1
|
Rosell-Hidalgo A, Bruhn C, Shardlow E, Barton R, Ryder S, Samatov T, Hackmann A, Aquino GR, Fernandes Dos Reis M, Galatenko V, Fritsch R, Dohrmann C, Walker PA. In-depth mechanistic analysis including high-throughput RNA sequencing in the prediction of functional and structural cardiotoxicants using hiPSC cardiomyocytes. Expert Opin Drug Metab Toxicol 2024; 20:685-707. [PMID: 37995132 DOI: 10.1080/17425255.2023.2273378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/05/2023] [Accepted: 09/15/2023] [Indexed: 11/25/2023]
Abstract
BACKGROUND Cardiotoxicity remains one of the most reported adverse drug reactions that lead to drug attrition during pre-clinical and clinical drug development. Drug-induced cardiotoxicity may develop as a functional change in cardiac electrophysiology (acute alteration of the mechanical function of the myocardium) and/or as a structural change, resulting in loss of viability and morphological damage to cardiac tissue. RESEARCH DESIGN AND METHODS Non-clinical models with better predictive value need to be established to improve cardiac safety pharmacology. To this end, high-throughput RNA sequencing (ScreenSeq) was combined with high-content imaging (HCI) and Ca2+ transience (CaT) to analyze compound-treated human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). RESULTS Analysis of hiPSC-CMs treated with 33 cardiotoxicants and 9 non-cardiotoxicants of mixed therapeutic indications facilitated compound clustering by mechanism of action, scoring of pathway activities related to cardiomyocyte contractility, mitochondrial integrity, metabolic state, diverse stress responses and the prediction of cardiotoxicity risk. The combination of ScreenSeq, HCI and CaT provided a high cardiotoxicity prediction performance with 89% specificity, 91% sensitivity and 90% accuracy. CONCLUSIONS Overall, this study introduces mechanism-driven risk assessment approach combining structural, functional and molecular high-throughput methods for pre-clinical risk assessment of novel compounds.
Collapse
|
2
|
Yoon SH, Lee HL, Jeong DU, Lim KM, Park SJ, Kim KS. Assessment of the proarrhythmic effects of repurposed antimalarials for COVID-19 treatment using a comprehensive in vitro proarrhythmia assay (CiPA). Front Pharmacol 2023; 14:1220796. [PMID: 37649890 PMCID: PMC10464612 DOI: 10.3389/fphar.2023.1220796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/07/2023] [Indexed: 09/01/2023] Open
Abstract
Due to the outbreak of the SARS-CoV-2 virus, drug repurposing and Emergency Use Authorization have been proposed to treat the coronavirus disease 2019 (COVID-19) during the pandemic. While the efficiency of the drugs has been discussed, it was identified that certain compounds, such as chloroquine and hydroxychloroquine, cause QT interval prolongation and potential cardiotoxic effects. Drug-induced cardiotoxicity and QT prolongation may lead to life-threatening arrhythmias such as torsades de pointes (TdP), a potentially fatal arrhythmic symptom. Here, we evaluated the risk of repurposed pyronaridine or artesunate-mediated cardiac arrhythmias alone and in combination for COVID-19 treatment through in vitro and in silico investigations using the Comprehensive in vitro Proarrhythmia Assay (CiPA) initiative. The potential effects of each drug or in combinations on cardiac action potential (AP) and ion channels were explored using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and Chinese hamster ovary (CHO) cells transiently expressing cardiac ion channels (Nav1.5, Cav1.2, and hERG). We also performed in silico computer simulation using the optimized O'Hara-Rudy human ventricular myocyte model (ORd model) to classify TdP risk. Artesunate and dihydroartemisinin (DHA), the active metabolite of artesunate, are classified as a low risk of inducing TdP based on the torsade metric score (TMS). Moreover, artesunate does not significantly affect the cardiac APs of hiPSC-CMs even at concentrations up to 100 times the maximum serum concentration (Cmax). DHA modestly prolonged at APD90 (10.16%) at 100 times the Cmax. When considering Cmax, pyronaridine, and the combination of both drugs (pyronaridine and artesunate) are classified as having an intermediate risk of inducing TdP. However, when considering the unbound concentration (the free fraction not bound to carrier proteins or other tissues inducing pharmacological activity), both drugs are classified as having a low risk of inducing TdP. In summary, pyronaridine, artesunate, and a combination of both drugs have been confirmed to pose a low proarrhythmogenic risk at therapeutic and supratherapeutic (up to 4 times) free Cmax. Additionally, the CiPA initiative may be suitable for regulatory use and provide novel insights for evaluating drug-induced cardiotoxicity.
Collapse
Affiliation(s)
- Seung-Hyun Yoon
- R&D Center for Advanced Pharmaceuticals and Evaluation, Korea Institute of Toxicology, Daejeon, Republic of Korea
- College of Veterinary Medicine, Research Institute of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Hyun-Lee Lee
- R&D Center for Advanced Pharmaceuticals and Evaluation, Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Da Un Jeong
- Intelligent Human Twin Research Center, Electronics and Telecommunications Research Institute, Daejeon, Republic of Korea
| | - Ki Moo Lim
- Department of IT Convergence Engineering, Kumoh National Institute of Technology, Gumi, Republic of Korea
| | - Seong-Jun Park
- College of Veterinary Medicine, Research Institute of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Ki-Suk Kim
- R&D Center for Advanced Pharmaceuticals and Evaluation, Korea Institute of Toxicology, Daejeon, Republic of Korea
| |
Collapse
|
3
|
Reisqs JB, Moreau A, Sleiman Y, Boutjdir M, Richard S, Chevalier P. Arrhythmogenic cardiomyopathy as a myogenic disease: highlights from cardiomyocytes derived from human induced pluripotent stem cells. Front Physiol 2023; 14:1191965. [PMID: 37250123 PMCID: PMC10210147 DOI: 10.3389/fphys.2023.1191965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited cardiomyopathy characterized by the replacement of myocardium by fibro-fatty infiltration and cardiomyocyte loss. ACM predisposes to a high risk for ventricular arrhythmias. ACM has initially been defined as a desmosomal disease because most of the known variants causing the disease concern genes encoding desmosomal proteins. Studying this pathology is complex, in particular because human samples are rare and, when available, reflect the most advanced stages of the disease. Usual cellular and animal models cannot reproduce all the hallmarks of human pathology. In the last decade, human-induced pluripotent stem cells (hiPSC) have been proposed as an innovative human cellular model. The differentiation of hiPSCs into cardiomyocytes (hiPSC-CM) is now well-controlled and widely used in many laboratories. This hiPSC-CM model recapitulates critical features of the pathology and enables a cardiomyocyte-centered comprehensive approach to the disease and the screening of anti-arrhythmic drugs (AAD) prescribed sometimes empirically to the patient. In this regard, this model provides unique opportunities to explore and develop new therapeutic approaches. The use of hiPSC-CMs will undoubtedly help the development of precision medicine to better cure patients suffering from ACM. This review aims to summarize the recent advances allowing the use of hiPSCs in the ACM context.
Collapse
Affiliation(s)
- J. B. Reisqs
- Cardiovascular Research Program, VA New York Harbor Healthcare System, Brooklyn, NY, United States
| | - A. Moreau
- Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, PhyMedExp, Montpellier, France
| | - Y. Sleiman
- Cardiovascular Research Program, VA New York Harbor Healthcare System, Brooklyn, NY, United States
| | - M. Boutjdir
- Cardiovascular Research Program, VA New York Harbor Healthcare System, Brooklyn, NY, United States
- Department of Medicine, Cell Biology and Pharmacology, State University of New York Downstate Health Sciences University, NY, United States
- Department of Medicine, New York University School of Medicine, NY, United States
| | - S. Richard
- Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, PhyMedExp, Montpellier, France
| | - P. Chevalier
- Neuromyogene Institute, Claude Bernard University, Lyon 1, Villeurbanne, France
- Service de Rythmologie, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
4
|
Reisqs JB, Moreau A, Sleiman Y, Charrabi A, Delinière A, Bessière F, Gardey K, Richard S, Chevalier P. Spironolactone as a Potential New Treatment to Prevent Arrhythmias in Arrhythmogenic Cardiomyopathy Cell Model. J Pers Med 2023; 13:jpm13020335. [PMID: 36836569 PMCID: PMC9960914 DOI: 10.3390/jpm13020335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/03/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a rare genetic disease associated with ventricular arrhythmias in patients. The occurrence of these arrhythmias is due to direct electrophysiological remodeling of the cardiomyocytes, namely a reduction in the action potential duration (APD) and a disturbance of Ca2+ homeostasis. Interestingly, spironolactone (SP), a mineralocorticoid receptor antagonist, is known to block K+ channels and may reduce arrhythmias. Here, we assess the direct effect of SP and its metabolite canrenoic acid (CA) in cardiomyocytes derived from human-induced pluripotent stem cells (hiPSC-CMs) of a patient bearing a missense mutation (c.394C>T) in the DSC2 gene coding for desmocollin 2 and for the amino acid replacement of arginine by cysteine at position 132 (R132C). SP and CA corrected the APD in the muted cells (vs. the control) in linking to a normalization of the hERG and KCNQ1 K+ channel currents. In addition, SP and CA had a direct cellular effect on Ca2+ homeostasis. They reduced the amplitude and aberrant Ca2+ events. In conclusion, we show the direct beneficial effects of SP on the AP and Ca2+ homeostasis of DSC2-specific hiPSC-CMs. These results provide a rationale for a new therapeutical approach to tackle mechanical and electrical burdens in patients suffering from ACM.
Collapse
Affiliation(s)
- Jean-Baptiste Reisqs
- Neuromyogene Institute, Claude Bernard University, Lyon 1, 69008 Villeurbanne, France
- PhyMedExp, INSERM, University of Montpellier, CNRS, 34000 Montpellier, France
| | - Adrien Moreau
- PhyMedExp, INSERM, University of Montpellier, CNRS, 34000 Montpellier, France
| | - Yvonne Sleiman
- PhyMedExp, INSERM, University of Montpellier, CNRS, 34000 Montpellier, France
| | - Azzouz Charrabi
- PhyMedExp, INSERM, University of Montpellier, CNRS, 34000 Montpellier, France
| | | | - Francis Bessière
- Service de Rythmologie, Hospices Civils de Lyon, 69500 Lyon, France
| | - Kevin Gardey
- Service de Rythmologie, Hospices Civils de Lyon, 69500 Lyon, France
| | - Sylvain Richard
- PhyMedExp, INSERM, University of Montpellier, CNRS, 34000 Montpellier, France
| | - Philippe Chevalier
- Neuromyogene Institute, Claude Bernard University, Lyon 1, 69008 Villeurbanne, France
- Service de Rythmologie, Hospices Civils de Lyon, 69500 Lyon, France
- Correspondence:
| |
Collapse
|
5
|
Malihi G, Nikoui V, Elson EL. A review on qualifications and cost effectiveness of induced pluripotent stem cells (IPSCs)-induced cardiomyocytes in drug screening tests. Arch Physiol Biochem 2023; 129:131-142. [PMID: 32783745 DOI: 10.1080/13813455.2020.1802600] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Human induced pluripotent stem cells (hIPSCs) have initiated a higher degree of successes in disease modelling, preclinical evaluation of drug therapy and pharmaco-toxicological testing. Since the discovery of iPSCs in 2006, many advanced techniques have been introduced to differentiate iPSCs to cardiomyocytes, which have been progressively improved. The disease models from iPSC-induced cardiomyocytes (iPSC-CM) have been successfully helping to study a variety of cardiac diseases such as long QT syndrome, drug-induced long QT, different cardiomyopathies related to mutations in mitochondria or desmosomal proteins and other rare genetic diseases. IPSC-CMs have also been used to screen the role of chemicals in cardiovascular drug discovery and individualisation of drug dosages. In this review, the quality of current procedures for characterisation and maturation of iPSC-CM lines will be discussed. Also, we will focus on time efficiency and cost of standard differentiation methods after reprogramming.
Collapse
Affiliation(s)
| | - Vahid Nikoui
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Elliot L Elson
- Department of Biochemistry and Molecular Biophysics, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
6
|
Lee J, Gänswein T, Ulusan H, Emmenegger V, Saguner AM, Duru F, Hierlemann A. Repeated and On-Demand Intracellular Recordings of Cardiomyocytes Derived from Human-Induced Pluripotent Stem Cells. ACS Sens 2022; 7:3181-3191. [PMID: 36166837 PMCID: PMC7613763 DOI: 10.1021/acssensors.2c01678] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Pharmaceutical compounds may have cardiotoxic properties, triggering potentially life-threatening arrhythmias. To investigate proarrhythmic effects of drugs, the patch clamp technique has been used as the gold standard for characterizing the electrophysiology of cardiomyocytes in vitro. However, the applicability of this technology for drug screening is limited, as it is complex to use and features low throughput. Recent studies have demonstrated that 3D-nanostructured electrodes enable to obtain intracellular signals from many cardiomyocytes in parallel; however, the tedious electrode fabrication and limited measurement duration still remain major issues for cardiotoxicity testing. Here, we demonstrate how porous Pt-black electrodes, arranged in high-density microelectrode arrays, can be used to record intracellular-like signals of cardiomyocytes at large scale repeatedly over an extended period of time. The developed technique, which yields highly parallelized electroporations using stimulation voltages around 1 V peak-to-peak amplitude, enabled intracellular-like recordings at high success rates without causing significant alteration in key electrophysiological features. In a proof-of-concept study, we investigated electrophysiological modulations induced by two clinically applied drugs, nifedipine and quinidine. As the obtained results were in good agreement with previously published data, we are confident that the developed technique has the potential to be routinely used in in vitro platforms for cardiotoxicity screening.
Collapse
Affiliation(s)
- Jihyun Lee
- Corresponding Authors Jihyun Lee — Bio Engineering Laboratory, ETH Zurich, 4058 Basel, Switzerland; ® Phone: +41 (0)61 387 31 28; jihyun.lee@ bsse.ethz.ch; Andreas Hierlemann — Bio Engineering Laboratory, ETH Zurich, 4058 Basel, Switzerland; Phone: +41 (0)61 387 31 50;
| | | | | | | | | | | | - Andreas Hierlemann
- Corresponding Authors Jihyun Lee — Bio Engineering Laboratory, ETH Zurich, 4058 Basel, Switzerland; ® Phone: +41 (0)61 387 31 28; jihyun.lee@ bsse.ethz.ch; Andreas Hierlemann — Bio Engineering Laboratory, ETH Zurich, 4058 Basel, Switzerland; Phone: +41 (0)61 387 31 50;
| |
Collapse
|
7
|
Faulkner-Jones A, Zamora V, Hortigon-Vinagre MP, Wang W, Ardron M, Smith GL, Shu W. A Bioprinted Heart-on-a-Chip with Human Pluripotent Stem Cell-Derived Cardiomyocytes for Drug Evaluation. Bioengineering (Basel) 2022; 9:bioengineering9010032. [PMID: 35049741 PMCID: PMC8773426 DOI: 10.3390/bioengineering9010032] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/06/2022] [Accepted: 01/10/2022] [Indexed: 12/12/2022] Open
Abstract
In this work, we show that valve-based bioprinting induces no measurable detrimental effects on human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). The aim of the current study was three-fold: first, to assess the response of hiPSC-CMs to several hydrogel formulations by measuring electrophysiological function; second, to customise a new microvalve-based cell printing mechanism in order to deliver hiPSC-CMs suspensions, and third, to compare the traditional manual pipetting cell-culture method and cardiomyocytes dispensed with the bioprinter. To achieve the first and third objectives, iCell2 (Cellular Dynamics International) hiPSC-CMs were used. The effects of well-known drugs were tested on iCell2 cultured by manual pipetting and bioprinting. Despite the results showing that hydrogels and their cross-linkers significantly reduced the electrophysiological performance of the cells compared with those cultured on fibronectin, the bio-ink droplets containing a liquid suspension of live cardiomyocytes proved to be an alternative to standard manual handling and could reduce the number of cells required for drug testing, with no significant differences in drug-sensitivity between both approaches. These results provide a basis for the development of a novel bioprinter with nanolitre resolution to decrease the required number of cells and to automate the cell plating process.
Collapse
Affiliation(s)
- Alan Faulkner-Jones
- Institute of Mechanical, Process and Energy Engineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, UK; (A.F.-J.); (W.W.)
| | - Victor Zamora
- Departamento de Ingeniería Mecánica, Energética y de los Materiales, Universidad de Extremadura, 06006 Badajoz, Spain;
| | - Maria P. Hortigon-Vinagre
- Departamento de Bioquímica y Biología Molecular y Genética, Universidad de Extremadura, Facultad de Ciencias, 06006 Badajoz, Spain
- Correspondence: ; Tel.: +34-924-289-300 (ext. 89053)
| | - Wenxing Wang
- Institute of Mechanical, Process and Energy Engineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, UK; (A.F.-J.); (W.W.)
| | - Marcus Ardron
- Renishaw PLC, Research Avenue North, Edinburgh EH14 4AP, UK;
| | - Godfrey L. Smith
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8QQ, UK;
- Clyde Biosciences, Glasgow G12 8QQ, UK
| | - Wenmiao Shu
- Department of Biomedical Engineering, Faculty of Engineering, University of Strathclyde, Glasgow G4 0NW, UK;
| |
Collapse
|
8
|
Laco F, Lam ATL, Woo TL, Tong G, Ho V, Soong PL, Grishina E, Lin KH, Reuveny S, Oh SKW. Selection of human induced pluripotent stem cells lines optimization of cardiomyocytes differentiation in an integrated suspension microcarrier bioreactor. Stem Cell Res Ther 2020; 11:118. [PMID: 32183888 PMCID: PMC7076930 DOI: 10.1186/s13287-020-01618-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/11/2020] [Accepted: 02/24/2020] [Indexed: 01/13/2023] Open
Abstract
Background The production of large quantities of cardiomyocyte is essential for the needs of cellular therapies. This study describes the selection of a human-induced pluripotent cell (hiPSC) line suitable for production of cardiomyocytes in a fully integrated bioprocess of stem cell expansion and differentiation in microcarrier stirred tank reactor. Methods Five hiPSC lines were evaluated first for their cardiac differentiation efficiency in monolayer cultures followed by their expansion and differentiation compatibility in microcarrier (MC) cultures under continuous stirring conditions. Results Three cell lines were highly cardiogenic but only one (FR202) of them was successfully expanded on continuous stirring MC cultures. FR202 was thus selected for cardiac differentiation in a 22-day integrated bioprocess under continuous stirring in a stirred tank bioreactor. In summary, we integrated a MC-based hiPSC expansion (phase 1), CHIR99021-induced cardiomyocyte differentiation step (phase 2), purification using the lactate-based treatment (phase 3) and cell recovery step (phase 4) into one process in one bioreactor, under restricted oxygen control (< 30% DO) and continuous stirring with periodic batch-type media exchanges. High density of undifferentiated hiPSC (2 ± 0.4 × 106 cells/mL) was achieved in the expansion phase. By controlling the stirring speed and DO levels in the bioreactor cultures, 7.36 ± 1.2 × 106 cells/mL cardiomyocytes with > 80% Troponin T were generated in the CHIR99021-induced differentiation phase. By adding lactate in glucose-free purification media, the purity of cardiomyocytes was enhanced (> 90% Troponin T), with minor cell loss as indicated by the increase in sub-G1 phase and the decrease of aggregate sizes. Lastly, we found that the recovery period is important for generating purer and functional cardiomyocytes (> 96% Troponin T). Three independent runs in a 300-ml working volume confirmed the robustness of this process. Conclusion A streamlined and controllable platform for large quantity manufacturing of pure functional atrial, ventricular and nodal cardiomyocytes on MCs in conventional-type stirred tank bioreactors was established, which can be further scaled up and translated to a good manufacturing practice-compliant production process, to fulfill the quantity requirements of the cellular therapeutic industry. Supplementary information The online version of this article (10.1186/s13287-020-01618-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Filip Laco
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore, 138668, Singapore
| | - Alan Tin-Lun Lam
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore, 138668, Singapore.
| | - Tsung-Liang Woo
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore, 138668, Singapore
| | - Gerine Tong
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore, 138668, Singapore
| | - Valerie Ho
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore, 138668, Singapore
| | - Poh-Loong Soong
- Ternion Biosciences, National Heart Centre of Singapore, Singapore, Singapore
| | - Elina Grishina
- Ternion Biosciences, National Heart Centre of Singapore, Singapore, Singapore
| | - Kun-Han Lin
- Ternion Biosciences, National Heart Centre of Singapore, Singapore, Singapore
| | - Shaul Reuveny
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore, 138668, Singapore
| | - Steve Kah-Weng Oh
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore, 138668, Singapore.
| |
Collapse
|
9
|
Lin YH, Ni XB, Zhang JW, Ou CW, He XQ, Dai WJ, Chen XM, Chen MS. Effect of puerarin on action potential and sodium channel activation in human hypertrophic cardiomyocytes. Biosci Rep 2020; 40:222020. [PMID: 32003781 PMCID: PMC7024842 DOI: 10.1042/bsr20193369] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 01/13/2020] [Accepted: 01/28/2020] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE To study the effect of puerarin on electrophysiology using a hypertrophic cardiomyocyte (HC) model. MATERIALS AND METHODS Human urine epithelial cells were used to generate the HC model (hiPSC-CM). Cardiomyocyte hypertrophy was induced by applying 10 nM endothelin-1 (ET-1). Effects of puerarin pre-treatment (PPr) and post-treatment (PPo) on action potential, sodium current (INa) activation and inactivation, and recovery following INa inactivation were tested using patch clamp electrophysiology. RESULTS Depolarization to repolarization 50% time (APD50) and repolarization 30% time (APD30) were significantly prolonged in the PPo and PPr groups compared with the controls. However, there were no significant differences in the action potential depolarization amplitude (APA) or the maximum depolarization velocity (Vmax) in phase 0. The PPr group had a slightly shortened APD90, and an extended APD50 and APD30, but did not exhibit any significant changes in stage A of APA and Vmax. The PPo group did not exhibit any significant changes in INa, while 12 h of PPr improved INa. However, puerarin did not significantly affect the activation, inactivation, or recovery of the sodium channel. CONCLUSIONS Cardiomyocyte hypertrophy significantly decreased the Vmax of the action potential and the peak density of INa. PPr inhibited the decrease in Vmax and increased the peak density of INa. Thus, puerarin could be used to stabilize the electrophysiological properties of hypertrophic cardiomyocytes and reduce arrhythmias.
Collapse
Affiliation(s)
- Yu-hui Lin
- Department of Cardiovascular Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiao-Bin Ni
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Jian-wu Zhang
- Department of Cardiovascular Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Cai-wen Ou
- Department of Cardiovascular Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiao-qing He
- Department of Cardiovascular Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wen-jun Dai
- Department of Cardiovascular Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xi-ming Chen
- Department of Cardiovascular Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Correspondence: Xi-ming Chen () or Min-sheng Chen ()
| | - Min-sheng Chen
- Department of Cardiovascular Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Correspondence: Xi-ming Chen () or Min-sheng Chen ()
| |
Collapse
|
10
|
Gasiūnienė M, Valatkaitė E, Navakauskienė R. Long-term cultivation of human amniotic fluid stem cells: The impact on proliferative capacity and differentiation potential. J Cell Biochem 2020; 121:3491-3501. [PMID: 31898359 DOI: 10.1002/jcb.29623] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 12/11/2019] [Indexed: 12/14/2022]
Abstract
Human amniotic fluid mesenchymal stem cells (AF-MSCs) are a valuable, easily obtainable alternative source of SCs for regenerative medicine. Usually, amounts of cells required for the translational purposes are large thus the goal of this study was to assess the potency of AF-MSCs to proliferate and differentiate during long-term cultivation in vitro. AF-MSCs were isolated from amniotic fluid of healthy women in the second trimester of pregnancy and cultivated in vitro. AF-MSCs were cultivated up to 42 passages and they still maintained pluripotency genes, such as OCT4, SOX2, and NANOG, expression at a similar level as in the initial passages as determined by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Fluorescence-activated cell sorting analysis demonstrated that the cell surface markers CD34 (negative), CD44, and CD105 (positive) expression was also stable, only the expression of SCs marker CD90 decreased during the cultivation. The morphology of AF-MSCs changed over passage, acridine orange/ethidium bromide staining revealed that more cells entered into apoptosis and the first signs of aging were detected only at late passages (later than p33) using SA-β-gal assay. Concomitantly, the differentiation potential towards cardiomyogenic lineage, induced with DNA methyltransferases inhibitors decitabine, zebularine, and RG108, was impaired when comparing AF-MSCs at p31/33 with p6. The expression of cardiomyocytes genes MYH6, TNNT2, DES together with ion channels genes of the heart (sodium, calcium, and potassium) decreased in p31/33 induced AF-MSCs. AF-MSCs have a great proliferative capacity and maintain most of the characteristics up to 33 passages; however, the cardiomyogenic differentiation capacity decreases to a certain extent during the long-term cultivation. These results provide useful insights for the potential use of AF-MSCs for biobanking and broad applications requiring high yield of cells or repeated infusions. Hence, it is vital to take into account the passage number of AF-MSCs, cultivated in culture, when utilizing them in vivo or in clinical experiments.
Collapse
Affiliation(s)
- Monika Gasiūnienė
- Department of Molecular Cell Biology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Elvina Valatkaitė
- Department of Molecular Cell Biology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Rūta Navakauskienė
- Department of Molecular Cell Biology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
11
|
Gasiūnienė M, Petkus G, Matuzevičius D, Navakauskas D, Navakauskienė R. Angiotensin II and TGF- β1 Induce Alterations in Human Amniotic Fluid-Derived Mesenchymal Stem Cells Leading to Cardiomyogenic Differentiation Initiation. Int J Stem Cells 2019; 12:251-264. [PMID: 31023001 PMCID: PMC6657950 DOI: 10.15283/ijsc18126] [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: 12/06/2018] [Revised: 02/07/2019] [Accepted: 02/11/2019] [Indexed: 12/17/2022] Open
Abstract
Background and Objectives Human amniotic fluid-derived mesenchymal stem cells (AF-MSCs) may be a valuable source for cardiovascular tissue engineering and cell therapy. The aim of this study is to verify angiotensin II and transforming growth factor-beta 1 (TGF-β1) as potential cardiomyogenic differentiation inducers of AF-MSCs. Methods and Results AF-MSCs were obtained from amniocentesis samples from second-trimester pregnant women, isolated and characterized by the expression of cell surface markers (CD44, CD90, CD105 positive; CD34 negative) and pluripotency genes (OCT4, SOX2, NANOG, REX1). Cardiomyogenic differentiation was induced using different concentrations of angiotensin II and TGF-β1. Successful initiation of differentiation was confirmed by alterations in cell morphology, upregulation of cardiac genes-markers NKX2-5, TBX5, GATA4, MYH6, TNNT2, DES and main cardiac ion channels genes (sodium, calcium, potassium) as determined by RT-qPCR. Western blot and immunofluorescence analysis revealed the increased expression of Connexin43, the main component of gap junctions, and Nkx2.5, the early cardiac transcription factor. Induced AF-MSCs switched their phenotype towards more energetic and started utilizing oxidative phosphorylation more than glycolysis for energy production as assessed using Agilent Seahorse XF analyzer. The immune analysis of chromatin-modifying enzymes DNMT1, HDAC1/2 and Polycomb repressive complex 1 and 2 (PRC1/2) proteins BMI1, EZH2 and SUZ12 as well as of modified histones H3 and H4 indicated global chromatin remodeling during the induced differentiation. Conclusions Angiotensin II and TGF-β1 are efficient cardiomyogenic inducers of human AF-MSCs; they initiate alterations at the gene and protein expression, metabolic and epigenetic levels in stem cells leading towards cardiomyocyte- like phenotype formation.
Collapse
Affiliation(s)
- Monika Gasiūnienė
- Department of Molecular Cell Biology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Gintautas Petkus
- Department of Molecular Cell Biology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Dalius Matuzevičius
- Electronic Systems Department, Electronics Faculty, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | - Dalius Navakauskas
- Electronic Systems Department, Electronics Faculty, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | - Rūta Navakauskienė
- Department of Molecular Cell Biology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
12
|
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: 24] [Impact Index Per Article: 4.0] [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
|
13
|
Exposure-response analysis of drug-induced QT interval prolongation in telemetered monkeys for translational prediction to human. J Pharmacol Toxicol Methods 2019; 99:106606. [PMID: 31255745 DOI: 10.1016/j.vascn.2019.106606] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/24/2019] [Accepted: 06/24/2019] [Indexed: 01/08/2023]
Abstract
INTRODUCTION The preclinical in vivo assay for QT prolongation is critical for predicting torsadogenic risk, but still difficult to extrapolate to humans. This study ran preclinical tests in cynomolgus monkeys on seven QT reference drugs containing the drugs used in the IQ-CSRC clinical trial and applied exposure-response (ER) analysis to the data to investigate the potential for translational information on the QT effect. METHODS In each of six participating facilities in the J-ICET project, telemetered monkeys were monitored for 24 h following administration of vehicle or 3 doses of test drugs, and pharmacokinetic profiles at the same doses were evaluated separately. An individual rate-corrected QT interval (QTca) was derived and the vehicle-adjusted change in QTca from baseline (∆∆QTca) was calculated. Then the relationship of concentration to QT effect was evaluated by ER analysis. RESULTS For QT-positive drugs in the IQ-CSRC study (dofetilide, dolasetron, moxifloxacin, ondansetron, and quinine) and levofloxacin, the slope of the total concentration-QTca effect was significantly positive, and the QT-prolonging effect, taken as the upper bound of the confidence interval for predicted ∆∆QTca, was confirmed to exceed 10 ms. The ER slope of the negative drug levocetirizine was not significantly positive and the QTca effect was below 10 ms at observed peak exposure. DISCUSSION Preclinical QT assessment in cynomolgus monkeys combined with ER analysis could identify the small QT effect induced by several QT drugs consistently with the outcomes in humans. Thus, the ER method should be regarded as useful for translational prediction of QT effects in humans.
Collapse
|
14
|
Zhang XH, Šarić T, Mehrjardi NZ, Hamad S, Morad M. Acid-Sensitive Ion Channels Are Expressed in Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Stem Cells Dev 2019; 28:920-932. [PMID: 31119982 DOI: 10.1089/scd.2018.0234] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are potential sources for cardiac regeneration and drug development. hiPSC-CMs express all the cardiac ion channels and the unique cardiac Ca2+-signaling phenotype. In this study, we tested for expression of acid sensing ion channels (ASICs) in spontaneously beating cardiomyocytes derived from three different hiPSC lines (IMR-90, iPSC-K3, and Ukki011-A). Rapid application of solutions buffered at pH 6.7, 6.0, or 5.0 triggered rapidly activating and slowly inactivating voltage-independent inward current that reversed at voltages positive to ENa, was suppressed by 5 μM amiloride and withdrawal of [Na+]o, like neuronal ASIC currents. ASIC currents were expressed at much lower percentages and densities in undifferentiated hiPSC and in dermal fibroblasts. ASIC1 mRNA and protein were measured in first 60 days but decreased in 100 days postdifferentiation hiPSC cultures. Hyperacidification (pH 5 and 6) also triggered large Ca2+ transients in intact hiPSC-CMs that were neither ruthenium red nor amiloride-sensitive, but were absent in whole cell-clamped hiPSC-CMs. Neither ASIC1 current nor its protein was detected in rat adult cardiomyocytes, but hyperacidification did activate smaller and slowly activating currents with drug sensitivity similar to TRPV channels. Considering ASIC expression in developing but not adult myocardium, a role in heart development is likely.
Collapse
Affiliation(s)
- Xiao-Hua Zhang
- 1Cardiac Signaling Center of University of South Carolina, Medical University of South Carolina, Clemson University, Charleston, South Carolina
| | - Tomo Šarić
- 2Medical Faculty, Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Cologne, Germany
| | - Narges Zare Mehrjardi
- 2Medical Faculty, Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Cologne, Germany
| | - Sarkawt Hamad
- 2Medical Faculty, Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Cologne, Germany
| | - Martin Morad
- 1Cardiac Signaling Center of University of South Carolina, Medical University of South Carolina, Clemson University, Charleston, South Carolina
| |
Collapse
|
15
|
Gasiūnienė M, Zubova A, Utkus A, Navakauskienė R. Epigenetic and metabolic alterations in human amniotic fluid stem cells induced to cardiomyogenic differentiation by DNA methyltransferases and p53 inhibitors. J Cell Biochem 2019; 120:8129-8143. [PMID: 30485506 DOI: 10.1002/jcb.28092] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 10/29/2018] [Indexed: 01/24/2023]
Abstract
Human amniotic fluid-derived mesenchymal stem cells (AF-MSCs) may be a valuable source for cell therapy and regenerative medicine. In this study, the potential of DNA methyltransferases (DNMT) inhibitors Decitabine, Zebularine, RG108 alone or combined with Zebularine and p53 inhibitor Pifithrin-α to induce cardiomyogenic differentiation of AF-MSCs was investigated. Differentiation into cardiomyocyte-like cells initiation was indicated with all agents by changes in the cell phenotype, upregulation of the relative expression of the main cardiac genes (NKX2-5, TNNT2, MYH6, and DES) as well as of cardiac ion channels genes (sodium, calcium, and potassium) as determined by reverse-transcription quantitative polymerase chain reaction and the increase in Connexin43 levels as detected from Western blot and immunofluorescence data. Cellular energetics and mitochondrial function in induced cells were assessed using Seahorse analyzer and revealed the initiation of AF-MSCs metabolic transformation into cardiomyocyte-like cells. All used inducers were nontoxic to AF-MSCs, arrested cell cycle at the G0/G1 phase, and upregulated p53 and p21 expression. The relative expression of miR-34a and miR-145 that are related to cell cycle regulation was also observed. Furthermore, the evaluated levels of chromatin remodeling proteins enhancer of zeste homolog 2, suppressor of zeste 12 protein homolog, DNMT1, histone deacetylase 1 (HDAC1), HDAC2, and heterochromatin protein 1α, as well as the rate of activating histone modifications, exhibited rearrangements of chromatin after the induction of cardiomyogenic differentiation. In conclusion, we demonstrated that all explored DNMT and p53 inhibitors initiated cardiomyogenesis-related alterations in AF-MSCs through rather similar mechanisms but to a different extent providing useful insights for the future research and potential applications of AF-MSCs.
Collapse
Affiliation(s)
- Monika Gasiūnienė
- Department of Molecular Cell Biology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Anastasija Zubova
- Department of Molecular Cell Biology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Algirdas Utkus
- Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Rūta Navakauskienė
- Department of Molecular Cell Biology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
16
|
Gasiūnienė M, Zentelytė A, Wojtas B, Baronaitė S, Krasovskaja N, Savickienė J, Gielniewski B, Kaminska B, Utkus A, Navakauskienė R. DNA methyltransferases inhibitors effectively induce gene expression changes suggestive of cardiomyogenic differentiation of human amniotic fluid-derived mesenchymal stem cells via chromatin remodeling. J Tissue Eng Regen Med 2019; 13:469-481. [PMID: 30637987 DOI: 10.1002/term.2800] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 10/31/2018] [Accepted: 01/09/2019] [Indexed: 12/17/2022]
Abstract
Human amniotic fluid-derived mesenchymal stem cells (AF-MSCs) are a new potential stem cell source for cell therapy and regenerative medicine. These are fetal mesenchymal stem cells with multilineage differentiation potential found in amniotic fluid. The aim of the present study was to evaluate in vitro differentiation initiation of AF-MSCs into cardiac progenitors upon application of inhibitors of DNA methyltransferases (DNMT), such as Decitabine (DEC; 5-aza-2'-deoxycytidine) and Zebularine (ZEB). We assessed epigenetic changes and explored patterns of genes, enriched in association with hyperacetylated H4 after induced differentiation. Upregulation of cardiomyogenesis-related genes (TNNT2, MYH6, ACTN2, and DES) and cardiac ion channels genes, downregulation of pluripotency genes markers as well as increase in Connexin43 expression indicated cardiomyogenic commitment. Evaluation of global epigenetic changes showed that levels of chromatin modifying enzymes, such as Polycomb repressive complex 2 proteins (EZH2, SUZ12), DNMT1, histone deacetylases 1 and 2 were reduced to the similar extent by both differentiation agents. Levels of specific histone marks keeping active state of chromatin (H3K4me3, H3K9Ac, and H4hyperAc) increased and marks of repressed chromatin state (H3K27me3 and H3K9me3) decreased after DEC or ZEB treatment. Chip-Seq analysis after chromatin immunoprecipitation with H4hyperAc demonstrated enrichment of around 100 functionally annotated genes, related to chromatin reorganization and cardiomyogenesis and confirmed relation between H4 hyperacetylation and gene expression. Our results demonstrate that both DEC and ZEB can be potentially used as cardiomyogenic differentiation inducers in AF-MSCs, and they cause various genetic and epigenetic changes resulting in global chromatin remodeling.
Collapse
Affiliation(s)
- Monika Gasiūnienė
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Aistė Zentelytė
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Bartosz Wojtas
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Sandra Baronaitė
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | | | - Jūratė Savickienė
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Bartlomiej Gielniewski
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Bozena Kaminska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Algirdas Utkus
- Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Rūta Navakauskienė
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
17
|
Isobe T, Honda M, Komatsu R, Tabo M. Cardiac safety assessment with motion field imaging analysis of human iPS cell-derived cardiomyocytes is improved by an integrated evaluation with cardiac ion channel profiling. J Toxicol Sci 2019; 44:859-870. [DOI: 10.2131/jts.44.859] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Takehito Isobe
- Research Division, Chugai Pharmaceutical Co., Ltd
- Translational Research Division, Chugai Pharmaceutical Co., Ltd
| | - Masaki Honda
- Research Division, Chugai Pharmaceutical Co., Ltd
| | | | | |
Collapse
|
18
|
Hypoxic cardiac fibroblasts from failing human hearts decrease cardiomyocyte beating frequency in an ALOX15 dependent manner. PLoS One 2018; 13:e0202693. [PMID: 30138423 PMCID: PMC6107211 DOI: 10.1371/journal.pone.0202693] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 08/07/2018] [Indexed: 11/19/2022] Open
Abstract
A common denominator for patients with heart failure is the correlation between elevated serum levels of proinflammatory cytokines and adverse clinical outcomes. Furthermore, lipoxygenase-induced inflammation is reportedly involved in the pathology of heart failure. Cardiac fibroblasts, which are abundant in cardiac tissue, are known to be activated by inflammation. We previously showed high expression of the lipoxygenase arachidonate 15 lipoxygenase (ALOX15), which catalyzes the conversion of arachidonic acid to 15-hydroxy eicosatetraenoic acid (15-HETE), in ischemic cardiac tissue. The exact roles of ALOX15 and 15-HETE in the pathogenesis of heart failure are however unknown. Biopsies were collected from all chambers of explanted failing human hearts from heart transplantation patients, as well as from the left ventricles from organ donors not suffering from chronic heart failure. Biopsies from the left ventricles underwent quantitative immunohistochemical analysis for ALOX15/B. Gene expression of ALOX enzymes, as well as 15-HETE levels, were examined in cardiac fibroblasts which had been cultured in either hypoxic or normoxic conditions after isolation from failing hearts. After the addition of fibroblast supernatants to human induced pluripotent stem cell-derived cardiomyocytes, intracellular calcium concentrations were measured to examine the effect of paracrine signaling on cardiomyocyte beating frequency. While ALOX15 and ALOX15B were expressed throughout failing hearts as well as in hearts from organ donors, ALOX15 was expressed at significantly higher levels in donor hearts. Hypoxia resulted in a significant increase in gene and protein expression of ALOX15 and ALOX15B in fibroblasts isolated from the different chambers of failing hearts. Finally, preconditioned medium from hypoxic fibroblasts decreased the beating frequency of human cardiomyocytes derived from induced pluripotent stem cells in an ALOX15-dependent manner. In summary, our results demonstrate that ALOX15/B signaling by hypoxic cardiac fibroblasts may play an important role in ischemic cardiomyopathy, by decreasing cardiomyocyte beating frequency.
Collapse
|
19
|
Gorabi AM, Hajighasemi S, Tafti HA, Atashi A, Soleimani M, Aghdami N, Saeid AK, Khori V, Panahi Y, Sahebkar A. TBX18 transcription factor overexpression in human-induced pluripotent stem cells increases their differentiation into pacemaker-like cells. J Cell Physiol 2018; 234:1534-1546. [PMID: 30078203 DOI: 10.1002/jcp.27018] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 06/26/2018] [Indexed: 12/24/2022]
Abstract
BACKGROUND The discovery of gene- and cell-based strategies has opened a new area to investigate novel approaches for the treatment of many conditions caused by cardiac cell failure. The TBX18 (T-box 18) transcription factor is considered as a prominent factor in the sinoatrial node (SAN) formation during the embryonic development. In this in vitro study, the effect of TBX18 gene expression on human-induced pluripotent-stem-cell-derived cardiomyocytes (hiPS-CMs) to induce pacemaker-like cells was examined. METHODS The human-dermal-fibroblast-derived iPSCs were transfected using chemical, physical, and Lentiviral methods of TBX18 gene delivery during differentiation into cardiomyocytes (CMs). After the differentiation process through small-molecule-based temporal modulation of the Wnt signaling pathway, the hiPSC-CMs were analyzed using the real-time polymerase chain reaction, immunocytochemistry, immunofluorescence, whole-cell patch-clamp recording, and western blotting to investigate the accuracy of differentiation and identify the effect exerted by TBX18. RESULTS The hiPS-CMs showed spontaneous beating and expressed specific markers of cardiac cells. The lentiviral-mediated TBX18 delivery was the most efficient method for transfection. The results showed the increment in Connexin 43 expression among untransfected hiPS-CMs, whereas this protein was significantly downregulated followed by TBX18 overexpression. TBX18-hiPSCMs were detected with pacemaker cell features. CONCLUSIONS It was demonstrated that the TBX18 gene is able to conduct hiPSCs to differentiate into pacemaker-like cells. The TBX18 gene delivery seems to have the potential for the development of biological pacemakers; however, more investigations are still needed to assess its usefulness to fix arrhythmic conditions with SAN failure basis.
Collapse
Affiliation(s)
- Armita M Gorabi
- Department of Basic and Clinical Research, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeideh Hajighasemi
- Department of Medical Biotechnology, Faculty of Paramedicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Hossein A Tafti
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Atashi
- Stem Cell and Tissue Engineering Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Nasser Aghdami
- Department of Regenerative Biomedicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Tehran, Iran
| | - Ali K Saeid
- Department of Cardiology, Tehran University of Medical Science, Tehran, Iran
| | - Vahid Khori
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Yunes Panahi
- Pharmacotherapy Department, School of Pharmacy, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Tehran, Iran
| |
Collapse
|
20
|
Fermini B, Coyne ST, Coyne KP. Clinical Trials in a Dish: A Perspective on the Coming Revolution in Drug Development. SLAS DISCOVERY 2018; 23:765-776. [PMID: 29862873 PMCID: PMC6104197 DOI: 10.1177/2472555218775028] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The pharmaceutical industry is facing unprecedented challenges as the cost of developing
new drugs has reached unsustainable levels, fueled in large parts by a high attrition rate
in clinical development. Strategies to bridge studies between preclinical testing and
clinical trials are needed to reduce the knowledge gap and allow earlier decisions to be
made on the continuation or discontinuation of further development of drugs. The discovery
and development of human induced pluripotent stem cells (hiPSCs) have opened up new
avenues that support the concept of screening for cell-based safety and toxicity at the
level of a population. This approach, termed “Clinical Trials in a Dish” (CTiD), allows
testing medical therapies for safety or efficacy on cells collected from a representative
sample of human patients, before moving into actual clinical trials. It can be applied to
the development of drugs for specific populations, and it allows predicting not only the
magnitude of effects but also the incidence of patients in a population who will benefit
or be harmed by these drugs. This, in turn, can lead to the selection of safer drugs to
move into clinical development, resulting in a reduction in attrition. The current article
offers a perspective of this new model for “humanized” preclinical drug development.
Collapse
|
21
|
Dinarelli S, Girasole M, Spitalieri P, Talarico RV, Murdocca M, Botta A, Novelli G, Mango R, Sangiuolo F, Longo G. AFM nano-mechanical study of the beating profile of hiPSC-derived cardiomyocytes beating bodies WT and DM1. J Mol Recognit 2018; 31:e2725. [PMID: 29748973 DOI: 10.1002/jmr.2725] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 03/20/2018] [Accepted: 04/13/2018] [Indexed: 12/12/2022]
Abstract
Myotonic Dystrophy type 1 (DM1) is the most common form of muscular dystrophy in adults, characterized by a variety of multisystemic features and associated with cardiac anomalies. Among cardiac phenomena, conduction defects, ventricular arrhythmias, and dilated cardiomyopathy represent the main cause of sudden death in DM1 patients. Patient-specific induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) represent a powerful in vitro model for molecular, biochemical, and physiological studies of disease in the target cells. Here, we used an Atomic Force Microscope (AFM) to measure the beating profiles of a large number of cells, organized in CM clusters (Beating Bodies, BBs), obtained from wild type (WT) and DM1 patients. We monitored the evolution over time of the frequency and intensity of the beating. We determined the variations between different BBs and over various areas of a single BB, caused by morphological and biomechanical variations. We exploited the AFM tip to apply a controlled force over the BBs, to carefully assess the biomechanical reaction of the different cell clusters over time, both in terms of beating frequency and intensity. Our measurements demonstrated differences between the WT and DM1 clusters highlighting, for the DM1 samples, an instability which was not observed in WT cells. We measured differences in the cellular response to the applied mechanical stimulus in terms of beating synchronicity over time and cell tenacity, which are in good agreement with the cellular behavior in vivo. Overall, the combination of hiPSC-CMs with AFM characterization can become a new tool to study the collective movements of cell clusters in different conditions and can be extended to the characterization of the BB response to chemical and pharmacological stimuli.
Collapse
Affiliation(s)
- S Dinarelli
- Institute for the Structure of Matter, CNR, Rome, Italy
| | - M Girasole
- Institute for the Structure of Matter, CNR, Rome, Italy
| | - P Spitalieri
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - R V Talarico
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - M Murdocca
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - A Botta
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - G Novelli
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - R Mango
- Department of Emergency and Critical Care, Polyclinic Tor Vergata, Rome, Italy
| | - F Sangiuolo
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - G Longo
- Institute for the Structure of Matter, CNR, Rome, Italy
| |
Collapse
|
22
|
Yonekura M, Kondoh N, Han C, Toyama Y, Ohba T, Ono K, Itagaki S, Tomita H, Murakami M. Medaka as a model for ECG analysis and the effect of verapamil. J Pharmacol Sci 2018; 137:55-60. [DOI: 10.1016/j.jphs.2018.04.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 02/20/2018] [Accepted: 02/22/2018] [Indexed: 11/17/2022] Open
|
23
|
Spitalieri P, Talarico RV, Caioli S, Murdocca M, Serafino A, Girasole M, Dinarelli S, Longo G, Pucci S, Botta A, Novelli G, Zona C, Mango R, Sangiuolo F. Modelling the pathogenesis of Myotonic Dystrophy type 1 cardiac phenotype through human iPSC-derived cardiomyocytes. J Mol Cell Cardiol 2018; 118:95-109. [PMID: 29551391 DOI: 10.1016/j.yjmcc.2018.03.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/12/2018] [Accepted: 03/14/2018] [Indexed: 12/20/2022]
Abstract
Myotonic Dystrophy type 1 (DM1) is a multisystemic disease, autosomal dominant, caused by a CTG repeat expansion in DMPK gene. We assessed the appropriateness of patient-specific induced pluripotent stem cell-derived cardiomyocytes (CMs) as a model to recapitulate some aspects of the pathogenetic mechanism involving cardiac manifestations in DM1 patients. Once obtained in vitro, CMs have been characterized for their morphology and their functionality. CMs DM1 show intranuclear foci and transcript markers abnormally spliced respect to WT ones, as well as several irregularities in nuclear morphology, probably caused by an unbalanced lamin A/C ratio. Electrophysiological characterization evidences an abnormal profile only in CMs DM1 such that the administration of antiarrythmic drugs to these cells highlights even more the functional defect linked to the disease. Finally, Atomic Force Measurements reveal differences in the biomechanical behaviour of CMs DM1, in terms of frequencies and synchronicity of the beats. Altogether the complex phenotype described in this work, strongly reproduces some aspects of the human DM1 cardiac phenotype. Therefore, the present study provides an in vitro model suggesting novel insights into the mechanisms leading to the development of arrhythmogenesis and dilatative cardiomyopathy to consider when approaching to DM1 patients, especially for the risk assessment of sudden cardiac death (SCD). These data could be also useful in identifying novel biomarkers effective in clinical settings and patient-tailored therapies.
Collapse
Affiliation(s)
- Paola Spitalieri
- Dept of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Rosa V Talarico
- Dept of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | | | - Michela Murdocca
- Dept of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | | | | | | | | | - Sabina Pucci
- Dept of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Annalisa Botta
- Dept of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Giuseppe Novelli
- Dept of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Cristina Zona
- I.R.C.C.S. S. Lucia, Rome, Italy; Dept of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | | | - Federica Sangiuolo
- Dept of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy.
| |
Collapse
|
24
|
Hyun SW, Kim BR, Lin D, Hyun SA, Yoon SS, Seo JW. The effects of gentamicin and penicillin/streptomycin on the electrophysiology of human induced pluripotent stem cell-derived cardiomyocytes in manual patch clamp and multi-electrode array system. J Pharmacol Toxicol Methods 2018; 91:1-6. [DOI: 10.1016/j.vascn.2017.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 11/29/2017] [Accepted: 12/09/2017] [Indexed: 02/06/2023]
|
25
|
Mulder P, de Korte T, Dragicevic E, Kraushaar U, Printemps R, Vlaming MLH, Braam SR, Valentin JP. Predicting cardiac safety using human induced pluripotent stem cell-derived cardiomyocytes combined with multi-electrode array (MEA) technology: A conference report. J Pharmacol Toxicol Methods 2018; 91:36-42. [PMID: 29355722 DOI: 10.1016/j.vascn.2018.01.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/21/2017] [Accepted: 01/10/2018] [Indexed: 12/20/2022]
Abstract
Safety pharmacology studies that evaluate drug candidates for potential cardiovascular liabilities remain a critical component of drug development. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have recently emerged as a new and promising tool for preclinical hazard identification and risk assessment of drugs. Recently, Pluriomics organized its first User Meeting entitled 'Combining Pluricyte® Cardiomyocytes & MEA for Safety Pharmacology applications', consisting of scientific sessions and live demonstrations, which provided the opportunity to discuss the application of hiPSC-CMs (Pluricyte® Cardiomyocytes) in cardiac safety assessment to support early decision making in safety pharmacology. This report summarizes the outline and outcome of this Pluriomics User Meeting, which took place on November 24-25, 2016 in Leiden (The Netherlands). To reflect the content of the communications presented at this meeting we have cited key scientific articles and reviews.
Collapse
Affiliation(s)
- Petra Mulder
- Pluriomics BV, Galileiweg 8, 2333 BD Leiden, The Netherlands
| | - Tessa de Korte
- Pluriomics BV, Galileiweg 8, 2333 BD Leiden, The Netherlands.
| | - Elena Dragicevic
- Nanion Technologies GmbH, Ganghoferstraße 70a, D-80339 Munich, Germany
| | - Udo Kraushaar
- NMI Natural and Medical Sciences Institute, Markwiesenstraße 55, 72770 Reutlingen, Germany
| | | | | | - Stefan R Braam
- Pluriomics BV, Galileiweg 8, 2333 BD Leiden, The Netherlands
| | - Jean-Pierre Valentin
- Investigative Toxicology, Non-Clinical Development, UCB-Biopharma, Chemin du Foriest, 1420 Braine l'Alleud, Belgium
| |
Collapse
|
26
|
Selga E, Sendfeld F, Martinez-Moreno R, Medine CN, Tura-Ceide O, Wilmut SI, Pérez GJ, Scornik FS, Brugada R, Mills NL. Sodium channel current loss of function in induced pluripotent stem cell-derived cardiomyocytes from a Brugada syndrome patient. J Mol Cell Cardiol 2018; 114:10-19. [PMID: 29024690 PMCID: PMC5807028 DOI: 10.1016/j.yjmcc.2017.10.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 09/15/2017] [Accepted: 10/04/2017] [Indexed: 12/24/2022]
Abstract
Brugada syndrome predisposes to sudden death due to disruption of normal cardiac ion channel function, yet our understanding of the underlying cellular mechanisms is incomplete. Commonly used heterologous expression models lack many characteristics of native cardiomyocytes and, in particular, the individual genetic background of a patient. Patient-specific induced pluripotent stem (iPS) cell-derived cardiomyocytes (iPS-CM) may uncover cellular phenotypical characteristics not observed in heterologous models. Our objective was to determine the properties of the sodium current in iPS-CM with a mutation in SCN5A associated with Brugada syndrome. Dermal fibroblasts from a Brugada syndrome patient with a mutation in SCN5A (c.1100G>A, leading to Nav1.5_p.R367H) were reprogrammed to iPS cells. Clones were characterized and differentiated to form beating clusters and sheets. Patient and control iPS-CM were structurally indistinguishable. Sodium current properties of patient and control iPS-CM were compared. These results were contrasted with those obtained in tsA201 cells heterologously expressing sodium channels with the same mutation. Patient-derived iPS-CM showed a 33.1-45.5% reduction in INa density, a shift in both activation and inactivation voltage-dependence curves, and faster recovery from inactivation. Co-expression of wild-type and mutant channels in tsA201 cells did not compromise channel trafficking to the membrane, but resulted in a reduction of 49.8% in sodium current density without affecting any other parameters. Cardiomyocytes derived from iPS cells from a Brugada syndrome patient with a mutation in SCN5A recapitulate the loss of function of sodium channel current associated with this syndrome; including pro-arrhythmic changes in channel function not detected using conventional heterologous expression systems.
Collapse
Affiliation(s)
- Elisabet Selga
- Cardiovascular Genetics Centre, Department of Medical Sciences, University of Girona, Girona, Spain; Institut d'Investigació Biomèdica de Girona (IDIBGI), Girona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Franziska Sendfeld
- Scottish Centre for Regenerative Medicine, University of Edinburgh, United Kingdom; BHF/University Centre for Cardiovascular Sciences, University of Edinburgh, United Kingdom
| | - Rebecca Martinez-Moreno
- Cardiovascular Genetics Centre, Department of Medical Sciences, University of Girona, Girona, Spain; Institut d'Investigació Biomèdica de Girona (IDIBGI), Girona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Claire N Medine
- Scottish Centre for Regenerative Medicine, University of Edinburgh, United Kingdom; BHF/University Centre for Cardiovascular Sciences, University of Edinburgh, United Kingdom
| | - Olga Tura-Ceide
- Department of Pulmonary Medicine, Hospital Clinic-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, University of Barcelona, Spain
| | - Sir Ian Wilmut
- Scottish Centre for Regenerative Medicine, University of Edinburgh, United Kingdom
| | - Guillermo J Pérez
- Cardiovascular Genetics Centre, Department of Medical Sciences, University of Girona, Girona, Spain; Institut d'Investigació Biomèdica de Girona (IDIBGI), Girona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Fabiana S Scornik
- Cardiovascular Genetics Centre, Department of Medical Sciences, University of Girona, Girona, Spain; Institut d'Investigació Biomèdica de Girona (IDIBGI), Girona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Ramon Brugada
- Cardiovascular Genetics Centre, Department of Medical Sciences, University of Girona, Girona, Spain; Institut d'Investigació Biomèdica de Girona (IDIBGI), Girona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain; Hospital Josep Trueta, Girona, Spain
| | - Nicholas L Mills
- BHF/University Centre for Cardiovascular Sciences, University of Edinburgh, United Kingdom.
| |
Collapse
|
27
|
Isobe T, Honda M, Komatsu R, Tabo M. Conduction and contraction properties of human iPS cell-derived cardiomyocytes: analysis by motion field imaging compared with the guinea-pig isolated heart model. J Toxicol Sci 2018; 43:493-506. [DOI: 10.2131/jts.43.493] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
| | - Masaki Honda
- Research Division, Chugai Pharmaceutical Co., Ltd
| | | | | |
Collapse
|
28
|
Takeda M, Miyagawa S, Fukushima S, Saito A, Ito E, Harada A, Matsuura R, Iseoka H, Sougawa N, Mochizuki-Oda N, Matsusaki M, Akashi M, Sawa Y. Development of In Vitro Drug-Induced Cardiotoxicity Assay by Using Three-Dimensional Cardiac Tissues Derived from Human Induced Pluripotent Stem Cells. Tissue Eng Part C Methods 2017; 24:56-67. [PMID: 28967302 PMCID: PMC5757089 DOI: 10.1089/ten.tec.2017.0247] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
An in vitro drug-induced cardiotoxicity assay is a critical step in drug discovery for clinical use. The use of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) is promising for this purpose. However, single hiPSC-CMs are limited in their ability to mimic native cardiac tissue structurally and functionally, and the generation of artificial cardiac tissue using hiPSC-CMs is an ongoing challenging. We therefore developed a new method of constructing three-dimensional (3D) artificial tissues in a short time by coating extracellular matrix (ECM) components on cell surfaces. We hypothesized that 3D cardiac tissues derived from hiPSC-CMs (3D-hiPSC-CT) could be used for an in vitro drug-induced cardiotoxicity assay. 3D-hiPSC-CT were generated by fibronectin and gelatin nanofilm coated single hiPSC-CMs. Histologically, 3D-hiPSC-CT exhibited a sarcomere structure in the myocytes and ECM proteins, such as fibronectin, collagen type I/III, and laminin. The administration of cytotoxic doxorubicin at 5.0 μM induced the release of lactate dehydrogenase, while that at 2.0 μM reduced the cell viability. E-4031, human ether-a-go-go related gene (hERG)-type potassium channel blocker, and isoproterenol induced significant changes both in the Ca transient parameters and contractile parameters in a dose-dependent manner. The 3D-hiPSC-CT exhibited doxorubicin-sensitive cytotoxicity and hERG channel blocker/isoproterenol-sensitive electrical activity in vitro, indicating its usefulness for drug-induced cardiotoxicity assays or drug screening systems for drug discovery.
Collapse
Affiliation(s)
- Maki Takeda
- 1 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Suita, Osaka, Japan
| | - Shigeru Miyagawa
- 1 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Suita, Osaka, Japan
| | - Satsuki Fukushima
- 1 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Suita, Osaka, Japan
| | - Atsuhiro Saito
- 1 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Suita, Osaka, Japan
| | - Emiko Ito
- 1 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Suita, Osaka, Japan
| | - Akima Harada
- 1 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Suita, Osaka, Japan
| | - Ryohei Matsuura
- 1 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Suita, Osaka, Japan
| | - Hiroko Iseoka
- 1 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Suita, Osaka, Japan
| | - Nagako Sougawa
- 1 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Suita, Osaka, Japan
| | - Noriko Mochizuki-Oda
- 1 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Suita, Osaka, Japan
| | - Michiya Matsusaki
- 2 Department of Applied Chemistry, Osaka University Graduate School of Engineering , Osaka, Japan
| | - Mitsuru Akashi
- 3 Building Block Science Joint Research Chair, Graduate School of Frontier Biosciences, Osaka University , Suita, Japan
| | - Yoshiki Sawa
- 1 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Suita, Osaka, Japan
| |
Collapse
|
29
|
Yoon JK, Lee TI, Bhang SH, Shin JY, Myoung JM, Kim BS. Stretchable Piezoelectric Substrate Providing Pulsatile Mechanoelectric Cues for Cardiomyogenic Differentiation of Mesenchymal Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2017; 9:22101-22111. [PMID: 28560866 DOI: 10.1021/acsami.7b03050] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Ex vivo induction of cardiomyogenic differentiation of mesenchymal stem cells (MSCs) before implantation would potentiate therapeutic efficacy of stem cell therapies for ischemic heart diseases because MSCs rarely undergo cardiomyogenic differentiation following implantation. In cardiac microenvironments, electric pulse and cyclic mechanical strain are sequentially produced. However, no study has applied the pulsatile mechanoelectric cues (PMEC) to stimulate cardiomyogenic differentiation of MSCs ex vivo. In this study, we developed a stretchable piezoelectric substrate (SPS) that can provide PMEC to human MSCs (hMSCs) for cardiomyogenic differentiation ex vivo. Our data showed that hMSCs subjected to PMEC by SPS underwent promoted cardiac phenotype development: cell alignment and the expression of cardiac markers (i.e., cardiac transcription factors, structural proteins, ion channel proteins, and gap junction proteins). The enhanced cardiac phenotype development was mediated by the upregulation of cardiomyogenic differentiation-related autocrine factor expression, focal adhesion kinase, and extracellular signal-regulated kinases signaling pathways. Thus, SPS providing electrical and mechanical regulation of stem cells may be utilized to potentiate hMSC therapies for myocardial infarction and provide a tool for the study of stem cell biology.
Collapse
Affiliation(s)
| | - Tae Il Lee
- Department of BioNano Technology, Gachon University , Seongnam 13557, Republic of Korea
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University , Suwon 16419, Republic of Korea
| | | | - Jae-Min Myoung
- Department of Materials Science and Engineering, Yonsei University , Seoul 03722, Republic of Korea
| | | |
Collapse
|
30
|
Hansen KJ, Favreau JT, Gershlak JR, Laflamme MA, Albrecht DR, Gaudette GR. Optical Method to Quantify Mechanical Contraction and Calcium Transients of Human Pluripotent Stem Cell-Derived Cardiomyocytes. Tissue Eng Part C Methods 2017; 23:445-454. [PMID: 28562232 DOI: 10.1089/ten.tec.2017.0190] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Differentiation of human pluripotent stem cells into cardiomyocytes (hPS-CMs) holds promise for myocardial regeneration therapies, drug discovery, and models of cardiac disease. Potential cardiotoxicities may affect hPS-CM mechanical contraction independent of calcium signaling. Herein, a method using an image capture system is described to measure hPS-CM contractility and intracellular calcium concurrently, with high spatial and temporal resolution. The image capture system rapidly alternates between brightfield and epifluorescent illumination of contracting cells. Mechanical contraction is quantified by a speckle tracking algorithm applied to brightfield image pairs, whereas calcium transients are measured by a fluorescent calcium reporter. This technique captured changes in contractile strain, calcium transients, and beat frequency of hPS-CMs over 21 days in culture, as well as acute responses to isoproterenol and Cytochalasin D. The technique described above can be applied without the need to alter the culture platform, allowing for determination of hPS-CM behavior over weeks in culture for drug discovery and myocardial regeneration applications.
Collapse
Affiliation(s)
- Katrina J Hansen
- 1 Department of Biomedical Engineering, Worcester Polytechnic Institute , Worcester, Massachusetts
| | - John T Favreau
- 1 Department of Biomedical Engineering, Worcester Polytechnic Institute , Worcester, Massachusetts
| | - Joshua R Gershlak
- 1 Department of Biomedical Engineering, Worcester Polytechnic Institute , Worcester, Massachusetts
| | - Michael A Laflamme
- 2 Toronto General Research Institute, McEwen Centre for Regenerative Medicine, University Health Network , Toronto, Canada
| | - Dirk R Albrecht
- 1 Department of Biomedical Engineering, Worcester Polytechnic Institute , Worcester, Massachusetts
| | - Glenn R Gaudette
- 1 Department of Biomedical Engineering, Worcester Polytechnic Institute , Worcester, Massachusetts
| |
Collapse
|
31
|
Affiliation(s)
- Yoshinori Yoshida
- From the Center for iPS Cell Research and Application, Kyoto University, Japan (Y.Y., S.Y.); and Gladstone Institute of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA (S.Y.)
| | - Shinya Yamanaka
- From the Center for iPS Cell Research and Application, Kyoto University, Japan (Y.Y., S.Y.); and Gladstone Institute of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA (S.Y.)
| |
Collapse
|
32
|
Overexpression of KCNJ2 in induced pluripotent stem cell-derived cardiomyocytes for the assessment of QT-prolonging drugs. J Pharmacol Sci 2017; 134:75-85. [DOI: 10.1016/j.jphs.2017.05.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 05/14/2017] [Accepted: 05/17/2017] [Indexed: 02/05/2023] Open
|
33
|
Pei F, Jiang J, Bai S, Cao H, Tian L, Zhao Y, Yang C, Dong H, Ma Y. Chemical-defined and albumin-free generation of human atrial and ventricular myocytes from human pluripotent stem cells. Stem Cell Res 2017; 19:94-103. [PMID: 28110125 DOI: 10.1016/j.scr.2017.01.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 01/07/2017] [Accepted: 01/09/2017] [Indexed: 12/17/2022] Open
Abstract
Most existing culture media for cardiac differentiation of human pluripotent stem cells (hPSCs) contain significant amounts of albumin. For clinical transplantation applications of hPSC-derived cardiomyocytes (hPSC-CMs), culturing cells in an albumin containing environment raises the concern of pathogen contamination and immunogenicity to the recipient patients. In addition, batch-to-batch variation of albumin may cause the inconsistent of hPSC cardiac differentiation. Here, we demonstrated that antioxidants l-ascorbic acid, trolox, N-acetyl-l-cysteine (NAC) and sodium pyruvate could functionally substitute albumin in the culture medium, and formulated an albumin-free, chemical-defined medium (S12 medium). We showed that S12 medium could support efficient hPSC cardiac differentiation with significantly improved reproducibility, and maintained long-term culture of hPSC-CMs. Furthermore, under chemical-defined and albumin-free conditions, human-induced pluripotent stem cells (hiPSCs) were established, and differentiated into highly homogenous atrial and ventricular myocytes in a scalable fashion with normal electrophysiological properties. Finally, we characterized the activity of three typical cardiac ion channels of those cells, and demonstrated that hPSC-derived ventricular cardiomyocytes (hPSC-vCMs) were suitable for drug cardiac safety evaluation. In summary, this simplified, chemical-defined and albumin-free culture medium supports efficient generation and maintaining of hPSC-CMs and facilitates both research and clinical applications of these cells.
Collapse
Affiliation(s)
- Fei Pei
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing 100101, China; Medical School of University of Chinese Academy of Sciences, Beijing 100101, China
| | - Junjie Jiang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing 100101, China; Medical School of University of Chinese Academy of Sciences, Beijing 100101, China
| | - Shuyun Bai
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing 100101, China; Medical School of University of Chinese Academy of Sciences, Beijing 100101, China
| | - Henghua Cao
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing 100101, China; Medical School of University of Chinese Academy of Sciences, Beijing 100101, China
| | - Luyang Tian
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing 100101, China; Medical School of University of Chinese Academy of Sciences, Beijing 100101, China
| | - Ya Zhao
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing 100101, China; Medical School of University of Chinese Academy of Sciences, Beijing 100101, China
| | - Chuanxiu Yang
- Biology Unit, Research Division, WuXi AppTec Co., Ltd., Shanghai 200131, China
| | - Haiheng Dong
- Biology Unit, Research Division, WuXi AppTec Co., Ltd., Shanghai 200131, China
| | - Yue Ma
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing 100101, China; Medical School of University of Chinese Academy of Sciences, Beijing 100101, China..
| |
Collapse
|
34
|
Hortigon-Vinagre MP, Zamora V, Burton FL, Green J, Gintant GA, Smith GL. The Use of Ratiometric Fluorescence Measurements of the Voltage Sensitive Dye Di-4-ANEPPS to Examine Action Potential Characteristics and Drug Effects on Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Toxicol Sci 2016; 154:320-331. [PMID: 27621282 PMCID: PMC5139069 DOI: 10.1093/toxsci/kfw171] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) and higher throughput platforms have emerged as potential tools to advance cardiac drug safety screening. This study evaluated the use of high bandwidth photometry applied to voltage-sensitive fluorescent dyes (VSDs) to assess drug-induced changes in action potential characteristics of spontaneously active hiPSC-CM. Human iPSC-CM from 2 commercial sources (Cor.4U and iCell Cardiomyocytes) were stained with the VSD di-4-ANEPPS and placed in a specialized photometry system that simultaneously monitors 2 wavebands of emitted fluorescence, allowing ratiometric measurement of membrane voltage. Signals were acquired at 10 kHz and analyzed using custom software. Action potential duration (APD) values were normally distributed in cardiomyocytes (CMC) from both sources though the mean and variance differed significantly (APD90: 229 ± 15 ms vs 427 ± 49 ms [mean ± SD, P < 0.01]; average spontaneous cycle length: 0.99 ± 0.02 s vs 1.47 ± 0.35 s [mean ± SD, P < 0.01], Cor.4U vs iCell CMC, respectively). The 10-90% rise time of the AP (Trise) was ∼6 ms and was normally distributed when expressed as 1/[Formula: see text] in both cell preparations. Both cell types showed a rate dependence analogous to that of adult human cardiac cells. Furthermore, nifedipine, ranolazine, and E4031 had similar effects on cardiomyocyte electrophysiology in both cell types. However, ranolazine and E4031 induced early after depolarization-like events and high intrinsic firing rates at lower concentrations in iCell CMC. These data show that VSDs provide a minimally invasive, quantitative, and accurate method to assess hiPSC-CM electrophysiology and detect subtle drug-induced effects for drug safety screening while highlighting a need to standardize experimental protocols across preparations.
Collapse
Affiliation(s)
- M P Hortigon-Vinagre
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Science, University of Glasgow 126 University Place, Glasgow G12 8TA, United Kingdom
- Clyde Biosciences Ltd, BioCity Scotland, Bo'Ness Road, Newhouse, Lanarkshire, Scotland ML1 5UH, United Kingdom
| | - V Zamora
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Science, University of Glasgow 126 University Place, Glasgow G12 8TA, United Kingdom
- Clyde Biosciences Ltd, BioCity Scotland, Bo'Ness Road, Newhouse, Lanarkshire, Scotland ML1 5UH, United Kingdom
| | - F L Burton
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Science, University of Glasgow 126 University Place, Glasgow G12 8TA, United Kingdom
- Clyde Biosciences Ltd, BioCity Scotland, Bo'Ness Road, Newhouse, Lanarkshire, Scotland ML1 5UH, United Kingdom
| | - J Green
- AbbVie, 1 North Waukegan Road, Department ZR-13, Building AP-9A, North Chicago, Illinois 60064-6119
| | - G A Gintant
- AbbVie, 1 North Waukegan Road, Department ZR-13, Building AP-9A, North Chicago, Illinois 60064-6119
| | - G L Smith
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Science, University of Glasgow 126 University Place, Glasgow G12 8TA, United Kingdom
- Clyde Biosciences Ltd, BioCity Scotland, Bo'Ness Road, Newhouse, Lanarkshire, Scotland ML1 5UH, United Kingdom
| |
Collapse
|
35
|
Points to consider for a validation study of iPS cell-derived cardiomyocytes using a multi-electrode array system. J Pharmacol Toxicol Methods 2016; 81:196-200. [DOI: 10.1016/j.vascn.2016.06.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 06/25/2016] [Accepted: 06/27/2016] [Indexed: 12/16/2022]
|
36
|
Dempsey GT, Chaudhary KW, Atwater N, Nguyen C, Brown BS, McNeish JD, Cohen AE, Kralj JM. Cardiotoxicity screening with simultaneous optogenetic pacing, voltage imaging and calcium imaging. J Pharmacol Toxicol Methods 2016; 81:240-50. [DOI: 10.1016/j.vascn.2016.05.003] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 05/02/2016] [Accepted: 05/09/2016] [Indexed: 12/23/2022]
|
37
|
|
38
|
Eder A, Vollert I, Hansen A, Eschenhagen T. Human engineered heart tissue as a model system for drug testing. Adv Drug Deliv Rev 2016; 96:214-24. [PMID: 26026976 DOI: 10.1016/j.addr.2015.05.010] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 05/11/2015] [Accepted: 05/21/2015] [Indexed: 12/29/2022]
Abstract
Drug development is time- and cost-intensive and, despite extensive efforts, still hampered by the limited value of current preclinical test systems to predict side effects, including proarrhythmic and cardiotoxic effects in clinical practice. Part of the problem may be related to species-dependent differences in cardiomyocyte biology. Therefore, the event of readily available human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CM) has raised hopes that this human test bed could improve preclinical safety pharmacology as well as drug discovery approaches. However, hiPSC-CM are immature and exhibit peculiarities in terms of ion channel function, gene expression, structural organization and functional responses to drugs that limit their present usefulness. Current efforts are thus directed towards improving hiPSC-CM maturity and high-content readouts. Culturing hiPSC-CM as 3-dimensional engineered heart tissue (EHT) improves CM maturity and anisotropy and, in a 24-well format using silicone racks, enables automated, multiplexed high content readout of contractile function. This review summarizes the principal technology and focuses on advantages and disadvantages of this technology and its potential for preclinical drug screening.
Collapse
|
39
|
Higa A, Hoshi H, Takagi M. Differing responses of human stem cell-derived cardiomyocytes to arrhythmogenic drugs, determined using impedance measurements. ACTA ACUST UNITED AC 2016. [DOI: 10.2131/fts.3.47] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Arisa Higa
- Medical-industrial Translational Research Center, Fukushima Medical University
| | - Hirotaka Hoshi
- Medical-industrial Translational Research Center, Fukushima Medical University
| | - Motoki Takagi
- Medical-industrial Translational Research Center, Fukushima Medical University
| |
Collapse
|
40
|
Bedada FB, Wheelwright M, Metzger JM. Maturation status of sarcomere structure and function in human iPSC-derived cardiac myocytes. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:1829-38. [PMID: 26578113 DOI: 10.1016/j.bbamcr.2015.11.005] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 11/05/2015] [Accepted: 11/09/2015] [Indexed: 12/15/2022]
Abstract
Human heart failure due to myocardial infarction is a major health concern. The paucity of organs for transplantation limits curative approaches for the diseased and failing adult heart. Human induced pluripotent stem cell-derived cardiac myocytes (hiPSC-CMs) have the potential to provide a long-term, viable, regenerative-medicine alternative. Significant progress has been made with regard to efficient cardiac myocyte generation from hiPSCs. However, directing hiPSC-CMs to acquire the physiological structure, gene expression profile and function akin to mature cardiac tissue remains a major obstacle. Thus, hiPSC-CMs have several hurdles to overcome before they find their way into translational medicine. In this review, we address the progress that has been made, the void in knowledge and the challenges that remain. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.
Collapse
Affiliation(s)
- Fikru B Bedada
- Department of Integrative Biology and Physiology, University of Minnesota Medical School Minneapolis, MN 55455, USA
| | - Matthew Wheelwright
- Department of Integrative Biology and Physiology, University of Minnesota Medical School Minneapolis, MN 55455, USA
| | - Joseph M Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School Minneapolis, MN 55455, USA.
| |
Collapse
|
41
|
Sallam K, Li Y, Sager PT, Houser SR, Wu JC. Finding the rhythm of sudden cardiac death: new opportunities using induced pluripotent stem cell-derived cardiomyocytes. Circ Res 2015; 116:1989-2004. [PMID: 26044252 DOI: 10.1161/circresaha.116.304494] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Sudden cardiac death is a common cause of death in patients with structural heart disease, genetic mutations, or acquired disorders affecting cardiac ion channels. A wide range of platforms exist to model and study disorders associated with sudden cardiac death. Human clinical studies are cumbersome and are thwarted by the extent of investigation that can be performed on human subjects. Animal models are limited by their degree of homology to human cardiac electrophysiology, including ion channel expression. Most commonly used cellular models are cellular transfection models, which are able to mimic the expression of a single-ion channel offering incomplete insight into changes of the action potential profile. Induced pluripotent stem cell-derived cardiomyocytes resemble, but are not identical, adult human cardiomyocytes and provide a new platform for studying arrhythmic disorders leading to sudden cardiac death. A variety of platforms exist to phenotype cellular models, including conventional and automated patch clamp, multielectrode array, and computational modeling. Induced pluripotent stem cell-derived cardiomyocytes have been used to study long QT syndrome, catecholaminergic polymorphic ventricular tachycardia, hypertrophic cardiomyopathy, and other hereditary cardiac disorders. Although induced pluripotent stem cell-derived cardiomyocytes are distinct from adult cardiomyocytes, they provide a robust platform to advance the science and clinical care of sudden cardiac death.
Collapse
Affiliation(s)
- Karim Sallam
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (K.S., Y.L., P.T.S., J.C.W.), Institute of Stem Cell Biology and Regenerative Medicine (K.S., Y.L., J.C.W.), Stanford University School of Medicine, CA; and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Yingxin Li
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (K.S., Y.L., P.T.S., J.C.W.), Institute of Stem Cell Biology and Regenerative Medicine (K.S., Y.L., J.C.W.), Stanford University School of Medicine, CA; and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Philip T Sager
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (K.S., Y.L., P.T.S., J.C.W.), Institute of Stem Cell Biology and Regenerative Medicine (K.S., Y.L., J.C.W.), Stanford University School of Medicine, CA; and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Steven R Houser
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (K.S., Y.L., P.T.S., J.C.W.), Institute of Stem Cell Biology and Regenerative Medicine (K.S., Y.L., J.C.W.), Stanford University School of Medicine, CA; and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.).
| | - Joseph C Wu
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (K.S., Y.L., P.T.S., J.C.W.), Institute of Stem Cell Biology and Regenerative Medicine (K.S., Y.L., J.C.W.), Stanford University School of Medicine, CA; and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.).
| |
Collapse
|
42
|
Lee S, Lee HA, Kim SJ, Kim KS. Cellular mechanisms for trazodone-induced cardiotoxicity. Hum Exp Toxicol 2015; 35:501-10. [DOI: 10.1177/0960327115595683] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The second-generation selective 5-HT2 receptor antagonists and reuptake inhibitors (SARIs) class antidepressants are known to have fewer cardiovascular side effects than the older ones. However, several case reports showed that trazodone, one of the second-generation SARIs, induces QT prolongation, cardiac arrhythmia, and ventricular tachycardia. Although these clinical cases suggested trazodone-induced cardiotoxicity, the toxicological actions of trazodone on cardiac action potentials (APs) beyond the human ether-a-go-go related gene (hERG) remain unclear. To elucidate the cellular mechanism for the adverse cardiac effects of trazodone, we investigated its effects on cardiac APs and ion channels using whole-cell patch clamp techniques in human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and transiently transfected human embryonic kidney cells (HEK293) with cardiac ion channel complementary DNA. Trazodone dose-dependently decreased the maximum upstroke velocity ( Vmax) and prolonged the AP duration, inducing early after depolarizations at 3 and 10 μM that triggered ventricular arrhythmias in hiPSC-CMs. Trazodone also inhibited all of the major ion channels ( IKr, IKs, INa, and ICa), with an especially high inhibitory potency on hERG. These data indicate that the prolonged AP duration and decreased Vmax due to trazodone are mainly the result of hERG and sodium ion inhibition, and its inhibitory effects on cardiac ion channels can be exhibited in hiPSC-CMs.
Collapse
Affiliation(s)
- S Lee
- Next-Generation Pharmaceutical Research Center, Korea Institute of Toxicology, Korea Research Institute of Chemical Technology, Yuseong-gu, Daejeon, Korea
| | - H-A Lee
- Next-Generation Pharmaceutical Research Center, Korea Institute of Toxicology, Korea Research Institute of Chemical Technology, Yuseong-gu, Daejeon, Korea
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - SJ Kim
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - K-S Kim
- Next-Generation Pharmaceutical Research Center, Korea Institute of Toxicology, Korea Research Institute of Chemical Technology, Yuseong-gu, Daejeon, Korea
| |
Collapse
|
43
|
Gintant G. Cardiac Sodium Current (Na v1.5). METHODS AND PRINCIPLES IN MEDICINAL CHEMISTRY 2015. [DOI: 10.1002/9783527673643.ch12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
44
|
Liu QN, Trudeau MC. Eag Domains Regulate LQT Mutant hERG Channels in Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. PLoS One 2015; 10:e0123951. [PMID: 25923442 PMCID: PMC4414485 DOI: 10.1371/journal.pone.0123951] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 03/09/2015] [Indexed: 01/08/2023] Open
Abstract
Human Ether á go-go Related Gene potassium channels form the rapid component of the delayed-rectifier (IKr) current in the heart. The N-terminal 'eag' domain, which is composed of a Per-Arnt-Sim (PAS) domain and a short PAS-cap region, is a critical regulator of hERG channel function. In previous studies, we showed that isolated eag (i-eag) domains rescued the dysfunction of long QT type-2 associated mutant hERG R56Q channels, by substituting for defective eag domains, when the channels were expressed in Xenopus oocytes or HEK 293 cells.Here, our goal was to determine whether the rescue of hERG R56Q channels by i-eag domains could be translated into the environment of cardiac myocytes. We expressed hERG R56Q channels in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and measured electrical properties of the cells with whole-cell patch-clamp recordings. We found that, like in non-myocyte cells, hERG R56Q had defective, fast closing (deactivation) kinetics when expressed in hiPSC-CMs. We report here that i-eag domains slowed the deactivation kinetics of hERG R56Q channels in hiPSC-CMs. hERG R56Q channels prolonged the AP of hiPSCs, and the AP was shortened by co-expression of i-eag domains and hERG R56Q channels. We measured robust Förster Resonance Energy Transfer (FRET) between i-eag domains tagged with Cyan fluorescent protein (CFP) and hERG R56Q channels tagged with Citrine fluorescent proteins (Citrine), indicating their close proximity at the cell membrane in live iPSC-CMs. Together, functional regulation and FRET spectroscopy measurements indicated that i-eag domains interacted directly with hERG R56Q channels in hiPSC-CMs. These results mean that the regulatory role of i-eag domains is conserved in the cellular environment of human cardiomyocytes, indicating that i-eag domains may be useful as a biological therapeutic.
Collapse
Affiliation(s)
- Qiang-ni Liu
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Matthew C. Trudeau
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
45
|
Wegener M, Bader A, Giri S. How to mend a broken heart: adult and induced pluripotent stem cell therapy for heart repair and regeneration. Drug Discov Today 2015; 20:667-85. [PMID: 25720353 DOI: 10.1016/j.drudis.2015.02.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 11/30/2014] [Accepted: 02/16/2015] [Indexed: 01/06/2023]
Abstract
The recently developed ability to differentiate primary adult stem cells and induced pluripotent stem cells (iPSCs) into cardiomyocytes is providing unprecedented opportunities to produce an unlimited supply of cardiomyocytes for use in patients with heart disease. Here, we examine the evidence for the preclinical use of such cells for successful heart regeneration. We also describe advances in the identification of new cardiac molecular and cellular targets to induce proliferation of cardiomyocytes for heart regeneration. Such new advances are paving the way for a new innovative drug development process for the treatment of heart disease.
Collapse
Affiliation(s)
- Marie Wegener
- Centre for Biotechnology and Biomedicine, Department of Cell Techniques and Applied Stem Cell Biology, Medical Faculty of University of Leipzig, Deutscher Platz 5, Leipzig D-04103, Germany
| | - Augustinus Bader
- Centre for Biotechnology and Biomedicine, Department of Cell Techniques and Applied Stem Cell Biology, Medical Faculty of University of Leipzig, Deutscher Platz 5, Leipzig D-04103, Germany
| | - Shibashish Giri
- Centre for Biotechnology and Biomedicine, Department of Cell Techniques and Applied Stem Cell Biology, Medical Faculty of University of Leipzig, Deutscher Platz 5, Leipzig D-04103, Germany.
| |
Collapse
|
46
|
Image-based evaluation of contraction–relaxation kinetics of human-induced pluripotent stem cell-derived cardiomyocytes: Correlation and complementarity with extracellular electrophysiology. J Mol Cell Cardiol 2014; 77:178-91. [DOI: 10.1016/j.yjmcc.2014.09.010] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Accepted: 09/10/2014] [Indexed: 01/05/2023]
|
47
|
Ben-Ari M, Schick R, Barad L, Novak A, Ben-Ari E, Lorber A, Itskovitz-Eldor J, Rosen MR, Weissman A, Binah O. From beat rate variability in induced pluripotent stem cell-derived pacemaker cells to heart rate variability in human subjects. Heart Rhythm 2014; 11:1808-1818. [PMID: 25052725 PMCID: PMC4283811 DOI: 10.1016/j.hrthm.2014.05.037] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Indexed: 01/27/2023]
Abstract
BACKGROUND We previously reported that induced pluripotent stem cell-derived cardiomyocytes manifest beat rate variability (BRV) resembling heart rate variability (HRV) in the human sinoatrial node. We now hypothesized the BRV-HRV continuum originates in pacemaker cells. OBJECTIVE To investigate whether cellular BRV is a source of HRV dynamics, we hypothesized 3 levels of interaction among different cardiomyocyte entities: (1) single pacemaker cells, (2) networks of electrically coupled pacemaker cells, and (3) the in situ sinoatrial node. METHODS We measured BRV/HRV properties in single pacemaker cells, induced pluripotent stem cell-derived contracting embryoid bodies (EBs), and electrocardiograms from the same individual. RESULTS Pronounced BRV/HRV was present at all 3 levels. The coefficient of variance of interbeat intervals and Poincaré plot indices SD1 and SD2 for single cells were 20 times greater than those for EBs (P < .05) and the in situ heart (the latter two were similar; P > .05). We also compared BRV magnitude among single cells, small EBs (~5-10 cells), and larger EBs (>10 cells): BRV indices progressively increased with the decrease in the cell number (P < .05). Disrupting intracellular Ca(2+) handling markedly augmented BRV magnitude, revealing a unique bimodal firing pattern, suggesting that intracellular mechanisms contribute to BRV/HRV and the fractal behavior of heart rhythm. CONCLUSION The decreased BRV magnitude in transitioning from the single cell to the EB suggests that the HRV of in situ hearts originates from the summation and integration of multiple cell-based oscillators. Hence, complex interactions among multiple pacemaker cells and intracellular Ca(2+) handling determine HRV in humans and cardiomyocyte networks.
Collapse
Affiliation(s)
- Meital Ben-Ari
- The Sohnis Family Stem Cells Center, Technion, Haifa, Israel
- The Rappaport Institute, Technion, Haifa, Israel
- Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Revital Schick
- The Sohnis Family Stem Cells Center, Technion, Haifa, Israel
- The Rappaport Institute, Technion, Haifa, Israel
- Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Lili Barad
- The Sohnis Family Stem Cells Center, Technion, Haifa, Israel
- The Rappaport Institute, Technion, Haifa, Israel
- Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Atara Novak
- The Sohnis Family Stem Cells Center, Technion, Haifa, Israel
- The Rappaport Institute, Technion, Haifa, Israel
- Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Erez Ben-Ari
- Department of Electrical Engineering, Technion, Haifa, Israel
| | - Avraham Lorber
- Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
- Department of Pediatric Cardiology, Rambam Medical Center
| | - Joseph Itskovitz-Eldor
- The Sohnis Family Stem Cells Center, Technion, Haifa, Israel
- Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
- Department of Obstetrics and Gynecology, Rambam Medical Center, Haifa, Israel
| | - Michael R Rosen
- Department of Pharmacology, College of Physicians and Surgeons of Columbia University, New York, USA
| | - Amir Weissman
- Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
- Department of Obstetrics and Gynecology, Rambam Medical Center, Haifa, Israel
| | - Ofer Binah
- The Sohnis Family Stem Cells Center, Technion, Haifa, Israel
- The Rappaport Institute, Technion, Haifa, Israel
- Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| |
Collapse
|
48
|
Clements M, Thomas N. High-throughput multi-parameter profiling of electrophysiological drug effects in human embryonic stem cell derived cardiomyocytes using multi-electrode arrays. Toxicol Sci 2014; 140:445-61. [PMID: 24812011 DOI: 10.1093/toxsci/kfu084] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Human stem cell derived cardiomyocytes (hESC-CM) provide a potential model for development of improved assays for pre-clinical predictive drug safety screening. We have used multi-electrode array (MEA) analysis of hESC-CM to generate multi-parameter data to profile drug impact on cardiomyocyte electrophysiology using a panel of 21 compounds active against key cardiac ion channels. Our study is the first to apply multi-parameter phenotypic profiling and clustering techniques commonly used for high-content imaging and microarray data to the analysis of electrophysiology data obtained by MEA analysis. Our data show good correlations with previous studies in stem cell derived cardiomyocytes and demonstrate improved specificity in compound risk assignment over convention single-parametric approaches. These analyses indicate great potential for multi-parameter MEA data acquired from hESC-CM to enable drug electrophysiological liabilities to be assessed in pre-clinical cardiotoxicity assays, facilitating informed decision making and liability management at the optimum point in drug development.
Collapse
Affiliation(s)
| | - Nick Thomas
- GE Healthcare Life Sciences, Cardiff CF14 7YT, UK
| |
Collapse
|
49
|
Aikawa N, Kunisato A, Nagao K, Kusaka H, Takaba K, Ohgami K. Detection of thalidomide embryotoxicity by in vitro embryotoxicity testing based on human iPS cells. J Pharmacol Sci 2014; 124:201-7. [PMID: 24451995 DOI: 10.1254/jphs.13162fp] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
The mouse embryonic stem cell test (mEST) is used to assess the embryotoxicity of drug candidates by evaluating the effects on the cardiac differentiation of stem cells. However, thalidomide embryotoxicity has not yet been reported using the mEST. To detect the effects of thalidomide, we used human induced pluripotent stem cells (hiPSCs) instead of mouse embryonic stem cells, and assessed three endpoints: the inhibition of cardiac differentiation, the cytotoxicity to hiPSCs, and the cytotoxicity to human dermal fibroblasts, according to the mEST. From these data (IC50 values), the embryotoxicity was classified into one of three different classes based on the mEST and our criteria. Valproate was used as a positive control and ascorbic acid was used as a negative control, and their effects were assessed. Similar to valproate, thalidomide was classified as a Class 2 agent, with weak embryotoxicity, by the mEST criteria, and was classified as Category 3 embryotoxic based on our criteria. Ascorbic acid was classified as a Class 1 / Category 1, non-embryotoxic agent, based on both criteria. Thalidomide embryotoxicity was detected in the embryonic stem cell test based on hiPSCs. This test system is thus considered to have a much greater predictive ability than the mEST.
Collapse
Affiliation(s)
- Nobuo Aikawa
- Drug Discovery Research Laboratories, Fuji Research Park, Research Division, Kyowa Hakko Kirin Co., Ltd., Japan
| | | | | | | | | | | |
Collapse
|
50
|
Olmer R, Martin U. Induced Pluripotent Stem Cells Differentiate into Functional Cardiomyocytes. STEM CELLS AND CANCER STEM CELLS, VOLUME 12 2014. [DOI: 10.1007/978-94-017-8032-2_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|