functional maturation enabling predictive assessment of inotropic compounds.
Collapse
|
6
|
Nguemfo Tchankugni A, Mbida M, Hescheler J, Nguemo F. Effect of Ethanolic Extract of Vernonia amygdalina on the Proliferation, Viability and Function of Mouse Induced Pluripotent Stem Cells and Cardiomyocytes. PLANTS (BASEL, SWITZERLAND) 2023; 12:1108. [PMID: 36903967 PMCID: PMC10005699 DOI: 10.3390/plants12051108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/03/2023] [Accepted: 02/15/2023] [Indexed: 06/18/2023]
Abstract
Vernonia amygdalina (V. amygdalina) leaves are commonly used in traditional medicine around the world for the treatment of a plethora disorders, including heart disease. The aim of this study was to examine and evaluate the cardiac effect of V. amygdalina leaf extracts using mouse induced pluripotent stem cells (miPSCs) and their cardiomyocytes' (CMs) derivatives. We used a well-established stem cell culture to assess the effect of V. amygdalina extract on miPSC proliferation, EB formation and the beating activity of miPS cell-derived CMs. To study the cytotoxic effect of our extract, undifferentiating miPSCs were exposed to different concentrations of V. amygdalina. Cell colony formation and EB morphology were assessed using microscopy, whereas the cell viability was accessed with an impedance-based method and immunocytochemistry following treatment with different concentrations of V. amygdalina. Ethanolic extract of V. amygdalina induced toxicity in miPSCs, as revealed by a decrease in cell proliferation and colony formation, and an increase in cell death at a concentration of ≥20 mg/mL. At a concentration of 10 mg/mL, the rate of beating EBs was observed with no significant difference regarding the yield of cardiac cells. In addition, V. amygdalina did not affect the sarcomeric organization, but induced positive or negative effects on miPS cell-derived CMs' differentiation in a concentration-dependent manner. Taken together, our findings demonstrate that the ethanolic extract of V. amygdalina affected cell proliferation, colony forming and cardiac beating capacities in a concentration-dependent manner.
Collapse
Affiliation(s)
- Arlette Nguemfo Tchankugni
- Research Unit of Biology and Applied Ecology, Department of Animal Biology, Faculty of Science, University of Dschang, Dschang P.O. Box 067, Cameroon
| | - Mpoame Mbida
- Research Unit of Biology and Applied Ecology, Department of Animal Biology, Faculty of Science, University of Dschang, Dschang P.O. Box 067, Cameroon
| | - Jürgen Hescheler
- Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Filomain Nguemo
- Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
7
|
Logun M, Colonna MB, Mueller KP, Ventarapragada D, Rodier R, Tondepu C, Piscopo NJ, Das A, Chvatal S, Hayes HB, Capitini CM, Brat DJ, Kotanchek T, Edison AS, Saha K, Karumbaiah L. Label-free in vitro assays predict the potency of anti-disialoganglioside chimeric antigen receptor T-cell products. Cytotherapy 2023; 25:670-682. [PMID: 36849306 PMCID: PMC10159906 DOI: 10.1016/j.jcyt.2023.01.008] [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: 10/28/2022] [Revised: 01/05/2023] [Accepted: 01/13/2023] [Indexed: 02/27/2023]
Abstract
BACKGROUND AIMS Chimeric antigen receptor (CAR) T cells have demonstrated remarkable efficacy against hematological malignancies; however, they have not experienced the same success against solid tumors such as glioblastoma (GBM). There is a growing need for high-throughput functional screening platforms to measure CAR T-cell potency against solid tumor cells. METHODS We used real-time, label-free cellular impedance sensing to evaluate the potency of anti-disialoganglioside (GD2) targeting CAR T-cell products against GD2+ patient-derived GBM stem cells over a period of 2 days and 7 days in vitro. We compared CAR T products using two different modes of gene transfer: retroviral transduction and virus-free CRISPR-editing. Endpoint flow cytometry, cytokine analysis and metabolomics data were acquired and integrated to create a predictive model of CAR T-cell potency. RESULTS Results indicated faster cytolysis by virus-free CRISPR-edited CAR T cells compared with retrovirally transduced CAR T cells, accompanied by increased inflammatory cytokine release, CD8+ CAR T-cell presence in co-culture conditions and CAR T-cell infiltration into three-dimensional GBM spheroids. Computational modeling identified increased tumor necrosis factor α concentrations with decreased glutamine, lactate and formate as being most predictive of short-term (2 days) and long-term (7 days) CAR T cell potency against GBM stem cells. CONCLUSIONS These studies establish impedance sensing as a high-throughput, label-free assay for preclinical potency testing of CAR T cells against solid tumors.
Collapse
Affiliation(s)
- Meghan Logun
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA; Division of Neuroscience, Biomedical and Health Sciences Institute, University of Georgia, Athens, Georgia, USA
| | - Maxwell B Colonna
- Department of Biochemistry & Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA; Institute of Bioinformatics, University of Georgia, Athens, Georgia, USA
| | - Katherine P Mueller
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin USA
| | | | - Riley Rodier
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA
| | - Chaitanya Tondepu
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA; Division of Neuroscience, Biomedical and Health Sciences Institute, University of Georgia, Athens, Georgia, USA; Edgar L. Rhodes Center for Animal and Dairy Science, College of Agriculture and Environmental Science, University of Georgia, Athens, Georgia, USA
| | - Nicole J Piscopo
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin USA
| | - Amritava Das
- Morgridge Institute for Research, Madison, Wisconsin, USA
| | | | | | - Christian M Capitini
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin USA; University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin USA
| | - Daniel J Brat
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois USA
| | | | - Arthur S Edison
- Department of Biochemistry & Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA; Institute of Bioinformatics, University of Georgia, Athens, Georgia, USA
| | - Krishanu Saha
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin USA; University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin USA
| | - Lohitash Karumbaiah
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA; Division of Neuroscience, Biomedical and Health Sciences Institute, University of Georgia, Athens, Georgia, USA; Edgar L. Rhodes Center for Animal and Dairy Science, College of Agriculture and Environmental Science, University of Georgia, Athens, Georgia, USA.
| |
Collapse
|
8
|
Iachetta G, Melle G, Colistra N, Tantussi F, De Angelis F, Dipalo M. Long-term in vitro recording of cardiac action potentials on microelectrode arrays for chronic cardiotoxicity assessment. Arch Toxicol 2023; 97:509-522. [PMID: 36607357 PMCID: PMC9859891 DOI: 10.1007/s00204-022-03422-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 11/15/2022] [Indexed: 01/07/2023]
Abstract
The reliable identification of chronic cardiotoxic effects in in vitro screenings is fundamental for filtering out toxic molecular entities before in vivo animal experimentation and clinical trials. Present techniques such as patch-clamp, voltage indicators, and standard microelectrode arrays do not offer at the same time high sensitivity for measuring transmembrane ion currents and low-invasiveness for monitoring cells over long time. Here, we show that optoporation applied to microelectrode arrays enables measuring action potentials from human-derived cardiac syncytia for more than 1 continuous month and provides reliable data on chronic cardiotoxic effects caused by known compounds such as pentamidine. The technique has high potential for detecting chronic cardiotoxicity in the early phases of drug development.
Collapse
Affiliation(s)
| | | | | | | | | | - Michele Dipalo
- Istituto Italiano di Tecnologia, Via Morego 30, 16163, Genova, Italy.
- FORESEE Biosystems Srl, Genova, Italy.
| |
Collapse
|
9
|
Pan Z, Liang P. Human-Induced Pluripotent Stem Cell-Based Differentiation of Cardiomyocyte Subtypes for Drug Discovery and Cell Therapy. Handb Exp Pharmacol 2023; 281:209-233. [PMID: 37421443 DOI: 10.1007/164_2023_663] [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: 07/10/2023]
Abstract
Drug attrition rates have increased over the past few years, accompanied with growing costs for the pharmaceutical industry and consumers. Lack of in vitro models connecting the results of toxicity screening assays with clinical outcomes accounts for this high attrition rate. The emergence of cardiomyocytes derived from human pluripotent stem cells provides an amenable source of cells for disease modeling, drug discovery, and cardiotoxicity screening. Functionally similar to to embryonic stem cells, but with fewer ethical concerns, induced pluripotent stem cells (iPSCs) can recapitulate patient-specific genetic backgrounds, which would be a huge revolution for personalized medicine. The generated iPSC-derived cardiomyocytes (iPSC-CMs) represent different subtypes including ventricular-, atrial-, and nodal-like cardiomyocytes. Purifying these subtypes for chamber-specific drug screening presents opportunities and challenges. In this chapter, we discuss the strategies for the purification of iPSC-CMs, the use of iPSC-CMs for drug discovery and cardiotoxicity test, and the current limitations of iPSC-CMs that should be overcome for wider and more precise cardiovascular applications.
Collapse
Affiliation(s)
- Ziwei Pan
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Ping Liang
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
10
|
Kanade PP, Oyunbaatar NE, Shanmugasundaram A, Jeong YJ, Kim ES, Lee BK, Lee DW. MEA-integrated cantilever platform for comparison of real-time change in electrophysiology and contractility of cardiomyocytes to drugs. Biosens Bioelectron 2022; 216:114675. [DOI: 10.1016/j.bios.2022.114675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 08/12/2022] [Accepted: 08/28/2022] [Indexed: 11/02/2022]
|
11
|
Ergir E, Oliver-De La Cruz J, Fernandes S, Cassani M, Niro F, Pereira-Sousa D, Vrbský J, Vinarský V, Perestrelo AR, Debellis D, Vadovičová N, Uldrijan S, Cavalieri F, Pagliari S, Redl H, Ertl P, Forte G. Generation and maturation of human iPSC-derived 3D organotypic cardiac microtissues in long-term culture. Sci Rep 2022; 12:17409. [PMID: 36257968 PMCID: PMC9579206 DOI: 10.1038/s41598-022-22225-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 10/11/2022] [Indexed: 01/12/2023] Open
Abstract
Cardiovascular diseases remain the leading cause of death worldwide; hence there is an increasing focus on developing physiologically relevant in vitro cardiovascular tissue models suitable for studying personalized medicine and pre-clinical tests. Despite recent advances, models that reproduce both tissue complexity and maturation are still limited. We have established a scaffold-free protocol to generate multicellular, beating human cardiac microtissues in vitro from hiPSCs-namely human organotypic cardiac microtissues (hOCMTs)-that show some degree of self-organization and can be cultured for long term. This is achieved by the differentiation of hiPSC in 2D monolayer culture towards cardiovascular lineage, followed by further aggregation on low-attachment culture dishes in 3D. The generated hOCMTs contain multiple cell types that physiologically compose the heart and beat without external stimuli for more than 100 days. We have shown that 3D hOCMTs display improved cardiac specification, survival and metabolic maturation as compared to standard monolayer cardiac differentiation. We also confirmed the functionality of hOCMTs by their response to cardioactive drugs in long-term culture. Furthermore, we demonstrated that they could be used to study chemotherapy-induced cardiotoxicity. Due to showing a tendency for self-organization, cellular heterogeneity, and functionality in our 3D microtissues over extended culture time, we could also confirm these constructs as human cardiac organoids (hCOs). This study could help to develop more physiologically-relevant cardiac tissue models, and represent a powerful platform for future translational research in cardiovascular biology.
Collapse
Affiliation(s)
- Ece Ergir
- grid.412752.70000 0004 0608 7557Center for Translational Medicine (CTM), International Clinical Research Centre (FNUSA-ICRC), St. Anne’s University Hospital, Studentská 812/6, 62500 Brno, Czech Republic ,grid.5329.d0000 0001 2348 4034Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, 1040 Vienna, Austria
| | - Jorge Oliver-De La Cruz
- grid.412752.70000 0004 0608 7557Center for Translational Medicine (CTM), International Clinical Research Centre (FNUSA-ICRC), St. Anne’s University Hospital, Studentská 812/6, 62500 Brno, Czech Republic
| | - Soraia Fernandes
- grid.412752.70000 0004 0608 7557Center for Translational Medicine (CTM), International Clinical Research Centre (FNUSA-ICRC), St. Anne’s University Hospital, Studentská 812/6, 62500 Brno, Czech Republic
| | - Marco Cassani
- grid.412752.70000 0004 0608 7557Center for Translational Medicine (CTM), International Clinical Research Centre (FNUSA-ICRC), St. Anne’s University Hospital, Studentská 812/6, 62500 Brno, Czech Republic
| | - Francesco Niro
- grid.412752.70000 0004 0608 7557Center for Translational Medicine (CTM), International Clinical Research Centre (FNUSA-ICRC), St. Anne’s University Hospital, Studentská 812/6, 62500 Brno, Czech Republic ,grid.10267.320000 0001 2194 0956Faculty of Medicine, Department of Biomedical Sciences, Masaryk University, 62500 Brno, Czech Republic
| | - Daniel Pereira-Sousa
- grid.412752.70000 0004 0608 7557Center for Translational Medicine (CTM), International Clinical Research Centre (FNUSA-ICRC), St. Anne’s University Hospital, Studentská 812/6, 62500 Brno, Czech Republic ,grid.10267.320000 0001 2194 0956Faculty of Medicine, Department of Biomedical Sciences, Masaryk University, 62500 Brno, Czech Republic
| | - Jan Vrbský
- grid.412752.70000 0004 0608 7557Center for Translational Medicine (CTM), International Clinical Research Centre (FNUSA-ICRC), St. Anne’s University Hospital, Studentská 812/6, 62500 Brno, Czech Republic
| | - Vladimír Vinarský
- grid.412752.70000 0004 0608 7557Center for Translational Medicine (CTM), International Clinical Research Centre (FNUSA-ICRC), St. Anne’s University Hospital, Studentská 812/6, 62500 Brno, Czech Republic
| | - Ana Rubina Perestrelo
- grid.412752.70000 0004 0608 7557Center for Translational Medicine (CTM), International Clinical Research Centre (FNUSA-ICRC), St. Anne’s University Hospital, Studentská 812/6, 62500 Brno, Czech Republic
| | - Doriana Debellis
- grid.25786.3e0000 0004 1764 2907Electron Microscopy Facility, Fondazione Istituto Italiano Di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Natália Vadovičová
- grid.10267.320000 0001 2194 0956Faculty of Medicine, Department of Biomedical Sciences, Masaryk University, 62500 Brno, Czech Republic
| | - Stjepan Uldrijan
- grid.10267.320000 0001 2194 0956Faculty of Medicine, Department of Biomedical Sciences, Masaryk University, 62500 Brno, Czech Republic
| | - Francesca Cavalieri
- grid.1008.90000 0001 2179 088XDepartment of Chemical Engineering, The University of Melbourne, Parkville, VIC 3010 Australia ,grid.6530.00000 0001 2300 0941Dipartimento di Scienze e Tecnologie Chimiche, Università degli Studi di Roma Tor Vergata, via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Stefania Pagliari
- grid.412752.70000 0004 0608 7557Center for Translational Medicine (CTM), International Clinical Research Centre (FNUSA-ICRC), St. Anne’s University Hospital, Studentská 812/6, 62500 Brno, Czech Republic
| | - Heinz Redl
- grid.454388.6Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, 1200 Vienna, Austria ,grid.511951.8Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Peter Ertl
- grid.5329.d0000 0001 2348 4034Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, 1040 Vienna, Austria ,grid.511951.8Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Giancarlo Forte
- grid.412752.70000 0004 0608 7557Center for Translational Medicine (CTM), International Clinical Research Centre (FNUSA-ICRC), St. Anne’s University Hospital, Studentská 812/6, 62500 Brno, Czech Republic ,grid.1374.10000 0001 2097 1371Department of Biomaterials Science, Institute of Dentistry, University of Turku, 20014 Turku, Finland
| |
Collapse
|
12
|
Snelders M, Koedijk IH, Schirmer J, Mulleners O, van Leeuwen J, de Wagenaar NP, Bartulos O, Voskamp P, Braam S, Guttenberg Z, Danser AJ, Majoor-Krakauer D, Meijering E, van der Pluijm I, Essers J. Contraction pressure analysis using optical imaging in normal and MYBPC3-mutated hiPSC-derived cardiomyocytes grown on matrices with tunable stiffness. BIOMATERIALS AND BIOSYSTEMS 2022; 8:100068. [PMID: 36824378 PMCID: PMC9934435 DOI: 10.1016/j.bbiosy.2022.100068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 09/09/2022] [Accepted: 10/15/2022] [Indexed: 12/04/2022] Open
Abstract
Current in vivo disease models and analysis methods for cardiac drug development have been insufficient in providing accurate and reliable predictions of drug efficacy and safety. Here, we propose a custom optical flow-based analysis method to quantitatively measure recordings of contracting cardiomyocytes on polydimethylsiloxane (PDMS), compatible with medium-throughput systems. Movement of the PDMS was examined by covalently bound fluorescent beads on the PDMS surface, differences caused by increased substrate stiffness were compared, and cells were stimulated with β-agonist. We further validated the system using cardiomyocytes treated with endothelin-1 and compared their contractions against control and cells incubated with receptor antagonist bosentan. After validation we examined two MYBPC3-mutant patient-derived cell lines. Recordings showed that higher substrate stiffness resulted in higher contractile pressure, while beating frequency remained similar to control. β-agonist stimulation resulted in both higher beating frequency as well as higher pressure values during contraction and relaxation. Cells treated with endothelin-1 showed an increased beating frequency, but a lower contraction pressure. Cells treated with both endothelin-1 and bosentan remained at control level of beating frequency and pressure. Lastly, both MYBPC3-mutant lines showed a higher beating frequency and lower contraction pressure. Our validated method is capable of automatically quantifying contraction of hiPSC-derived cardiomyocytes on a PDMS substrate of known shear modulus, returning an absolute value. Our method could have major benefits in a medium-throughput setting.
Collapse
Affiliation(s)
- Matthijs Snelders
- Department of Molecular Genetics, Erasmus MC, Rotterdam, the Netherlands
| | - Iris H. Koedijk
- Department of Molecular Genetics, Erasmus MC, Rotterdam, the Netherlands
| | | | - Otto Mulleners
- Department of Molecular Genetics, Erasmus MC, Rotterdam, the Netherlands
| | | | - Nathalie P. de Wagenaar
- Department of Molecular Genetics, Erasmus MC, Rotterdam, the Netherlands,Department of Cardiology, Erasmus MC, Rotterdam, the Netherlands
| | | | | | | | | | - A.H. Jan Danser
- Department of Internal Medicine - Pharmacology, Erasmus MC, Rotterdam, the Netherlands
| | | | - Erik Meijering
- School of Computer Science and Engineering, University of New South Wales, Sydney, Australia
| | - Ingrid van der Pluijm
- Department of Molecular Genetics, Erasmus MC, Rotterdam, the Netherlands,Department of Vascular Surgery, Erasmus MC, Rotterdam, the Netherlands
| | - Jeroen Essers
- Department of Molecular Genetics, Erasmus MC, Rotterdam, the Netherlands,Department of Vascular Surgery, Erasmus MC, Rotterdam, the Netherlands,Department of Radiotherapy, Erasmus MC, Rotterdam, the Netherlands,Corresponding author: Erasmus Medical Center, Wytemaweg 80, Rotterdam 3015CN, The Netherlands
| |
Collapse
|
13
|
Wei F, Pence L, Woodling K, Bagam P, Beger R, Gamboa da Costa G, Pang L. Effects of Serum and Compound Preparation Methods on Delayed Repolarization Evaluation With Human iPSC-CMs. Toxicol Sci 2022; 188:48-61. [PMID: 35478258 DOI: 10.1093/toxsci/kfac043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2024] Open
Abstract
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have been widely used in the Comprehensive in vitro Proarrhythmia Assay (CiPA). The notable difference of the electrophysiological (EP) responses of hiPSC-CMs in serum and serum-free media (SFM) is puzzling and may impact regulatory decision-making on the cardiac safety of candidate drugs in inducing QT prolongation and torsade de pointes (TdP). In this study, we compared the EP responses of hiPSC-CMs to 10 CiPA compounds and moxifloxacin in serum and SFM; explained the potential reason behind the different EP responses—abiotic compound loss to plastic tubes/plates of hydrophobic compounds prepared in SFM; and investigated the impact of compound preparation methods on drug bioavailability in exposure media, which affects the TdP risk prediction of drugs tested in serum-containing and SFM. For assays to be conducted in SFM, awareness of abiotic compound loss of hydrophobic compounds in serum-free preparations is critical for delay repolarization evaluation and data extrapolation from in vitro to in vivo.
Collapse
Affiliation(s)
- Feng Wei
- Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson, Arkansas 72079, USA
- Department of Structural Heart Disease, the First Affiliated Hospital of Xi’an Jiaotong University , Xi’an, Shaanxi 710061, China
| | - Lisa Pence
- Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson, Arkansas 72079, USA
| | - Kellie Woodling
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson, Arkansas 72079, USA
| | - Prathyusha Bagam
- Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson, Arkansas 72079, USA
| | - Richard Beger
- Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson, Arkansas 72079, USA
| | - Goncalo Gamboa da Costa
- Office of the Director, National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson, Arkansas 72079, USA
| | - Li Pang
- Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson, Arkansas 72079, USA
| |
Collapse
|
14
|
Li M, Gong J, Gao L, Zou T, Kang J, Xu H. Advanced human developmental toxicity and teratogenicity assessment using human organoid models. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 235:113429. [PMID: 35325609 DOI: 10.1016/j.ecoenv.2022.113429] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/12/2022] [Accepted: 03/16/2022] [Indexed: 06/14/2023]
Abstract
Tremendous progress has been made in the field of toxicology leading to the advance of developmental toxicity assessment. Conventional animal models and in vitro two-dimensional models cannot accurately describe toxic effects and predict actual in vivo responses due to obvious inter-species differences between humans and animals, as well as the lack of a physiologically relevant tissue microenvironment. Human embryonic stem cell (hESC)- and induced pluripotent stem cell (iPSC)-derived three-dimensional organoids are ideal complex and multicellular organotypic models, which are indispensable in recapitulating morphogenesis, cellular interactions, and molecular processes of early human organ development. Recently, human organoids have been used for drug discovery, chemical toxicity and safety in vitro assessment. This review discusses the recent advances in the use of human organoid models, (i.e., brain, retinal, cardiac, liver, kidney, lung, and intestinal organoid models) for developmental toxicity and teratogenicity assessment of distinct tissues/organs following exposure to pharmaceutical compounds, heavy metals, persistent organic pollutants, nanomaterials, and ambient air pollutants. Combining next-generation organoid models with innovative engineering technologies generates novel and powerful tools for developmental toxicity and teratogenicity assessment, and the rapid progress in this field is expected to continue.
Collapse
Affiliation(s)
- Minghui Li
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Jing Gong
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Lixiong Gao
- Department of Ophthalmology, Third Medical Center of PLA General Hospital, Beijing 100039, China
| | - Ting Zou
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Jiahui Kang
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China.
| |
Collapse
|
15
|
Human Induced Pluripotent Stem Cell as a Disease Modeling and Drug Development Platform-A Cardiac Perspective. Cells 2021; 10:cells10123483. [PMID: 34943991 PMCID: PMC8699880 DOI: 10.3390/cells10123483] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 02/07/2023] Open
Abstract
A comprehensive understanding of the pathophysiology and cellular responses to drugs in human heart disease is limited by species differences between humans and experimental animals. In addition, isolation of human cardiomyocytes (CMs) is complicated because cells obtained by biopsy do not proliferate to provide sufficient numbers of cells for preclinical studies in vitro. Interestingly, the discovery of human-induced pluripotent stem cell (hiPSC) has opened up the possibility of generating and studying heart disease in a culture dish. The combination of reprogramming and genome editing technologies to generate a broad spectrum of human heart diseases in vitro offers a great opportunity to elucidate gene function and mechanisms. However, to exploit the potential applications of hiPSC-derived-CMs for drug testing and studying adult-onset cardiac disease, a full functional characterization of maturation and metabolic traits is required. In this review, we focus on methods to reprogram somatic cells into hiPSC and the solutions for overcome immaturity of the hiPSC-derived-CMs to mimic the structure and physiological properties of the adult human CMs to accurately model disease and test drug safety. Finally, we discuss how to improve the culture, differentiation, and purification of CMs to obtain sufficient numbers of desired types of hiPSC-derived-CMs for disease modeling and drug development platform.
Collapse
|
16
|
Long Y, Hou J, Tang F, Lin Z, Huang X, Li W, Chen Y, Li Z, Wu Z. Proarrhythmic effects induced by benzethonium chloride and domiphen bromide in vitro and in vivo. Toxicol Appl Pharmacol 2021; 431:115731. [PMID: 34592322 DOI: 10.1016/j.taap.2021.115731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/13/2021] [Accepted: 09/23/2021] [Indexed: 12/31/2022]
Abstract
Benzethonium chloride (BZT) and domiphen bromide (DMP) are widely used as antimicrobials in drugs, vaccines and industry. However, no cardiac safety data has been developed on both compounds. Previously we reported BZT and DMP as high-affinity human ether-a-go-go related gene (HERG) channel inhibitors with unknown proarrhythmic risk. Here, we investigate the cardiotoxicity of BZT and DMP in vitro and in vivo, aiming to improve the safety-in-use of both antimicrobials. In the present study, human iPSC derived cardiomyocytes (hiPSC-CMs) were generated and rabbit models were used to examine the proarrhythmic potential of BZT and DMP. Our results found that BZT and DMP induced time- and dose-dependent decrease in the contractile parameters of hiPSC-CMs, prolonged FPDc (≥ 0.1 μM), caused tachycardia/fibrillation-like oscillation (0.3-1 μM), ultimately progressing to irreversible arrest of beating (≥ 1 μM). The IC50 values of BZT and DMP derived from normalized beat rate were 0.13 μM and 0.10 μM on hiPSC-CMs at 76 days. Moreover, in vivo rabbit ECG data demonstrated that 12.85 mg/kg BZT and 3.85 mg/kg DMP evoked QTc prolongation, noncomplex arrhythmias and ventricular tachycardias. Our findings support the cardiac safety of 0.01 μM BZT/DMP in vitro and the intravenous infusion of 3.85 mg/kg BZT and 1.28 mg/kg DMP in vivo, whereas higher concentrations of both compounds cause mild to moderate cardiotoxicity that should not be neglected during medical and industrial applications.
Collapse
Affiliation(s)
- Yan Long
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; Central Laboratory, Shenzhen Samii Medical Center, Shenzhen, China
| | - Jian Hou
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Feng Tang
- Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zuoxian Lin
- Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xiaolin Huang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Wei Li
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yili Chen
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhiyuan Li
- Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.
| | - Zhongkai Wu
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
17
|
Blebbistatin protects iPSC-CMs from hypercontraction and facilitates automated patch-clamp based electrophysiological study. Stem Cell Res 2021; 56:102565. [PMID: 34638057 DOI: 10.1016/j.scr.2021.102565] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/01/2021] [Accepted: 10/06/2021] [Indexed: 01/11/2023] Open
Abstract
Recently, there have been great advances in cardiovascular channelopathy modeling and drug safety pharmacology using human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). The automated patch-clamp (APC) technique overcomes the disadvantages of the manual patch-clamp (MPC) technique, which is labor intensive and gives low output. However, the application of the APC platform is still limited in iPSC-CM based research, due to the difficulty in maintaining the high quality of single iPSC-CMs during dissociation and recording. In this study, we improved the method for single iPSC-CM preparation by applying 2.5 µM blebbistatin (BB, an excitation-contraction coupling uncoupler) throughout APC procedures (dissociation, filtration, storage, and recording). Under non-BB buffered condition, iPSC-CMs in suspension showed a severe bleb-like morphology. However, BB-supplement led to significant improvements in morphology and INa recording, and we even obtained several CMs that showed spontaneous action potentials with typical morphology. Furthermore, APC faithfully recapitulated the single-cell electrophysiological phenotypes of iPSC-CMs derived from Brugada syndrome patients, as detected with MPC. Our study indicates that APC is capable of replacing MPC in the modeling of cardiac channelopathies using human iPSC-CMs by providing high-quality data with higher throughput.
Collapse
|
18
|
Ohya T, Ohtomo H, Kikuchi T, Sasaki D, Kawamura Y, Matsuura K, Shimizu T, Fukuda K, Someya T, Umezu S. Simultaneous measurement of contractile force and field potential of dynamically beating human iPS cell-derived cardiac cell sheet-tissue with flexible electronics. LAB ON A CHIP 2021; 21:3899-3909. [PMID: 34636821 DOI: 10.1039/d1lc00411e] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Human induced pluripotent stem (iPS) cell-derived cardiomyocytes are used for in vitro pharmacological and pathological studies worldwide. In particular, the functional assessment of cardiac tissues created from iPS cell-derived cardiomyocytes is expected to provide precise prediction of drug effects and thus streamline the process of drug development. However, the current format of electrophysiological and contractile assessment of cardiomyocytes on a rigid substrate is not appropriate for cardiac tissues that beat dynamically. Here, we show a novel simultaneous measurement system for contractile force and extracellular field potential of iPS cell-derived cardiac cell sheet-tissues using 500 nm-thick flexible electronic sheets. It was confirmed that the developed system is applicable for pharmacological studies and assessments of excitation-contraction coupling-related parameters, such as the electro-mechanical window. Our results indicate that flexible electronics with cardiac tissue engineering provide an advanced platform for drug development. This system will contribute to gaining new insight in pharmacological study of human cardiac function.
Collapse
Affiliation(s)
- Takashi Ohya
- Department of Modern Mechanical Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan.
- Thin-Film Device Laboratory & Center for Emergent Matter Science, RIKEN, 2-1, Hirosawa, Wako, Saitama 351-0198, Japan.
| | - Haruki Ohtomo
- Department of Modern Mechanical Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan.
| | - Tetsutaro Kikuchi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-Ku, Tokyo 162-8666, Japan
| | - Daisuke Sasaki
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-Ku, Tokyo 162-8666, Japan
| | - Yohei Kawamura
- Thin-Film Device Laboratory & Center for Emergent Matter Science, RIKEN, 2-1, Hirosawa, Wako, Saitama 351-0198, Japan.
- Department of Integrative Bioscience and Biomedical Engineering, TWIns, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Katsuhisa Matsuura
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-Ku, Tokyo 162-8666, Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-Ku, Tokyo 162-8666, Japan
| | - Kenjiro Fukuda
- Thin-Film Device Laboratory & Center for Emergent Matter Science, RIKEN, 2-1, Hirosawa, Wako, Saitama 351-0198, Japan.
| | - Takao Someya
- Thin-Film Device Laboratory & Center for Emergent Matter Science, RIKEN, 2-1, Hirosawa, Wako, Saitama 351-0198, Japan.
- Electrical and Electronic Engineering and Information Systems, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Shinjiro Umezu
- Department of Modern Mechanical Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan.
| |
Collapse
|
19
|
Grafton F, Ho J, Ranjbarvaziri S, Farshidfar F, Budan A, Steltzer S, Maddah M, Loewke KE, Green K, Patel S, Hoey T, Mandegar MA. Deep learning detects cardiotoxicity in a high-content screen with induced pluripotent stem cell-derived cardiomyocytes. eLife 2021; 10:68714. [PMID: 34338636 PMCID: PMC8367386 DOI: 10.7554/elife.68714] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/27/2021] [Indexed: 12/15/2022] Open
Abstract
Drug-induced cardiotoxicity and hepatotoxicity are major causes of drug attrition. To decrease late-stage drug attrition, pharmaceutical and biotechnology industries need to establish biologically relevant models that use phenotypic screening to detect drug-induced toxicity in vitro. In this study, we sought to rapidly detect patterns of cardiotoxicity using high-content image analysis with deep learning and induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). We screened a library of 1280 bioactive compounds and identified those with potential cardiotoxic liabilities in iPSC-CMs using a single-parameter score based on deep learning. Compounds demonstrating cardiotoxicity in iPSC-CMs included DNA intercalators, ion channel blockers, epidermal growth factor receptor, cyclin-dependent kinase, and multi-kinase inhibitors. We also screened a diverse library of molecules with unknown targets and identified chemical frameworks that show cardiotoxic signal in iPSC-CMs. By using this screening approach during target discovery and lead optimization, we can de-risk early-stage drug discovery. We show that the broad applicability of combining deep learning with iPSC technology is an effective way to interrogate cellular phenotypes and identify drugs that may protect against diseased phenotypes and deleterious mutations.
Collapse
Affiliation(s)
| | - Jaclyn Ho
- Tenaya Therapeutics, South San Francisco, United States
| | - Sara Ranjbarvaziri
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, United States
| | | | | | | | | | | | | | - Snahel Patel
- Tenaya Therapeutics, South San Francisco, United States
| | - Tim Hoey
- Tenaya Therapeutics, South San Francisco, United States
| | | |
Collapse
|
20
|
Comparison of 10 Control hPSC Lines for Drug Screening in an Engineered Heart Tissue Format. Stem Cell Reports 2021; 15:983-998. [PMID: 33053362 PMCID: PMC7561618 DOI: 10.1016/j.stemcr.2020.09.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 12/19/2022] Open
Abstract
Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) are commercially available, and cardiac differentiation established routine. Systematic evaluation of several control hiPSC-CM is lacking. We investigated 10 different control hiPSC-CM lines and analyzed function and suitability for drug screening. Five commercial and 5 academic hPSC-CM lines were casted in engineered heart tissue (EHT) format. Spontaneous and stimulated EHT contractions were analyzed, and 7 inotropic indicator compounds investigated on 8 cell lines. Baseline contractile force, kinetics, and rate varied widely among the different lines (e.g., relaxation time range: 118-471 ms). In contrast, the qualitative correctness of responses to BayK-8644, nifedipine, EMD-57033, isoprenaline, and digoxin in terms of force and kinetics varied only between 80% and 93%. Large baseline differences between control cell lines support the request for isogenic controls in disease modeling. Variability appears less relevant for drug screening but needs to be considered, arguing for studies with more than one line.
Collapse
|
21
|
Meek AT, Kronenberg NM, Morton A, Liehm P, Murawski J, Dalaka E, Booth JH, Powis SJ, Gather MC. Real-time imaging of cellular forces using optical interference. Nat Commun 2021; 12:3552. [PMID: 34117241 PMCID: PMC8196160 DOI: 10.1038/s41467-021-23734-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 04/30/2021] [Indexed: 12/01/2022] Open
Abstract
Important dynamic processes in mechanobiology remain elusive due to a lack of tools to image the small cellular forces at play with sufficient speed and throughput. Here, we introduce a fast, interference-based force imaging method that uses the illumination of an elastic deformable microcavity with two rapidly alternating wavelengths to map forces. We show real-time acquisition and processing of data, obtain images of mechanical activity while scanning across a cell culture, and investigate sub-second fluctuations of the piconewton forces exerted by macrophage podosomes. We also demonstrate force imaging of beating neonatal cardiomyocytes at 100 fps which reveals mechanical aspects of spontaneous oscillatory contraction waves in between the main contraction cycles. These examples illustrate the wider potential of our technique for monitoring cellular forces with high throughput and excellent temporal resolution. Studying dynamic processes in mechanobiology has been challenging due to lack of appropriate tools. Here, the authors present an interference-based method, illuminated via two rapidly alternating wavelengths, which enables real-time mapping of nanoscale forces with sub-second mechanical fluctuations.
Collapse
Affiliation(s)
- Andrew T Meek
- SUPA, School of Physics and Astronomy, University of St Andrews, North Haugh, St Andrews, UK
| | - Nils M Kronenberg
- SUPA, School of Physics and Astronomy, University of St Andrews, North Haugh, St Andrews, UK.,Humboldt Centre for Nano- and Biophotonics, Department of Chemistry, University of Cologne, Cologne, Germany
| | - Andrew Morton
- SUPA, School of Physics and Astronomy, University of St Andrews, North Haugh, St Andrews, UK
| | - Philipp Liehm
- SUPA, School of Physics and Astronomy, University of St Andrews, North Haugh, St Andrews, UK
| | - Jan Murawski
- SUPA, School of Physics and Astronomy, University of St Andrews, North Haugh, St Andrews, UK
| | - Eleni Dalaka
- SUPA, School of Physics and Astronomy, University of St Andrews, North Haugh, St Andrews, UK
| | - Jonathan H Booth
- SUPA, School of Physics and Astronomy, University of St Andrews, North Haugh, St Andrews, UK
| | - Simon J Powis
- School of Medicine, University of St Andrews, North Haugh, St Andrews, UK.,Centre of Biophotonics, University of St Andrews, North Haugh, St Andrews, UK
| | - Malte C Gather
- SUPA, School of Physics and Astronomy, University of St Andrews, North Haugh, St Andrews, UK. .,Humboldt Centre for Nano- and Biophotonics, Department of Chemistry, University of Cologne, Cologne, Germany. .,Centre of Biophotonics, University of St Andrews, North Haugh, St Andrews, UK.
| |
Collapse
|
22
|
Palmer JA, Smith AM, Gryshkova V, Donley ELR, Valentin JP, Burrier RE. A Targeted Metabolomics-Based Assay Using Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes Identifies Structural and Functional Cardiotoxicity Potential. Toxicol Sci 2021; 174:218-240. [PMID: 32040181 DOI: 10.1093/toxsci/kfaa015] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Implementing screening assays that identify functional and structural cardiotoxicity earlier in the drug development pipeline has the potential to improve safety and decrease the cost and time required to bring new drugs to market. In this study, a metabolic biomarker-based assay was developed that predicts the cardiotoxicity potential of a drug based on changes in the metabolism and viability of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM). Assay development and testing was conducted in 2 phases: (1) biomarker identification and (2) targeted assay development. In the first phase, metabolomic data from hiPSC-CM spent media following exposure to 66 drugs were used to identify biomarkers that identified both functional and structural cardiotoxicants. Four metabolites that represent different metabolic pathways (arachidonic acid, lactic acid, 2'-deoxycytidine, and thymidine) were identified as indicators of cardiotoxicity. In phase 2, a targeted, exposure-based biomarker assay was developed that measured these metabolites and hiPSC-CM viability across an 8-point concentration curve. Metabolite-specific predictive thresholds for identifying the cardiotoxicity potential of a drug were established and optimized for balanced accuracy or sensitivity. When predictive thresholds were optimized for balanced accuracy, the assay predicted the cardiotoxicity potential of 81 drugs with 86% balanced accuracy, 83% sensitivity, and 90% specificity. Alternatively, optimizing the thresholds for sensitivity yields a balanced accuracy of 85%, 90% sensitivity, and 79% specificity. This new hiPSC-CM-based assay provides a paradigm that can identify structural and functional cardiotoxic drugs that could be used in conjunction with other endpoints to provide a more comprehensive evaluation of a drug's cardiotoxicity potential.
Collapse
Affiliation(s)
| | - Alan M Smith
- Stemina Biomarker Discovery, Inc, Madison, Wisconsin
| | - Vitalina Gryshkova
- UCB Biopharma SPRL, Investigative Toxicology, Development Science, B-1420 Braine L'Alleud, Belgium
| | | | - Jean-Pierre Valentin
- UCB Biopharma SPRL, Investigative Toxicology, Development Science, B-1420 Braine L'Alleud, Belgium
| | | |
Collapse
|
23
|
Lee-Montiel FT, Laemmle A, Charwat V, Dumont L, Lee CS, Huebsch N, Okochi H, Hancock MJ, Siemons B, Boggess SC, Goswami I, Miller EW, Willenbring H, Healy KE. Integrated Isogenic Human Induced Pluripotent Stem Cell-Based Liver and Heart Microphysiological Systems Predict Unsafe Drug-Drug Interaction. Front Pharmacol 2021; 12:667010. [PMID: 34025426 PMCID: PMC8138446 DOI: 10.3389/fphar.2021.667010] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 04/14/2021] [Indexed: 12/14/2022] Open
Abstract
Three-dimensional (3D) microphysiological systems (MPSs) mimicking human organ function in vitro are an emerging alternative to conventional monolayer cell culture and animal models for drug development. Human induced pluripotent stem cells (hiPSCs) have the potential to capture the diversity of human genetics and provide an unlimited supply of cells. Combining hiPSCs with microfluidics technology in MPSs offers new perspectives for drug development. Here, the integration of a newly developed liver MPS with a cardiac MPS—both created with the same hiPSC line—to study drug–drug interaction (DDI) is reported. As a prominent example of clinically relevant DDI, the interaction of the arrhythmogenic gastroprokinetic cisapride with the fungicide ketoconazole was investigated. As seen in patients, metabolic conversion of cisapride to non-arrhythmogenic norcisapride in the liver MPS by the cytochrome P450 enzyme CYP3A4 was inhibited by ketoconazole, leading to arrhythmia in the cardiac MPS. These results establish integration of hiPSC-based liver and cardiac MPSs to facilitate screening for DDI, and thus drug efficacy and toxicity, isogenic in the same genetic background.
Collapse
Affiliation(s)
- Felipe T Lee-Montiel
- Departments of Bioengineering, and Materials Science & Engineering, University of California Berkeley, Berkeley, CA, United States
| | - Alexander Laemmle
- Department of Surgery, Division of Transplant Surgery, Liver Center and Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, United States.,Institute of Clinical Chemistry and Department of Pediatrics, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Verena Charwat
- Departments of Bioengineering, and Materials Science & Engineering, University of California Berkeley, Berkeley, CA, United States
| | - Laure Dumont
- Department of Surgery, Division of Transplant Surgery, Liver Center and Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, United States
| | - Caleb S Lee
- Departments of Bioengineering, and Materials Science & Engineering, University of California Berkeley, Berkeley, CA, United States
| | - Nathaniel Huebsch
- Departments of Bioengineering, and Materials Science & Engineering, University of California Berkeley, Berkeley, CA, United States
| | - Hideaki Okochi
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA, United States
| | | | - Brian Siemons
- Departments of Bioengineering, and Materials Science & Engineering, University of California Berkeley, Berkeley, CA, United States
| | - Steven C Boggess
- Department of Chemistry, University of California Berkeley, Berkeley, CA, United States
| | - Ishan Goswami
- Departments of Bioengineering, and Materials Science & Engineering, University of California Berkeley, Berkeley, CA, United States
| | - Evan W Miller
- Departments of Chemistry and Molecular & Cell Biology, and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, United States
| | - Holger Willenbring
- Department of Surgery, Division of Transplant Surgery, Liver Center and Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, United States
| | - Kevin E Healy
- Departments of Bioengineering, and Materials Science & Engineering, University of California Berkeley, Berkeley, CA, United States
| |
Collapse
|
24
|
Ahmed U, Ahmed R, Masoud MS, Tariq M, Ashfaq UA, Augustine R, Hasan A. Stem cells based in vitro models: trends and prospects in biomaterials cytotoxicity studies. Biomed Mater 2021; 16:042003. [PMID: 33686970 DOI: 10.1088/1748-605x/abe6d8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Advanced biomaterials are increasingly used for numerous medical applications from the delivery of cancer-targeted therapeutics to the treatment of cardiovascular diseases. The issues of foreign body reactions induced by biomaterials must be controlled for preventing treatment failure. Therefore, it is important to assess the biocompatibility and cytotoxicity of biomaterials on cell culture systems before proceeding to in vivo studies in animal models and subsequent clinical trials. Direct use of biomaterials on animals create technical challenges and ethical issues and therefore, the use of non-animal models such as stem cell cultures could be useful for determination of their safety. However, failure to recapitulate the complex in vivo microenvironment have largely restricted stem cell cultures for testing the cytotoxicity of biomaterials. Nevertheless, properties of stem cells such as their self-renewal and ability to differentiate into various cell lineages make them an ideal candidate for in vitro screening studies. Furthermore, the application of stem cells in biomaterials screening studies may overcome the challenges associated with the inability to develop a complex heterogeneous tissue using primary cells. Currently, embryonic stem cells, adult stem cells, and induced pluripotent stem cells are being used as in vitro preliminary biomaterials testing models with demonstrated advantages over mature primary cell or cell line based in vitro models. This review discusses the status and future directions of in vitro stem cell-based cultures and their derivatives such as spheroids and organoids for the screening of their safety before their application to animal models and human in translational research.
Collapse
Affiliation(s)
- Uzair Ahmed
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad 38000 Punjab, Pakistan
| | | | | | | | | | | | | |
Collapse
|
25
|
Burnett SD, Blanchette AD, Chiu WA, Rusyn I. Human induced pluripotent stem cell (iPSC)-derived cardiomyocytes as an in vitro model in toxicology: strengths and weaknesses for hazard identification and risk characterization. Expert Opin Drug Metab Toxicol 2021; 17:887-902. [PMID: 33612039 DOI: 10.1080/17425255.2021.1894122] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Human induced pluripotent stem cell (iPSC)-derived cardiomyocytes is one of the most widely used cell-based models that resulted from the discovery of how non-embryonic stem cells can be differentiated into multiple cell types. In just one decade, iPSC-derived cardiomyocytes went from a research lab to widespread use in biomedical research and preclinical safety evaluation for drugs and other chemicals. AREAS COVERED This manuscript reviews data on toxicology applications of human iPSC-derived cardiomyocytes. We detail the outcome of a systematic literature search on their use (i) in hazard assessment for cardiotoxicity liabilities, (ii) for risk characterization, (iii) as models for population variability, and (iv) in studies of personalized medicine and disease. EXPERT OPINION iPSC-derived cardiomyocytes are useful to increase the accuracy, precision, and efficiency of cardiotoxicity hazard identification for both drugs and non-pharmaceuticals, with recent efforts beginning to demonstrate their utility for risk characterization. Notable limitations include the needs to improve the maturation of cells in culture, to better understand their potential use identifying structural cardiotoxicity, and for additional case studies involving population-wide and disease-specific risk characterization. Ultimately, the greatest future benefits are likely for non-pharmaceutical chemicals, filling a critical gap where no routine testing for cardiotoxicity is currently performed.
Collapse
Affiliation(s)
- Sarah D Burnett
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Alexander D Blanchette
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Weihsueh A Chiu
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| |
Collapse
|
26
|
Iachetta G, Colistra N, Melle G, Deleye L, Tantussi F, De Angelis F, Dipalo M. Improving reliability and reducing costs of cardiotoxicity assessments using laser-induced cell poration on microelectrode arrays. Toxicol Appl Pharmacol 2021; 418:115480. [PMID: 33689843 DOI: 10.1016/j.taap.2021.115480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/07/2021] [Accepted: 03/02/2021] [Indexed: 10/22/2022]
Abstract
Drug-induced cardiotoxicity is a major barrier to drug development and a main cause of withdrawal of marketed drugs. Drugs can strongly alter the spontaneous functioning of the heart by interacting with the cardiac membrane ion channels. If these effects only surface during in vivo preclinical tests, clinical trials or worse after commercialization, the societal and economic burden will be significant and seriously hinder the efficient drug development process. Hence, cardiac safety pharmacology requires in vitro electrophysiological screening assays of all drug candidates to predict cardiotoxic effects before clinical trials. In the past 10 years, microelectrode array (MEA) technology began to be considered a valuable approach in pharmaceutical applications. However, an effective tool for high-throughput intracellular measurements, compatible with pharmaceutical standards, is not yet available. Here, we propose laser-induced optoacoustic poration combined with CMOS-MEA technology as a reliable and effective platform to detect cardiotoxicity. This approach enables the acquisition of high-quality action potential recordings from large numbers of cardiomyocytes within the same culture well, providing reliable data using single-well MEA devices and single cardiac syncytia per each drug. Thus, this technology could be applied in drug safety screening platforms reducing times and costs of cardiotoxicity assessments, while simultaneously improving the data reliability.
Collapse
Affiliation(s)
| | - Nicolò Colistra
- Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Giovanni Melle
- Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Lieselot Deleye
- Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | | | | | - Michele Dipalo
- Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy.
| |
Collapse
|
27
|
Luo YS, Chen Z, Blanchette AD, Zhou YH, Wright FA, Baker ES, Chiu WA, Rusyn I. Relationships between constituents of energy drinks and beating parameters in human induced pluripotent stem cell (iPSC)-Derived cardiomyocytes. Food Chem Toxicol 2021; 149:111979. [PMID: 33450301 DOI: 10.1016/j.fct.2021.111979] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/24/2022]
Abstract
Consumption of energy drinks has been associated with adverse cardiovascular effects; however, little is known about the ingredients that may contribute to these effects. We therefore characterized the chemical profiles and in vitro effects of energy drinks and their ingredients on human induced pluripotent stem cell (iPSC)-derived cardiomyocytes, and identified the putative active ingredients using a multivariate prediction model. Energy drinks from 17 widely-available over-the-counter brands were evaluated in this study. The concentrations of six common ingredients (caffeine, taurine, riboflavin, pantothenic acid, adenine, and L-methionine) were quantified by coupling liquid chromatography with a triple quadrupole mass spectrometer for the acquisition of LC-MS/MS spectra. In addition, untargeted analyses for each beverage were performed with a platform combining LC, ion mobility spectrometry and mass spectrometry (LC-IMS-MS) measurements. Approximately 300 features were observed across samples in the untargeted studies, and of these ~100 were identified. In vitro effects of energy drinks and some of their ingredients were then tested in iPSC-derived cardiomyocytes. Data on the beat rate (positive and negative chronotropy), ion channel function (QT prolongation), and cytotoxicity were collected in a dilution series. We found that some of the energy drinks elicited adverse effects on the cardiomyocytes with the most common being an increase in the beat rate, while QT prolongation was also observed at the lowest concentrations. Finally, concentration addition modeling using quantitative data from the 6 common ingredients and multivariate prediction modeling was used to determine potential ingredients responsible for the adverse effects on the cardiomyocytes. These analyses suggested theophylline, adenine, and azelate as possibly contributing to the in vitro effects of energy drinks on QT prolongation in cardiomyocytes.
Collapse
Affiliation(s)
- Yu-Syuan Luo
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Zunwei Chen
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Alexander D Blanchette
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Yi-Hui Zhou
- Departments of Statistics and Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Fred A Wright
- Departments of Statistics and Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Erin S Baker
- Department of Chemistry, North Carolina State University, Raleigh, NC, USA
| | - Weihsueh A Chiu
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
28
|
Chanthra N, Uosaki H. Maturity of Pluripotent Stem Cell-Derived Cardiomyocytes and Future Perspectives for Regenerative Medicine. Stem Cells 2021. [DOI: 10.1007/978-3-030-77052-5_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
29
|
Wei X, Zhuang L, Li H, He C, Wan H, Hu N, Wang P. Advances in Multidimensional Cardiac Biosensing Technologies: From Electrophysiology to Mechanical Motion and Contractile Force. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2005828. [PMID: 33230867 DOI: 10.1002/smll.202005828] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Indexed: 06/11/2023]
Abstract
Cardiovascular disease is currently a leading killer to human, while drug-induced cardiotoxicity remains the main cause of the withdrawal and attrition of drugs. Taking clinical correlation and throughput into account, cardiomyocyte is perfect as in vitro cardiac model for heart disease modeling, drug discovery, and cardiotoxicity assessment by accurately measuring the physiological multiparameters of cardiomyocytes. Remarkably, cardiomyocytes present both electrophysiological and biomechanical characteristics due to the unique excitation-contraction coupling, which plays a significant role in studying the cardiomyocytes. This review mainly focuses on the recent advances of biosensing technologies for the 2D and 3D cardiac models with three special properties: electrophysiology, mechanical motion, and contractile force. These high-performance multidimensional cardiac models are popular and effective to rebuild and mimic the heart in vitro. To help understand the high-quality and accurate physiologies, related detection techniques are highly demanded, from microtechnology to nanotechnology, from extracellular to intracellular recording, from multiple cells to single cell, and from planar to 3D models. Furthermore, the characteristics, advantages, limitations, and applications of these cardiac biosensing technologies, as well as the future development prospects should contribute to the systematization and expansion of knowledge.
Collapse
Affiliation(s)
- Xinwei Wei
- Department of Biomedical Engineering, Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Liujing Zhuang
- Department of Biomedical Engineering, Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Hongbo Li
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou, 510006, China
| | - Chuanjiang He
- Department of Biomedical Engineering, Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China
| | - Hao Wan
- Department of Biomedical Engineering, Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Ning Hu
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou, 510006, China
| | - Ping Wang
- Department of Biomedical Engineering, Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| |
Collapse
|
30
|
Veldhuizen J, Cutts J, Brafman DA, Migrino RQ, Nikkhah M. Engineering anisotropic human stem cell-derived three-dimensional cardiac tissue on-a-chip. Biomaterials 2020; 256:120195. [DOI: 10.1016/j.biomaterials.2020.120195] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/15/2020] [Accepted: 06/09/2020] [Indexed: 01/02/2023]
|
31
|
Assessment of Cardiotoxicity With Stem Cell-based Strategies. Clin Ther 2020; 42:1892-1910. [PMID: 32938533 DOI: 10.1016/j.clinthera.2020.08.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/20/2020] [Accepted: 08/25/2020] [Indexed: 02/08/2023]
Abstract
PURPOSE Adverse cardiovascular drug effects pose a substantial medical risk and represent a common cause of drug withdrawal from the market. Thus, current in vitro assays and in vivo animal models still have shortcomings in assessing cardiotoxicity. A human model for more accurate preclinical cardiotoxicity assessment is highly desirable. Current differentiation protocols allow for the generation of human pluripotent stem cell-derived cardiomyocytes in basically unlimited numbers and offer the opportunity to study drug effects on human cardiomyocytes. The purpose of this review is to provide a brief overview of the current approaches to translate studies with pluripotent stem cell-derived cardiomyocytes from basic science to preclinical risk assessment. METHODS A review of the literature was performed to gather data on the pathophysiology of cardiotoxicity, the current cardiotoxicity screening assays, stem cell-derived cardiomyocytes, and their application in cardiotoxicity screening. FINDINGS There is increasing evidence that stem cell-derived cardiomyocytes predict arrhythmogenicity with high accuracy. Cardiomyocyte immaturity represents the major limitation so far. However, strategies are being developed to overcome this hurdle, such as tissue engineering. In addition, stem cell-based strategies offer the possibility to assess structural drug toxicity (eg, by anticancer drugs) on complex models that more closely mirror the structure of the heart and contain endothelial cells and fibroblasts. IMPLICATIONS Pluripotent stem cell-derived cardiomyocytes have the potential to substantially change how preclinical cardiotoxicity screening is performed. To which extent they will replace or complement current approaches is being evaluated.
Collapse
|
32
|
Li H, Fang J, Wei X, Xu D, Zhang T, Xiang Y, Chen HJ, Liu F, Xie X, Wang P, Hu N. Specific recognition of ion channel blocker by high-content cardiomyocyte electromechanical integrated correlation. Biosens Bioelectron 2020; 162:112273. [DOI: 10.1016/j.bios.2020.112273] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 04/29/2020] [Accepted: 05/02/2020] [Indexed: 12/22/2022]
|
33
|
Choi SW, Cho YW, Kim JG, Kim YJ, Kim E, Chung HM, Kang SW. Effect of Cell Labeling on the Function of Human Pluripotent Stem Cell-Derived Cardiomyocytes. Int J Stem Cells 2020; 13:287-294. [PMID: 32323512 PMCID: PMC7378900 DOI: 10.15283/ijsc19138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 03/01/2020] [Accepted: 03/03/2020] [Indexed: 11/09/2022] Open
Abstract
Cell labeling technologies are required to monitor the fate of transplanted cells in vivo and to select target cells for the observation of certain changes in vitro. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have been transplanted for the treatment of heart injuries or used in vitro for preclinical cardiac safety assessments. Cardiomyocyte (CM) labeling has been used in these processes to facilitate target cell monitoring. However, the functional effect of the labeling agent on hiPSC-CMs has not been studied. Therefore, we investigated the effects of labeling agents on CM cellular functions. 3'-Dioctadecyloxacarbocyanine perchlorate (DiO), quantum dots (QDs), and a DNA plasmid expressing EGFP using Lipo2K were used to label hiPSC-CMs. We conclude that the hiPSC-CM labeling with DiO and QDs does not induce arrhythmogenic effects but rather improves the mRNA expression of cardiac ion channels and Ca2+ influx by L-type Ca2+ channels. Thus, DiO and QD labeling agents may be useful tools to monitor transplanted CMs, and further in vivo influences of the labeling agents should be investigated in the future.
Collapse
Affiliation(s)
- Seong Woo Choi
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Young-Woo Cho
- Department of Pharmacy, Chungbuk National University College of Pharmacy, Cheongju, Korea.,Division of Drug Evaluation, NDDC, Oseong Medical Innovation Foundation, Cheongju, Korea
| | - Jae Gon Kim
- Research Group for Biomimetic Advanced Technology, Korea Institute of Toxicology, Daejeon, Korea
| | - Yong-Jin Kim
- R&D Unit, Amorepacific Corporation, Yongin, Korea
| | - Eunmi Kim
- R&D Unit, Amorepacific Corporation, Yongin, Korea
| | - Hyung-Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Sun-Woong Kang
- Department of Human and Environmental Toxicology, University of Science and Technology, Daejeon, Korea
| |
Collapse
|
34
|
Modeling Cardiovascular Diseases with hiPSC-Derived Cardiomyocytes in 2D and 3D Cultures. Int J Mol Sci 2020; 21:ijms21093404. [PMID: 32403456 PMCID: PMC7246991 DOI: 10.3390/ijms21093404] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 12/15/2022] Open
Abstract
In the last decade, the generation of cardiac disease models based on human-induced pluripotent stem cells (hiPSCs) has become of common use, providing new opportunities to overcome the lack of appropriate cardiac models. Although much progress has been made toward the generation of hiPSC-derived cardiomyocytes (hiPS-CMs), several lines of evidence indicate that two-dimensional (2D) cell culturing presents significant limitations, including hiPS-CMs immaturity and the absence of interaction between different cell types and the extracellular matrix. More recently, new advances in bioengineering and co-culture systems have allowed the generation of three-dimensional (3D) constructs based on hiPSC-derived cells. Within these systems, biochemical and physical stimuli influence the maturation of hiPS-CMs, which can show structural and functional properties more similar to those present in adult cardiomyocytes. In this review, we describe the latest advances in 2D- and 3D-hiPSC technology for cardiac disease mechanisms investigation, drug development, and therapeutic studies.
Collapse
|
35
|
Thavandiran N, Hale C, Blit P, Sandberg ML, McElvain ME, Gagliardi M, Sun B, Witty A, Graham G, Do VTH, Bakooshli MA, Le H, Ostblom J, McEwen S, Chau E, Prowse A, Fernandes I, Norman A, Gilbert PM, Keller G, Tagari P, Xu H, Radisic M, Zandstra PW. Functional arrays of human pluripotent stem cell-derived cardiac microtissues. Sci Rep 2020; 10:6919. [PMID: 32332814 PMCID: PMC7181791 DOI: 10.1038/s41598-020-62955-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 03/18/2020] [Indexed: 11/09/2022] Open
Abstract
To accelerate the cardiac drug discovery pipeline, we set out to develop a platform that would be capable of quantifying tissue-level functions such as contractile force and be amenable to standard multiwell-plate manipulations. We report a 96-well-based array of 3D human pluripotent stem cell (hPSC)-derived cardiac microtissues - termed Cardiac MicroRings (CaMiRi) - in custom 3D-print-molded multiwell plates capable of contractile force measurement. Within each well, two elastomeric microcantilevers are situated above a circumferential ramp. The wells are seeded with cell-laden collagen, which, in response to the gradual slope of the circumferential ramp, self-organizes around tip-gated microcantilevers to form contracting CaMiRi. The contractile force exerted by the CaMiRi is measured and calculated using the deflection of the cantilevers. Platform responses were robust and comparable across wells, and we used it to determine an optimal tissue formulation. We validated the contractile force response of CaMiRi using selected cardiotropic compounds with known effects. Additionally, we developed automated protocols for CaMiRi seeding, image acquisition, and analysis to enable the measurement of contractile force with increased throughput. The unique tissue fabrication properties of the platform, and the consequent effects on tissue function, were demonstrated upon adding hPSC-derived epicardial cells to the system. This platform represents an open-source contractile force screening system useful for drug screening and tissue engineering applications.
Collapse
Affiliation(s)
- Nimalan Thavandiran
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.,Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Christopher Hale
- Amgen Discovery Research, Amgen Inc., South San Francisco, CA, USA
| | | | | | | | - Mark Gagliardi
- McEwen Centre for Regenerative Medicine, University Health Network, Toronto, Ontario, Canada
| | - Bo Sun
- McEwen Centre for Regenerative Medicine, University Health Network, Toronto, Ontario, Canada
| | - Alec Witty
- McEwen Centre for Regenerative Medicine, University Health Network, Toronto, Ontario, Canada
| | | | | | - Mohsen Afshar Bakooshli
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Hon Le
- Amgen Discovery Research, Amgen Inc., South San Francisco, CA, USA
| | - Joel Ostblom
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Samuel McEwen
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Erik Chau
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario, Canada
| | | | - Ian Fernandes
- McEwen Centre for Regenerative Medicine, University Health Network, Toronto, Ontario, Canada
| | | | - Penney M Gilbert
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.,Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada.,Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada.,Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Gordon Keller
- McEwen Centre for Regenerative Medicine, University Health Network, Toronto, Ontario, Canada
| | - Philip Tagari
- Amgen Discovery Research, Amgen Inc., South San Francisco, CA, USA
| | - Han Xu
- A2 Biotherapeutics Inc., Agoura Hills, CA, USA.
| | - Milica Radisic
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada. .,Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada. .,Heart and Stroke/Richard Lewar Centre of Excellence, University of Toronto, Toronto, Ontario, Canada.
| | - Peter W Zandstra
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada. .,CCRM, Toronto, Ontario, Canada. .,Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada. .,Michael Smith Laboratories, School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
36
|
Gintant G, Traebert M. The roles of human induced pluripotent stem cell-derived cardiomyocytes in drug discovery: managing in vitro safety study expectations. Expert Opin Drug Discov 2020; 15:719-729. [PMID: 32129680 DOI: 10.1080/17460441.2020.1736549] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Human induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) preparations are increasingly employed in in vitro cardiac safety studies to support candidate drug selection and regulatory submissions. The value of hiPSC-CM-based approaches depends on their ability to recapitulate the cellular mechanisms responsible for cardiotoxicity as well as overall assay characteristics (thus defining model performance). Different expectations at different drug development stages define the utility of these human-derived models. AREAS COVERED Herein, the authors review the importance of understanding the functional characteristics of the evolving spectrum of simpler (2D) and more complex (co-cultures, 3D constructs, and engineered tissues) human-derived cardiac preparations, and how their performance may be evaluated based on analytical sensitivity, variability, and reproducibility in order to correctly match preparations with expectations of different safety assays. The need for consensus clinical examples of electrophysiologic, contractile, and structural cardiotoxicities essential for benchmarking human-derived models is also discussed. EXPERT OPINION It is helpful (but not essential) that hiPSC-CMs preparations fully recapitulate pharmacological responses of native adult human ventricular myocytes when evaluating cardiotoxicity in vitro. Further calibration and model standardization (aligning concordance with clinical findings) are necessary to understand the role of hiPSC-CMs in guiding cardiotoxicity assessments in early drug discovery efforts.
Collapse
Affiliation(s)
- Gary Gintant
- Department of Integrative Pharmacology (ZR13), AP-9A-LL, AbbVie Inc. , North Chicago, IL, USA
| | - Martin Traebert
- Novartis Institutes for Biomedical Research , Safety Pharmacology, Basel, Switzerland
| |
Collapse
|
37
|
Golforoush P, Schneider MD. Intensive care for human hearts in pluripotent stem cell models. NPJ Regen Med 2020; 5:4. [PMID: 32194989 PMCID: PMC7060343 DOI: 10.1038/s41536-020-0090-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 02/06/2020] [Indexed: 12/12/2022] Open
Abstract
Successful drug discovery is ultimately contingent on the availability of workable, relevant, predictive model systems. Conversely, for cardiac muscle, the lack of human preclinical models to inform target validation and compound development has likely contributed to the perennial problem of clinical trial failures, despite encouraging non-human results. By contrast, human cardiomyocytes produced from pluripotent stem cell models have recently been applied to safety pharmacology, phenotypic screening, target validation and high-throughput assays, facilitating cardiac drug discovery. Here, we review the impact of human pluripotent stem cell models in cardiac drug discovery, discussing the range of applications, readouts, and disease models employed, along with the challenges and prospects to advance this fruitful mode of research further.
Collapse
Affiliation(s)
- Pelin Golforoush
- National Heart and Lung Institute, Imperial College London, London, W12 0NN UK
| | | |
Collapse
|
38
|
Huo J, Wei F, Cai C, Lyn-Cook B, Pang L. Sex-Related Differences in Drug-Induced QT Prolongation and Torsades de Pointes: A New Model System with Human iPSC-CMs. Toxicol Sci 2020; 167:360-374. [PMID: 30247688 DOI: 10.1093/toxsci/kfy239] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Numerous drugs have the potential to prolong the QT interval and may cause accidental cardiac arrest (torsades de pointes [TdP]). Women are at a higher risk than men for experiencing drug-induced TdP. Due to the lack of appropriate tools, few studies have investigated whether genetic differences between men and women have any effects on drug-induced proarrhythmia. Sex hormones are believed to play a predominant role in the induction of TdP. Recently, progress in induced pluripotent stem cell (iPSC) technologies has made it possible to utilize human iPSC-derived cardiomyocytes (hiPSC-CMs) to investigate the influence of both genetics and sex hormones on cardiac ion channel gene expression and cardiomyocyte function. In this study, we investigated genetic and hormonal effects on sex differences of drug-induced QT prolongation and TdP with hiPSC-CMs from healthy male and female donors. We found that despite batch variations in beating rates and field potential durations (FPD), female-derived hiPSC-CMs showed steeper slopes of FPD to interspike interval ratios and were more sensitive to IKr blocker-induced FPD prolongation. 17β-estradiol increased FPD and 5α-dihydrotestosterone shortened FPD, but the addition of sex hormones had limited effect on the responses of hiPSC-CMs to IKr blockades. The differential expression of KCNE1 gene and reduced repolarization reserve in female-derived hiPSC-CMs compared with male-derived hiPSC-CMs may partially explain why females are more susceptible to proarrhythmias. Human iPSC-CMs can be a useful new model to study mechanisms of sex differences in cardiomyocyte repolarization processes and aid in the prediction of drug-induced proarrhythmias in both men and women.
Collapse
Affiliation(s)
- Jianhua Huo
- Division of Systems Biology, National Center for Toxicological Research, U.S. FDA, Jefferson, Arkansas 72079.,Department of Cardiovascular Medicine, First Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Feng Wei
- Division of Systems Biology, National Center for Toxicological Research, U.S. FDA, Jefferson, Arkansas 72079.,Department of Cardiovascular Medicine, First Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Chengzhong Cai
- Division of Systems Biology, National Center for Toxicological Research, U.S. FDA, Jefferson, Arkansas 72079
| | - Beverly Lyn-Cook
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. FDA, Jefferson, Arkansas 72079
| | - Li Pang
- Division of Systems Biology, National Center for Toxicological Research, U.S. FDA, Jefferson, Arkansas 72079
| |
Collapse
|
39
|
Takasuna K, Kazusa K, Hayakawa T. Comprehensive Cardiac Safety Assessment using hiPS-cardiomyocytes (Consortium for Safety Assessment using Human iPS Cells: CSAHi). Curr Pharm Biotechnol 2019; 21:829-841. [PMID: 31749424 DOI: 10.2174/1389201020666191024172425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 09/16/2019] [Accepted: 09/24/2019] [Indexed: 11/22/2022]
Abstract
Current cardiac safety assessment platforms (in vitro hERG-centric, APD, and/or in vivo animal QT assays) are not fully predictive of drug-induced Torsades de Pointes (TdP) and do not address other mechanism-based arrhythmia, including ventricular tachycardia or ventricular fibrillation, or cardiac safety liabilities such as contractile and structural cardiotoxicity which are another growing safety concerns. We organized the Consortium for Safety Assessment using Human iPS cells (CSAHi; http://csahi.org/en/) in 2013, based on the Japan Pharmaceutical Manufacturers Association (JPMA), to verify the application of human iPS/ES cell-derived cardiomyocytes for drug safety evaluation. The CSAHi HEART team focused on comprehensive screening strategies to predict a diverse range of cardiotoxicities using recently introduced platforms such as the Multi-Electrode Array (MEA), cellular impedance, Motion Field Imaging (MFI), and optical imaging of Ca transient to identify strengths and weaknesses of each platform. Our study showed that hiPS-CMs used in these platforms could detect pharmacological responses that were more relevant to humans compared to existing hERG, APD, or Langendorff (MAPD/contraction) assays. Further, MEA and other methods such as impedance, MFI, and Ca transient assays provided paradigm changes of platforms for predicting drug-induced QT risk and/or arrhythmia or contractile dysfunctions. In contrast, since discordances such as overestimation (false positive) of arrhythmogenicity, oversight, or opposite conclusions in positive inotropic and negative chronotropic activities to some compounds were also confirmed, possibly due to their functional immaturity of hiPS-CMs, hiPS-CMs should be used in these platforms for cardiac safety assessment based upon their advantages and disadvantages.
Collapse
Affiliation(s)
- Kiyoshi Takasuna
- Consortium for Safety Assessment using Human iPS Cells (CSAHi), Heart Team, Japan
| | - Katsuyuki Kazusa
- Consortium for Safety Assessment using Human iPS cells (CSAHi), Heart team, Japan
| | - Tomohiro Hayakawa
- Consortium for Safety Assessment using Human iPS cells (CSAHi), Heart team, Japan
| |
Collapse
|
40
|
Pang L, Sager P, Yang X, Shi H, Sannajust F, Brock M, Wu JC, Abi-Gerges N, Lyn-Cook B, Berridge BR, Stockbridge N. Workshop Report: FDA Workshop on Improving Cardiotoxicity Assessment With Human-Relevant Platforms. Circ Res 2019; 125:855-867. [PMID: 31600125 DOI: 10.1161/circresaha.119.315378] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Given that cardiovascular safety concerns remain the leading cause of drug attrition at the preclinical drug development stage, the National Center for Toxicological Research of the US Food and Drug Administration hosted a workshop to discuss current gaps and challenges in translating preclinical cardiovascular safety data to humans. This white paper summarizes the topics presented by speakers from academia, industry, and government intended to address the theme of improving cardiotoxicity assessment in drug development. The main conclusion is that to reduce cardiovascular safety liabilities of new therapeutic agents, there is an urgent need to integrate human-relevant platforms/approaches into drug development. Potential regulatory applications of human-derived cardiomyocytes and future directions in employing human-relevant platforms to fill the gaps and overcome barriers and challenges in preclinical cardiovascular safety assessment were discussed. This paper is intended to serve as an initial step in a public-private collaborative development program for human-relevant cardiotoxicity tools, particularly for cardiotoxicities characterized by contractile dysfunction or structural injury.
Collapse
Affiliation(s)
- Li Pang
- From the Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration (L.P.)
| | | | - Xi Yang
- Division of Cardiovascular and Renal Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration (X.Y.)
| | - Hong Shi
- Discovery Toxicology, Bristol-Myers Squibb (BMS) Company (H.S.)
| | - Frederick Sannajust
- Safety & Exploratory Pharmacology Department, SALAR Division, Merck & Co (F.S.)
| | | | - Joseph C Wu
- Stanford University School of Medicine, Stanford Cardiovascular Institute (J.C.W.)
| | | | - Beverly Lyn-Cook
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration (B.L.-C.)
| | - Brian R Berridge
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health (B.R.B.)
| | - Norman Stockbridge
- Division of Cardiovascular and Renal Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration (N.S.)
| |
Collapse
|
41
|
van Meer BJ, Krotenberg A, Sala L, Davis RP, Eschenhagen T, Denning C, Tertoolen LGJ, Mummery CL. Simultaneous measurement of excitation-contraction coupling parameters identifies mechanisms underlying contractile responses of hiPSC-derived cardiomyocytes. Nat Commun 2019; 10:4325. [PMID: 31541103 PMCID: PMC6754438 DOI: 10.1038/s41467-019-12354-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 09/04/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiomyocytes from human induced pluripotent stem cells (hiPSC-CMs) are increasingly recognized as valuable for determining the effects of drugs on ion channels but they do not always accurately predict contractile responses of the human heart. This is in part attributable to their immaturity but the sensitivity of measurement tools may also be limiting. Measuring action potential, calcium flux or contraction individually misses critical information that is captured when interrogating the complete excitation-contraction coupling cascade simultaneously. Here, we develop an hypothesis-based statistical algorithm that identifies mechanisms of action. We design and build a high-speed optical system to measure action potential, cytosolic calcium and contraction simultaneously using fluorescent sensors. These measurements are automatically processed, quantified and then assessed by the algorithm. Multiplexing these three critical physical features of hiPSC-CMs allows identification of all major drug classes affecting contractility with detection sensitivities higher than individual measurement of action potential, cytosolic calcium or contraction.
Collapse
Affiliation(s)
- Berend J van Meer
- Dept. of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZD, Leiden, The Netherlands
| | - Ana Krotenberg
- Dept. of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZD, Leiden, The Netherlands
| | - Luca Sala
- Dept. of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZD, Leiden, The Netherlands.,Istituto Auxologico Italiano, IRCCS, Center for Cardiac Arrhythmias of Genetic Origin, Laboratory of Cardiovascular Genetics, Via Zucchi 18, 20095, Cusano Milanino, MI, Italy
| | - Richard P Davis
- Dept. of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZD, Leiden, The Netherlands
| | - Thomas Eschenhagen
- Dept. of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Chris Denning
- Dept. of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Leon G J Tertoolen
- Dept. of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZD, Leiden, The Netherlands
| | - Christine L Mummery
- Dept. of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZD, Leiden, The Netherlands. .,Dept. of Applied Stem Cell Technologies, University of Twente, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands.
| |
Collapse
|
42
|
Kügler P, Rast G, Guth BD. Comparison of in vitro and computational experiments on the relation of inter-beat interval and duration of repolarization in a specific type of human induced pluripotent stem cell-derived cardiomyocytes. PLoS One 2019; 14:e0221763. [PMID: 31498812 PMCID: PMC6733510 DOI: 10.1371/journal.pone.0221763] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 08/14/2019] [Indexed: 12/20/2022] Open
Abstract
We compared a published computational model of the action potential of a specific type of human induced pluripotent stem cell -derived cardiomyocytes (hiPSC-CM) with experimental field potential data with regard to their inter-beat interval and the duration of repolarization. In particular, concomitant changes in inter-beat interval and duration of repolarization were calculated after reduction and/or augmentation of specific ion channel conductances as a surrogate for pharmacological manipulation. The observed mismatches between calculations and experimental data indicate that there is information missing about the cellular test system. Based on our results we hypothesize that, among other currents, the actual If (“funny current”) may deviate from the prediction. We show that replacement of the If formulation by alternative equations causes the model predictions to change qualitatively, however, none of the available formulations is actually achieving a satisfactory match with experimental data. We suggest a strategy to clarify whether the mismatch can be completely resolved at all using single cell models and, if yes, how this goal could be reached.
Collapse
Affiliation(s)
- Philipp Kügler
- Institute of Applied Mathematics and Statistics, Computational Science Lab, University of Hohenheim, Stuttgart, Germany
| | - Georg Rast
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
- * E-mail:
| | - Brian D. Guth
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
- Department of Pharmaceutical Sciences, North-West University, Potchefstroom Campus, Potchefstroom, South Africa
| |
Collapse
|
43
|
HiPSC-CMs from different sex and ethnic origin donors exhibit qualitatively different responses to several classes of pharmacological challenges. J Pharmacol Toxicol Methods 2019; 99:106598. [DOI: 10.1016/j.vascn.2019.106598] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/29/2019] [Accepted: 06/11/2019] [Indexed: 12/28/2022]
|
44
|
Guth BD, Engwall M, Eldridge S, Foley CM, Guo L, Gintant G, Koerner J, Parish ST, Pierson JB, Ribeiro AJS, Zabka T, Chaudhary KW, Kanda Y, Berridge B. Considerations for an In Vitro, Cell-Based Testing Platform for Detection of Adverse Drug-Induced Inotropic Effects in Early Drug Development. Part 1: General Considerations for Development of Novel Testing Platforms. Front Pharmacol 2019; 10:884. [PMID: 31447679 PMCID: PMC6697071 DOI: 10.3389/fphar.2019.00884] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 07/15/2019] [Indexed: 01/10/2023] Open
Abstract
Drug-induced effects on cardiac contractility can be assessed through the measurement of the maximal rate of pressure increase in the left ventricle (LVdP/dtmax) in conscious animals, and such studies are often conducted at the late stage of preclinical drug development. Detection of such effects earlier in drug research using simpler, in vitro test systems would be a valuable addition to our strategies for identifying the best possible drug development candidates. Thus, testing platforms with reasonably high throughput, and affordable costs would be helpful for early screening purposes. There may also be utility for testing platforms that provide mechanistic information about how a given drug affects cardiac contractility. Finally, there could be in vitro testing platforms that could ultimately contribute to the regulatory safety package of a new drug. The characteristics needed for a successful cell or tissue-based testing platform for cardiac contractility will be dictated by its intended use. In this article, general considerations are presented with the intent of guiding the development of new testing platforms that will find utility in drug research and development. In the following article (part 2), specific aspects of using human-induced stem cell-derived cardiomyocytes for this purpose are addressed.
Collapse
Affiliation(s)
- Brian D Guth
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany.,PreClinical Drug Development Platform (PCDDP), North-West University, Potchefstroom, South Africa
| | - Michael Engwall
- Safety Pharmacology and Animal Research Center, Amgen Research, Thousand Oaks, CA, United States
| | - Sandy Eldridge
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - C Michael Foley
- Department of Integrative Pharmacology, Integrated Sciences and Technology, AbbVie, North Chicago, IL, United States
| | - Liang Guo
- Laboratory of Investigative Toxicology, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | - Gary Gintant
- Department of Integrative Pharmacology, Integrated Sciences and Technology, AbbVie, North Chicago, IL, United States
| | - John Koerner
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Stanley T Parish
- Health and Environmental Sciences Institute, Washington, DC, United States
| | - Jennifer B Pierson
- Health and Environmental Sciences Institute, Washington, DC, United States
| | - Alexandre J S Ribeiro
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translation Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Tanja Zabka
- Department of Safety Assessment, Genentech, South San Francisco, CA, United States
| | - Khuram W Chaudhary
- Global Safety Pharmacology, GlaxoSmithKline plc, Collegeville, PA, United States
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, Kanagawa, Japan
| | - Brian Berridge
- National Toxicology Program, National Institute of Environmental Health Sciences, Durham, NC, United States
| |
Collapse
|
45
|
Kernik DC, Morotti S, Wu H, Garg P, Duff HJ, Kurokawa J, Jalife J, Wu JC, Grandi E, Clancy CE. A computational model of induced pluripotent stem-cell derived cardiomyocytes incorporating experimental variability from multiple data sources. J Physiol 2019; 597:4533-4564. [PMID: 31278749 PMCID: PMC6767694 DOI: 10.1113/jp277724] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 07/05/2019] [Indexed: 12/22/2022] Open
Abstract
Key points Induced pluripotent stem cell‐derived cardiomyocytes (iPSC‐CMs) capture patient‐specific genotype–phenotype relationships, as well as cell‐to‐cell variability of cardiac electrical activity Computational modelling and simulation provide a high throughput approach to reconcile multiple datasets describing physiological variability, and also identify vulnerable parameter regimes We have developed a whole‐cell model of iPSC‐CMs, composed of single exponential voltage‐dependent gating variable rate constants, parameterized to fit experimental iPSC‐CM outputs We have utilized experimental data across multiple laboratories to model experimental variability and investigate subcellular phenotypic mechanisms in iPSC‐CMs This framework links molecular mechanisms to cellular‐level outputs by revealing unique subsets of model parameters linked to known iPSC‐CM phenotypes
Abstract There is a profound need to develop a strategy for predicting patient‐to‐patient vulnerability in the emergence of cardiac arrhythmia. A promising in vitro method to address patient‐specific proclivity to cardiac disease utilizes induced pluripotent stem cell‐derived cardiomyocytes (iPSC‐CMs). A major strength of this approach is that iPSC‐CMs contain donor genetic information and therefore capture patient‐specific genotype–phenotype relationships. A cited detriment of iPSC‐CMs is the cell‐to‐cell variability observed in electrical activity. We postulated, however, that cell‐to‐cell variability may constitute a strength when appropriately utilized in a computational framework to build cell populations that can be employed to identify phenotypic mechanisms and pinpoint key sensitive parameters. Thus, we have exploited variation in experimental data across multiple laboratories to develop a computational framework for investigating subcellular phenotypic mechanisms. We have developed a whole‐cell model of iPSC‐CMs composed of simple model components comprising ion channel models with single exponential voltage‐dependent gating variable rate constants, parameterized to fit experimental iPSC‐CM data for all major ionic currents. By optimizing ionic current model parameters to multiple experimental datasets, we incorporate experimentally‐observed variability in the ionic currents. The resulting population of cellular models predicts robust inter‐subject variability in iPSC‐CMs. This approach links molecular mechanisms to known cellular‐level iPSC‐CM phenotypes, as shown by comparing immature and mature subpopulations of models to analyse the contributing factors underlying each phenotype. In the future, the presented models can be readily expanded to include genetic mutations and pharmacological interventions for studying the mechanisms of rare events, such as arrhythmia triggers. Induced pluripotent stem cell‐derived cardiomyocytes (iPSC‐CMs) capture patient‐specific genotype–phenotype relationships, as well as cell‐to‐cell variability of cardiac electrical activity Computational modelling and simulation provide a high throughput approach to reconcile multiple datasets describing physiological variability, and also identify vulnerable parameter regimes We have developed a whole‐cell model of iPSC‐CMs, composed of single exponential voltage‐dependent gating variable rate constants, parameterized to fit experimental iPSC‐CM outputs We have utilized experimental data across multiple laboratories to model experimental variability and investigate subcellular phenotypic mechanisms in iPSC‐CMs This framework links molecular mechanisms to cellular‐level outputs by revealing unique subsets of model parameters linked to known iPSC‐CM phenotypes
Collapse
Affiliation(s)
- Divya C Kernik
- Department of Physiology and Membrane Biology, Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, USA
| | - Stefano Morotti
- Department of Pharmacology, School of Medicine, University of California, Davis, CA, USA
| | - HaoDi Wu
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Priyanka Garg
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Henry J Duff
- Libin Cardiovascular Institute of Alberta, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
| | - Junko Kurokawa
- Department of Bio-Informational Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - José Jalife
- Department of Internal Medicine, Center for Arrhythmia Research, Cardiovascular Research Center, University of Michigan, Ann Arbor, MI, USA.,Centro Nacional de Investigaciones Cardiovasculares (CNIC), and CIBERV, Madrid, Spain
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Eleonora Grandi
- Department of Pharmacology, School of Medicine, University of California, Davis, CA, USA
| | - Colleen E Clancy
- Department of Physiology and Membrane Biology, Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, USA
| |
Collapse
|
46
|
Pfeiffer-Kaushik ER, Smith GL, Cai B, Dempsey GT, Hortigon-Vinagre MP, Zamora V, Feng S, Ingermanson R, Zhu R, Hariharan V, Nguyen C, Pierson J, Gintant GA, Tung L. Electrophysiological characterization of drug response in hSC-derived cardiomyocytes using voltage-sensitive optical platforms. J Pharmacol Toxicol Methods 2019; 99:106612. [PMID: 31319140 DOI: 10.1016/j.vascn.2019.106612] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 06/30/2019] [Accepted: 07/10/2019] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Voltage-sensitive optical (VSO) sensors offer a minimally invasive method to study the time course of repolarization of the cardiac action potential (AP). This Comprehensive in vitro Proarrhythmia Assay (CiPA) cross-platform study investigates protocol design and measurement variability of VSO sensors for preclinical cardiac electrophysiology assays. METHODS Three commercial and one academic laboratory completed a limited study of the effects of 8 blinded compounds on the electrophysiology of 2 commercial lines of human induced pluripotent stem-cell derived cardiomyocytes (hSC-CMs). Acquisition technologies included CMOS camera and photometry; fluorescent voltage sensors included di-4-ANEPPS, FluoVolt and genetically encoded QuasAr2. The experimental protocol was standardized with respect to cell lines, plating and maintenance media, blinded compounds, and action potential parameters measured. Serum-free media was used to study the action of drugs, but the exact composition and the protocols for cell preparation and drug additions varied among sites. RESULTS Baseline AP waveforms differed across platforms and between cell types. Despite these differences, the relative responses to four selective ion channel blockers (E-4031, nifedipine, mexiletine, and JNJ 303 blocking IKr, ICaL, INa, and IKs, respectively) were similar across all platforms and cell lines although the absolute changes differed. Similarly, four mixed ion channel blockers (flecainide, moxifloxacin, quinidine, and ranolazine) had comparable effects in all platforms. Differences in repolarisation time course and response to drugs could be attributed to cell type and experimental method differences such as composition of the assay media, stimulated versus spontaneous activity, and single versus cumulative compound addition. DISCUSSION In conclusion, VSOs represent a powerful and appropriate method to assess the electrophysiological effects of drugs on iPSC-CMs for the evaluation of proarrhythmic risk. Protocol considerations and recommendations are provided toward standardizing conditions to reduce variability of baseline AP waveform characteristics and drug responses.
Collapse
Affiliation(s)
| | - Godfrey L Smith
- Clyde Biosciences Ltd, BioCity Scotland, Bo'Ness Road, Newhouse, Lanarkshire, Scotland ML1 5UH, United Kingdom; Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Science, University of Glasgow, 126 University Place, Glasgow G12 8TA, United Kingdom
| | - Beibei Cai
- Vala Sciences Inc., 6370 Nancy Ridge Drive, Suite 106, San Diego, CA 92121, USA
| | - Graham T Dempsey
- Q-State Biosciences Inc., 179 Sidney Street, Cambridge, MA 02139, USA
| | - Maria P Hortigon-Vinagre
- Clyde Biosciences Ltd, BioCity Scotland, Bo'Ness Road, Newhouse, Lanarkshire, Scotland ML1 5UH, United Kingdom; Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Science, University of Glasgow, 126 University Place, Glasgow G12 8TA, United Kingdom
| | - Victor Zamora
- Clyde Biosciences Ltd, BioCity Scotland, Bo'Ness Road, Newhouse, Lanarkshire, Scotland ML1 5UH, United Kingdom; Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Science, University of Glasgow, 126 University Place, Glasgow G12 8TA, United Kingdom
| | - Shuyun Feng
- Vala Sciences Inc., 6370 Nancy Ridge Drive, Suite 106, San Diego, CA 92121, USA
| | - Randall Ingermanson
- Vala Sciences Inc., 6370 Nancy Ridge Drive, Suite 106, San Diego, CA 92121, USA
| | - Renjun Zhu
- Department of Biomedical Engineering, The Johns Hopkins University, 720 Rutland Ave., Baltimore, MD 21205, USA
| | - Venkatesh Hariharan
- Department of Biomedical Engineering, The Johns Hopkins University, 720 Rutland Ave., Baltimore, MD 21205, USA
| | - Cuong Nguyen
- Q-State Biosciences Inc., 179 Sidney Street, Cambridge, MA 02139, USA
| | - Jennifer Pierson
- Health and Environmental Sciences Institute, Washington, D.C. 20009, USA.
| | - Gary A Gintant
- AbbVie, 1 North Waukegan Road, Department ZR-13, Building AP-9A, North Chicago, IL 60064-6119, USA
| | - Leslie Tung
- Department of Biomedical Engineering, The Johns Hopkins University, 720 Rutland Ave., Baltimore, MD 21205, USA
| |
Collapse
|
47
|
Zeng H, Wang J, Clouse H, Lagrutta A, Sannajust F. Human-induced pluripotent stem cell-derived cardiomyocytes have limited I Ks for repolarization reserve as revealed by specific KCNQ1/KCNE1 blocker. JRSM Cardiovasc Dis 2019; 8:2048004019854919. [PMID: 31217965 PMCID: PMC6558757 DOI: 10.1177/2048004019854919] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/06/2019] [Accepted: 05/11/2019] [Indexed: 01/18/2023] Open
Abstract
Objective We investigated if there is IKs, and if there is repolarization
reserve by IKs in human-induced pluripotent stem cell-derived
cardiomyocytes (hiPSC-CMs). Design We used a specific KCNQ1/KCNE1 channel blocker, L-000768673, with an
IC50 of 9 nM, and four hERG-specific blockers, astemizole,
cisapride, dofetilide, and E-4031 to investigate the issue. Results L-000768673 concentration-dependently prolonged feature point duration
(FPD)―a surrogate signal of action potential duration―from 1 to 30 nM
without pacing or paced at 1.2 Hz, resulting from IKs blockade in
hiPSC-CMs. At higher concentrations, the effect of L-000768673 on
IKs was mitigated by its effect on ICa-L,
resulting in shortened FPD, reduced impedance amplitude, and increased
beating rate at 1 µM and above, recapitulating the self-limiting properties
of L-000768673 on action potentials. All four hERG-specific blockers
prolonged FPD as expected. Co-application of L-000768673 at sub-threshold
(0.1 and 0.3 nM) and threshold (1 nM) concentrations failed to
synergistically enhance the effects of hERG blockers on FPD prolongation,
rather it showed additive effects, inconsistent with the repolarization
reserve role of IKs in mature human myocytes that enhanced
IKr response, implying a difference between hiPSC-CMs used in
this study and mature human cardiomyocytes. Conclusion There was IKs current in hiPSC-CMs, and blockade of IKs
current caused prolongation of action potential of hiPSC-CMs. However, we
could not demonstrate any synergistic effects on action potential duration
prolongation of hiPSC-CMs by blocking hERG current and IKs
current simultaneously, implying little or no repolarization reserve by
IKs current in hiPSC-CMs used in this study.
Collapse
Affiliation(s)
- Haoyu Zeng
- Merck & Co., Inc., Safety and Exploratory Pharmacology, West Point, PA, USA
| | - Jixin Wang
- Merck & Co., Inc., Safety and Exploratory Pharmacology, West Point, PA, USA
| | - Holly Clouse
- Merck & Co., Inc., Safety and Exploratory Pharmacology, West Point, PA, USA
| | - Armando Lagrutta
- Merck & Co., Inc., Safety and Exploratory Pharmacology, West Point, PA, USA
| | | |
Collapse
|
48
|
Gamal W, Wu H, Underwood I, Jia J, Smith S, Bagnaninchi PO. Impedance-based cellular assays for regenerative medicine. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0226. [PMID: 29786561 DOI: 10.1098/rstb.2017.0226] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2018] [Indexed: 12/22/2022] Open
Abstract
Therapies based on regenerative techniques have the potential to radically improve healthcare in the coming years. As a result, there is an emerging need for non-destructive and label-free technologies to assess the quality of engineered tissues and cell-based products prior to their use in the clinic. In parallel, the emerging regenerative medicine industry that aims to produce stem cells and their progeny on a large scale will benefit from moving away from existing destructive biochemical assays towards data-driven automation and control at the industrial scale. Impedance-based cellular assays (IBCA) have emerged as an alternative approach to study stem-cell properties and cumulative studies, reviewed here, have shown their potential to monitor stem-cell renewal, differentiation and maturation. They offer a novel method to non-destructively assess and quality-control stem-cell cultures. In addition, when combined with in vitro disease models they provide complementary insights as label-free phenotypic assays. IBCA provide quantitative and very sensitive results that can easily be automated and up-scaled in multi-well format. When facing the emerging challenge of real-time monitoring of three-dimensional cell culture dielectric spectroscopy and electrical impedance tomography represent viable alternatives to two-dimensional impedance sensing.This article is part of the theme issue 'Designer human tissue: coming to a lab near you'.
Collapse
Affiliation(s)
- W Gamal
- School of Electronic Engineering, Bangor University, Bangor LL57 1UT, UK
| | - H Wu
- School of Engineering, University of Edinburgh, Edinburgh EH9 3FB, UK
| | - I Underwood
- School of Engineering, University of Edinburgh, Edinburgh EH9 3FB, UK
| | - J Jia
- School of Engineering, University of Edinburgh, Edinburgh EH9 3FB, UK
| | - S Smith
- School of Engineering, University of Edinburgh, Edinburgh EH9 3FB, UK
| | - P O Bagnaninchi
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| |
Collapse
|
49
|
Veldhuizen J, Migrino RQ, Nikkhah M. Three-dimensional microengineered models of human cardiac diseases. J Biol Eng 2019; 13:29. [PMID: 30988697 PMCID: PMC6448321 DOI: 10.1186/s13036-019-0155-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 03/13/2019] [Indexed: 01/17/2023] Open
Abstract
In vitro three-dimensional (3D) microengineered tissue models have been the recent focus of pathophysiological studies, particularly in the field of cardiovascular research. These models, as classified by 3D biomimetic tissues within micrometer-scale platforms, enable precise environmental control on the molecular- and cellular-levels to elucidate biological mechanisms of disease progression and enhance efficacy of therapeutic research. Microengineered models also incorporate directed stem cell differentiation and genome modification techniques that warrant derivation of patient-specific and genetically-edited human cardiac cells for precise recapitulation of diseased tissues. Additionally, integration of added functionalities and/or structures into these models serves to enhance the capability to further extract disease-specific phenotypic, genotypic, and electrophysiological information. This review highlights the recent progress in the development of in vitro 3D microengineered models for study of cardiac-related diseases (denoted as CDs). We will primarily provide a brief overview on currently available 2D assays and animal models for studying of CDs. We will further expand our discussion towards currently available 3D microengineered cardiac tissue models and their implementation for study of specific disease conditions.
Collapse
Affiliation(s)
- Jaimeson Veldhuizen
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, 501 E Tyler Mall Building ECG, Suite 334, Tempe, AZ 85287-9709 USA
| | | | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, 501 E Tyler Mall Building ECG, Suite 334, Tempe, AZ 85287-9709 USA
| |
Collapse
|
50
|
Balasubramanian B, Belak V, Verma I, Prysiazhniuk Y, Sannajust F, Trepakova ES. Cell culture conditions affect the ability of high content imaging assay to detect drug-induced changes in cellular parameters in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Toxicol Rep 2019; 6:305-320. [PMID: 31011540 PMCID: PMC6460330 DOI: 10.1016/j.toxrep.2019.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 01/30/2019] [Accepted: 02/22/2019] [Indexed: 01/17/2023] Open
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are widely used for drug safety and efficacy testing with various techniques, including high content imaging (HCI). Upon drug treatment, a significant number of hiPSC-CMs grown in regular 96-well plates coated with fibronectin detached from the bottom of the plate, complicating data acquisition. Several cell culture configurations were tested to improve cell adherence, and the effects of these configurations on total cell number, separation of feature values between the negative (DMSO 0.1%) and positive (antimycin, staurosporine) controls, scale of feature value differences, and data variability were statistically calculated. hiPSC-CMs were plated on fibronectin- (in “blanket” configuration) or MaxGel- (in “sandwich” configuration) coated plates and covered with a layer of either HydroMatrix or MaxGel 2, 7, or 11d after plating. After a total of 14d in culture, cells were treated with compounds, labeled with four fluorescent dyes (Hoechst, TMRM, NucView, and RedDot), and imaged with GE INCell2000. Based on the statistical parameters calculated, the MaxGel 25% 7d “sandwich” was superior to all other tested conditions when the cells were treated with 0.3 μM antimycin for 2 h and test compounds 10 μM crizotinib and 30 μM amiodarone for 48 h. For staurosporine treatment, the best culturing condition varied between MaxGel “sandwich” systems, depending on which parameters were under consideration. Thus, cell culturing conditions can significantly affect the ability of high content imaging to detect changes in cellular features during compound treatment and should be thoroughly evaluated before committing to compound testing.
Collapse
Affiliation(s)
- Bharathi Balasubramanian
- Department of Safety and Exploratory Pharmacology, Safety Assessment and Laboratory Animal Resources, MRL, Merck & Co., Inc, West Point, PA, USA
| | - Vaclav Belak
- Deparment of Data Science, MSD Global IT Innovation Center, Prague, Czech Republic
| | - Isha Verma
- Department of Data Development, Informatics & Analytics, Palo Alto, CA, USA
| | - Yeva Prysiazhniuk
- Deparment of Data Science, MSD Global IT Innovation Center, Prague, Czech Republic
| | - Frederick Sannajust
- Department of Safety and Exploratory Pharmacology, Safety Assessment and Laboratory Animal Resources, MRL, Merck & Co., Inc, West Point, PA, USA
| | - Elena S Trepakova
- Department of Safety and Exploratory Pharmacology, Safety Assessment and Laboratory Animal Resources, MRL, Merck & Co., Inc, West Point, PA, USA
| |
Collapse
|