Phenotypic assays for analyses of pluripotent stem cell–derived cardiomyocytes†
Martin Pesl
Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
ICRC, St. Anne's University Hospital, Brno, Czech Republic
Search for more papers by this authorGuido Caluori
ICRC, St. Anne's University Hospital, Brno, Czech Republic
CEITEC, Masaryk University, Brno, Czech Republic
Search for more papers by this authorVratislav Cmiel
Department of Biomedical Engineering, Faculty of Electrical Engineering and Communication, Brno University of Technology, Brno, Czech Republic
Search for more papers by this authorIvana Acimovic
Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
Search for more papers by this authorSarka Jelinkova
Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
Search for more papers by this authorPetr Dvorak
Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
ICRC, St. Anne's University Hospital, Brno, Czech Republic
Search for more papers by this authorZdenek Starek
ICRC, St. Anne's University Hospital, Brno, Czech Republic
Search for more papers by this authorPetr Skladal
CEITEC, Masaryk University, Brno, Czech Republic
Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
Search for more papers by this authorCorresponding Author
Vladimir Rotrekl
Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
ICRC, St. Anne's University Hospital, Brno, Czech Republic
Correspondence
Vladimir Rotrekl, Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.
Email: [email protected]
Search for more papers by this authorMartin Pesl
Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
ICRC, St. Anne's University Hospital, Brno, Czech Republic
Search for more papers by this authorGuido Caluori
ICRC, St. Anne's University Hospital, Brno, Czech Republic
CEITEC, Masaryk University, Brno, Czech Republic
Search for more papers by this authorVratislav Cmiel
Department of Biomedical Engineering, Faculty of Electrical Engineering and Communication, Brno University of Technology, Brno, Czech Republic
Search for more papers by this authorIvana Acimovic
Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
Search for more papers by this authorSarka Jelinkova
Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
Search for more papers by this authorPetr Dvorak
Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
ICRC, St. Anne's University Hospital, Brno, Czech Republic
Search for more papers by this authorZdenek Starek
ICRC, St. Anne's University Hospital, Brno, Czech Republic
Search for more papers by this authorPetr Skladal
CEITEC, Masaryk University, Brno, Czech Republic
Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
Search for more papers by this authorCorresponding Author
Vladimir Rotrekl
Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
ICRC, St. Anne's University Hospital, Brno, Czech Republic
Correspondence
Vladimir Rotrekl, Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.
Email: [email protected]
Search for more papers by this authorAbstract
Stem cell–derived cardiomyocytes (CMs) hold great hopes for myocardium regeneration because of their ability to produce functional cardiac cells in large quantities. They also hold promise in dissecting the molecular principles involved in heart diseases and also in drug development, owing to their ability to model the diseases using patient-specific human pluripotent stem cell (hPSC)–derived CMs. The CM properties essential for the desired applications are frequently evaluated through morphologic and genotypic screenings. Even though these characterizations are necessary, they cannot in principle guarantee the CM functionality and their drug response. The CM functional characteristics can be quantified by phenotype assays, including electrophysiological, optical, and/or mechanical approaches implemented in the past decades, especially when used to investigate responses of the CMs to known stimuli (eg, adrenergic stimulation). Such methods can be used to indirectly determine the electrochemomechanics of the cardiac excitation-contraction coupling, which determines important functional properties of the hPSC-derived CMs, such as their differentiation efficacy, their maturation level, and their functionality. In this work, we aim to systematically review the techniques and methodologies implemented in the phenotype characterization of hPSC-derived CMs. Further, we introduce a novel approach combining atomic force microscopy, fluorescent microscopy, and external electrophysiology through microelectrode arrays. We demonstrate that this novel method can be used to gain unique information on the complex excitation-contraction coupling dynamics of the hPSC-derived CMs.
REFERENCES
- 1Pilkerton CS, Singh SS, Thomas K, Bias TK, Frisbee SJ. Changes in cardiovascular health in the United States, 2003–2011. J Am Heart Assoc. 2015; 4: e001650.
- 2Mozaffarian D, Benjamin EJ, Go AS, et al. Heart disease and stroke statistics—2016 update: A report from the American Heart Association. Circulation. 2015. doi: 10.1161/CIR.0000000000000350
10.1161/CIR.0000000000000350 Google Scholar
- 3Aistrup GL, Shiferaw Y, Kapur S, Kadish AH, Wasserstrom JA. Mechanisms underlying the formation and dynamics of subcellular calcium alternans in the intact rat heart. Circ Res. 2009; 104: 639–649.
- 4Stienen GJM. Pathomechanisms in heart failure: The contractile connection. J Muscle Res Cell Motil. 2015; 36: 47–60.
- 5Holzmann M, Nicko A, Kühl U, et al. Complication rate of right ventricular endomyocardial biopsy via the femoral approach: A retrospective and prospective study analyzing 3048 diagnostic procedures over an 11-year period. Circulation. 2008; 118: 1722–1728.
- 6Imamura T, Kinugawa K, Nitta D, et al. Is the internal jugular vein or femoral vein a better approach site for endomyocardial biopsy in heart transplant recipients? Int Heart J. 2015; 56: 67–72.
- 7Thomson J, Itskovitz-Eldor J, Shapiro SS, et al. Embryonic stem cell lines derived from human blastocysts. Science. 1998; 282: 1145–1147.
- 8Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007; 131: 861–872.
- 9Jiang Y, Jahagirdar BN, Reinhardt RL, et al. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature. 2002; 418: 41–49.
- 10Acimovic I, Vilotic A, Pesl M, et al. Human pluripotent stem cell-derived cardiomyocytes as research and therapeutic tools. Biomed Res Int. 2014; 2014: 512831
- 11Ribeiro MC, Tertoolen LG, Guadix JA, et al. Biomaterials functional maturation of human pluripotent stem cell derived cardiomyocytes in vitro—correlation between contraction force and electrophysiology. Biomaterials. 2015; 51: 138–150.
- 12Zhu R, Blazeski A, Poon E, Coska KD, Tung L, Boheler KR. Physical developmental cues for the maturation of human pluripotent stem cell-derived cardiomyocytes. Stem Cell Res Ther. 2014; 5: 117
- 13Ranga A, Gjorevski N, Lutolf MP. Drug discovery through stem cell-based organoid models. Adv Drug Deliv Rev. 2014; 69-70: 19–28.
- 14Del Álamo JC, Lemons D, Serrano R, et al. High throughput physiological screening of iPSC-derived cardiomyocytes for drug development. Biochim Biophys Acta. 2016; 1863: 1717–1727.
- 15Lundy SD, Zhu W-Z, Regnier M, Laflamme MA. Structural and functional maturation of cardiomyocytes derived from human pluripotent stem cells. Stem Cells Dev. 2013; 22: 1991–2002.
- 16Pesl M, Acimovic I, Pribyl J, et al. Forced aggregation and defined factors allow highly uniform-sized embryoid bodies and functional cardiomyocytes from human embryonic and induced pluripotent stem cells. Heart Vessels. 2014; 29: 834–846.
- 17Moretti A, Laugwitz KL, Dorn T, Sinnecker D, Mummery C. Pluripotent stem cell models of human heart disease. Cold Spring Harb Perspect Med. 2013; 3:
- 18Burridge PW, Keller G, Gold JD, Wu JC. Production of de novo cardiomyocytes: Human pluripotent stem cell differentiation and direct reprogramming. Cell Stem Cell. 2012; 10: 16–28.
- 19Devalla HD, Schwach V, Ford JW, et al. Atrial-like cardiomyocytes from human pluripotent stem cells are a robust preclinical model for assessing atrial-selective pharmacology. EMBO Mol Med. 2015; 7: 394–410.
- 20Mummery CL, Zhang J, Ng ES, Elliott DA, Elefanty AG, Kamp TJ. Differentiation of human embryonic stem cells and induced pluripotent stem cells to cardiomyocytes: A methods overview. Circ Res. 2012; 111: 344–358.
- 21Thomas 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: 445–461.
- 22Wang T, Hu N, Cao J, Wu J, Su K, Wang P. A cardiomyocyte-based biosensor for antiarrhythmic drug evaluation by simultaneously monitoring cell growth and beating. Biosens Bioelectron. 2013; 49: 9–13.
- 23Swinney DC, Anthony J. How were new medicines discovered? Nat Rev Drug Discov. 2011; 10: 507–519.
- 24Scott CW, Peters MF, Dragan YP. Human induced pluripotent stem cells and their use in drug discovery for toxicity testing. Toxicol Lett. 2013; 219: 49–58.
- 25Chow M, Boheler KR, Li RA. Human pluripotent stem cell-derived cardiomyocytes for heart regeneration, drug discovery and disease modeling: From the genetic, epigenetic, and tissue modeling perspectives. Stem Cell Res Ther. 2013; 4: 97
- 26Dell'Era P, Benzoni P, Crescini E, et al. Cardiac disease modeling using induced pluripotent stem cell–derived human cardiomyocytes. World J Stem Cells. 2015; 7: 329–342.
- 27Mathur A, Loskill P, Shao K, et al. Human iPSC-based cardiac microphysiological system for drug screening applications. Sci Rep. 2015; 5: 8883
- 28Peters MF, Lamore SD, Guo L, Scott CW, Kolaja KL. Human stem cell–derived cardiomyocytes in cellular impedance assays: Bringing cardiotoxicity screening to the front line. Cardiovasc Toxicol. 2015; 15: 127–139.
- 29Mordwinkin NM, Burridge PW, Wu JC. A review of human pluripotent stem cell–derived cardiomyocytes for high-throughput drug discovery, cardiotoxicity screening and publication standards. J Cardiovasc Transl Res. 2014; 6: 22–30.
- 30Yoshida Y, Yamanaka S. Recent stem cell advances: induced pluripotent stem cells for disease modeling and stem cell–based regeneration. Circulation. 2010; 122: 80–87.
- 31Kehat I, Khimovich L, Caspi O, et al. Electromechanical integration of cardiomyocytes derived from human embryonic stem cells. Nat Biotechnol. 2004; 22: 1282–1289.
- 32Hirt MN, Hansen A, Eschenhagen T. Cardiac tissue engineering: State of the art. Circ Res. 2014; 114: 354–367.
- 33Lan F, Lee AS, Liang P, et al. Abnormal calcium handling properties underlie familial hypertrophic cardiomyopathy pathology in patient-specific induced pluripotent stem cells. Cell Stem Cell. 2013; 12: 101–113.
- 34Ojala M, Prajapati C, Pölönen R-P, et al. Mutation-specific phenotypes in hiPSC-derived cardiomyocytes carrying either myosin-binding protein C or α-tropomyosin mutation for hypertrophic cardiomyopathy. Stem Cells Int. 2016; 2016:1684792.
- 35Tse HF, Ho JC, Choi SW, et al. Patient-specific induced-pluripotent stem cells–derived cardiomyocytes recapitulate the pathogenic phenotypes of dilated cardiomyopathy due to a novel DES mutation identified by whole exome sequencing. Hum Mol Genet. 2013; 22: 1395–1403.
- 36Wang G, McCain ML, Yang L, et al. Modeling the mitochondrial cardiomyopathy of Barth syndrome with iPSC and heart-on-chip technologies. Nat Med. 2014; 20: 616–623.
- 37Moretti A, Bellin M, Welling A, et al. Patient-specific induced pluripotent stem-cells models for long-QT syndrome. N Engl J Med. 2010; 363: 981–991.
- 38Itzhaki I, Maizels L, Huber I, et al. Modeling of catecholaminergic polymorphic ventricular tachycardia with patient-specific human-induced pluripotent stem cells. J Am Coll Cardiol. 2012; 60: 990–1000.
- 39Kim C, Wong J, Wen J, et al. Studying arrhythmogenic right ventricular dysplasia with patient-specific iPSCs. Nature. 2013; 494: 105–110.
- 40Carvajal-Vergara X, Sevilla A, D'Souza SL, et al. Patient-specific induced pluripotent stem-cell–derived models of LEOPARD syndrome. Nature. 2010; 465: 808–812.
- 41Lin B, Li Y, Han L, et al. Modeling and studying mechanism of dilated cardiomyopathy using induced pluripotent stem cells derived from Duchenne muscular dystrophy (DMD) patients. Dis Model Mech. 2015. doi: 10.1242/dmm.019505
- 42Drawnel FM, Boccardo S, Prummer M, et al. Disease modeling and phenotypic drug screening for diabetic cardiomyopathy using human induced pluripotent stem cells. Cell Rep. 2014; 9: 810–820.
- 43Wu SM, Hochedlinger K. Harnessing the potential of induced pluripotent stem cells for regenerative medicine. Nat Cell Biol. 2013; 13: 497–505.
- 44Sinnecker D, Laugwitz KL, Moretti A. Induced pluripotent stem cell-derived cardiomyocytes for drug development and toxicity testing. Pharmacol Ther. 2014; 143: 246–252.
- 45Bellin M, Casini S, Davis RP, et al. Isogenic human pluripotent stem cell pairs reveal the role of a KCNH2 mutation in long-QT syndrome. EMBO J. 2013; 32: 3161–3175.
- 46Gore A, Li Z, Fung H-L, et al. Somatic coding mutations in human induced pluripotent stem cells. Nature. 2011; 470: 63–67.
- 47González F, Boué S, Belmonte JCI. Methods for making induced plu-Q11 ripotent stem cells: Reprogramming à la carte. Nat Rev Genet. 2011; 12: 231–242.
- 48Laverty HG, Benson C, Cartwright E, et al. How can we improve our understanding of cardiovascular safety liabilities to develop safer medicines? Br J Pharmacol. 2011; 163: 675–693.
- 49Jurcut R, Wildiers H, Ganame J, D'hooge J, Paridaens R, Voigt JU. Detection and monitoring of cardiotoxicity—what does modern cardiology offer? Support Care Cancer. 2008; 16: 437–445.
- 50Liang P, Lan F, Lee AS, et al. Drug screening using a library of human induced pluripotent stem cell–derived cardiomyocytes reveals disease-specific patterns of cardiotoxicity. Circulation. 2013; 127: 1677–1691.
- 51Guo S, Olm-Shipman A, Walters A, et al. A cell-based phenotypic assay to identify cardioprotective agents. Circ Res. 2012; 110: 948–957.
- 52Segers VFM, Lee RT. Stem-cell therapy for cardiac disease. Nature. 2008; 451: 937–942.
- 53Sanganalmath SK, Bolli R. Cell therapy for heart failure: A comprehensive overview of experimental and clinical studies, current challenges, and future directions. Circ Res. 2013; 113: 810–834.
- 54Garbern JC, Lee RT. Cardiac stem cell therapy and the promise of heart regeneration. Cell Stem Cell. 2013; 12: 689–698.
- 55Orlic D, Hill JM, Arai AE. Stem cells for myocardial regeneration. Circ Res. 2002; 91: 1092–1102.
- 56Ambrosi CM, Entcheva E. Cardiac tissue engineering. Methods Mol Biol. 2014; 1181: 215–228.
- 57Kong CW, Akar FG, Li RA. Translational potential of human embryonic and induced pluripotent stem cells for myocardial repair: Insights from experimental models. Thromb Haemost. 2010; 104: 30–38.
- 58Shiba Y, Filice D, Fernandes S, et al. Electrical integration of human embryonic stem cell–derived cardiomyocytes in a guinea pig chronic infarct model. J Cardiovasc Pharmacol Ther. 2014; 19: 368–381.
- 59Ménard C, Hagège AA, Agbulut O, et al. Transplantation of cardiaccommitted mouse embryonic stem cells to infarcted sheep myocardium: A preclinical study. Lancet. 2005; 366: 1005–1012.
- 60Shiba Y, Fernandes S, Zhu W-Z, et al. Human ES-cell–derived cardiomyocytes electrically couple and suppress arrhythmias in injured hearts. Nature. 2012; 489: 322–325.
- 61Freund C, Mummery CL. Prospects for pluripotent stem cell–derived cardiomyocytes in cardiac cell therapy and as disease models. J Cell Biochem. 2009; 107: 592–599.
- 62Chong JJ, Yang X, Don CW, et al. Human embryonic-stem-cell-derived cardiomyocytes regenerate non-human primate hearts. Nature. 2014; 510: 273–277.
- 63Reinecke H, Minami E, Zhu WZ, Laflamme MA. Cardiogenic differentiation and transdifferentiation of progenitor cells. Circ Res. 2008; 103: 1058–1071.
- 64Yang X, Pabon L, Murry CE. Engineering adolescence: maturation of human pluripotent stem cell–derived cardiomyocytes. Circ Res. 2014; 114: 511–523.
- 65Rao C, Prodromakis T, Kolker L, et al. The effect of microgrooved culture substrates on calcium cycling of cardiac myocytes derived from human induced pluripotent stem cells. Biomaterials. 2013; 34: 2399–2411.
- 66Sartiani L, Bettiol E, Stillitano F, Mugelli A, Cerbai E, Jaconi ME. Developmental changes in cardiomyocytes differentiated from human embryonic stem cells: a molecular and electrophysiological approach. Stem Cells. 2007; 25: 1136–1144.
- 67Bedada FB, Wheelwright M, Metzger JM. Maturation status of sarcomere structure and function in human iPSC-derived cardiac myocytes. Biochim Biophys Acta—Mol Cell Res. 1863; 2016: 1829–1838.
- 68Lundy SD, Zhu WZ, Regnier M, Laflamme MA. Structural and Functional Maturation of Cardiomyocytes Derived from Human Pluripotent Stem Cells. Stem Cells Dev. 2013; 22: 1991–2002.
- 69Rao C, Prodromakis T, Kolker L, et al. The effect of microgrooved culture substrates on calcium cycling of cardiac myocytes derived from human induced pluripotent stem cells. Biomaterials. 2013; 34: 2399–2411.
- 70Lieu DK, Fu JD, Chiamvimonvat N, et al. Mechanism-based facilitated maturation of human pluripotent stem cell–derived cardiomyocytes. Circ Arrhythm Electrophysiol. 2013; 6: 191–201.
- 71Nunes SS, Miklas JW, Radisic M. Maturation of stem cell–derived human heart tissue by mimicking fetal heart rate. Future Cardiol. 2013; 9: 751–754.
- 72Yang X, Rodriguez M, Pabon L, et al. Tri-iodo-l-thyronine promotes the maturation of human cardiomyocytes-derived from induced pluripotent stem cells. J Mol Cell Cardiol. 2014; 72: 296–304.
- 73Ivashchenko CY, Pipers GC, Lozinskaya IM, et al. Human-induced pluripotent stem cell–derived cardiomyocytes exhibit temporal changes in phenotype. Am J Physiol Heart Circ Physiol. 2013; 305: H913–H922.
- 74Pesl M, Pribyl J, Acimovic I, et al. Atomic force microscopy combined with human pluripotent stem cell derived cardiomyocytes for biomechanical sensing. Biosens Bioelectron. 2016; 85: 751–757.
- 75Zhang J, Wilson G, Soerens A. Functional cardiomyocytes derived from human induced pluripotent stem cells. Circ Res. 2009; 104: e30–e41.
- 76He J-Q, Ma Y, Lee Y, Thomson JA, Kamp TJ. Human embryonic stem cells develop into multiple types of cardiac myocytes: Action potential characterization. Circ Res. 2003; 93: 32–39.
- 77Salick MR, Napiwocki BN, Sha J, et al. Micropattern width dependent sarcomere development in human ESC–derived cardiomyocytes. Biomaterials. 2015; 35: 4454–4464.
- 78Yanagi K, Takano M, Narazaki G, et al. Hyperpolarization-activated cyclic nucleotide-gated channels and T-type calcium channels confer automaticity of embryonic stem. Stem Cells. 2007; 25: 2712–2719. doi: 10.1634/stemcells.2006-0388
10.1634/stemcells.2006-0388 Google Scholar
- 79Hoshino S, Omatsu-Kanbe M, Nakagawa M, Matsuura H. Postnatal developmental decline in IK1 in mouse ventricular myocytes isolated by the Langendorff perfusion method: Comparison with the chunk method. Pflugers Arch Eur J Physiol. 2012; 463: 649–668.
- 80Jacot JG, Mcculloch AD, Omens JH. Substrate stiffness affects the functional maturation of neonatal rat ventricular myocytes. Biophys J. 2008; 95: 3479–3487.
- 81Veerman CC, Kosmidis G, Mummery CL, Casini S, Verkerk AO, Bellin M. Immaturity of human stem-cell–derived cardiomyocytes in culture: Fatal flaw or soluble problem? Stem Cells Dev. 2015; 24: 1035–1052.
- 82Snir M, Kehat I, Gepstein A, et al. Assessment of the ultrastructural and proliferative properties of human embryonic stem cell–derived cardiomyocytes. Am J Physiol Heart Circ Physiol. 2003; 285: H2355–H2363. doi: 10.1152/ajpheart.00020.2003
- 83Saffitz JE, Kléber AG. Effects of mechanical forces and mediators of hypertrophy on remodeling of gap junctions in the heart. Circ Res. 2004; 94: 585–591. doi: 10.1161/01.RES.0000121575.34653.50
- 84Rohr S. Role of gap junctions in the propagation of the cardiac action potential. Cardiovasc Res. 2004; 62: 309–322.
- 85Gourdie RG, Harfst E, Severs NJ, Green CR. Cardiac gap junctions in rat ventricle: Localization using site-directed antibodies and laser scanning confocal microscopy. Cardioscience. 1990; 1: 75–82.
- 86Dhein S. Pharmacology of gap junctions in the cardiovascular system. Cardiovasc Res. 2004; 62: 287–298.
- 87Pekkanen-Mattila M, Chapman H, Kerkelä E, et al. Human embryonic stem cell–derived cardiomyocytes: Demonstration of a portion of cardiac cells with fairly mature electrical phenotype. Exp Biol Med (Maywood). 2010; 235: 522–530.
- 88Shaw RM, Rudy Y. Ionic mechanisms of propagation in cardiac tissue: roles of the sodium and L-type calcium currents during reduced excitability and decreased gap junction coupling. Circ Res. 1997; 81: 727–741.
- 89Kucera JP, Rohr S, Rudy Y. Localization of sodium channels in intercalated disks modulates cardiac conduction. Circ Res. 2002; 91: 1176–1182.
- 90Maier SKG, Westenbroek RE, McCormick KA, Curtis R, Scheuer T, Catterall WA. Distinct subcellular localization of different sodium channel α and β subunits in single ventricular myocytes from mouse heart. Circulation. 2004; 109: 1421–1427.
- 91Seidel T, Salameh A, Dhein S. A simulation study of cellular hypertrophy and connexin lateralization in cardiac tissue. Biophys J. 2010; 99: 2821–2830.
- 92Kleber AG, Rudy Y. Basic mechanisms of cardiac impulse propagation and associated arrhythmias. Physiol Rev. 2004; 84: 431–488.
- 93Babiarz JE, Ravon M, Sridhar S, et al. Determination of the human cardiomyocyte mRNA and miRNA differentiation network by fine-scale profiling. Stem Cells Dev. 2012; 21: 1956–1965.
- 94Gupta MK, Illich DJ, Gaarz A, et al. Global transcriptional profiles of beating clusters derived from human induced pluripotent stem cells and embryonic stem cells are highly similar. BMC Dev Biol. 2010; 10: 98.
- 95Wamstad JA, Alexander JM, Truly RM, et al. Dynamic and coordinated epigenetic regulation of developmental transitions in the cardiac lineage. Cell. 2012; 151: 206–220.
- 96Paige SL, Thomas S, Stoick-Cooper CL, et al. A temporal chromatin signature in human embryonic stem cells identifies regulators of cardiac development. Cell. 2012; 151: 221–232.
- 97Aggarwal P, Turner A, Matter A, et al. RNA expression profiling of human iPSC-derived cardiomyocytes in a cardiac hypertrophy model. PLoS One. 2014; 9: 1–10.
- 98McGivern JV, Ebert AD. Exploiting pluripotent stem cell technology for drug discovery, screening, safety, and toxicology assessments. Adv Drug Deliv Rev. 2014; 69-70: 170–178.
- 99Aravanis AM, DeBusschere BD, Chruscinski AJ, Gilchrist KH, Kobilka BK, Kovacs GT. A genetically engineered cell-based biosensor for functional classification of agents. Biosens Bioelectron. 2001; 16: 571–577.
- 100Edmondson R, Broglie JJ, Adcock AF, Yang L. Three-dimensional cell culture systems and their applications in drug discovery and cell-based biosensors. Assay Drug Dev Technol. 2014; 12: 207–218.
- 101Cheng MS, Lau SH, Chan KP, Toh CS, Chow VT. Impedimetric cellbased biosensor for real-time monitoring of cytopathic effects induced by dengue viruses. Biosens Bioelectron. 2015; 70: 74–80.
- 102Zhang L, Yang F, Cai J-Y, Yang P-H, Liang Z-H. In-situ detection of resveratrol inhibition effect on epidermal growth factor receptor of living MCF-7 cells by atomic force microscopy. Biosens Bioelectron. 2014; 56: 271–277.
- 103Rodriguez-Mozaz S, Lopez de Alda MJ, Marco M-P, Barcelò D. Biosensors for environmental: A global perspective. Talanta. 2005; 65: 291–297.
- 104Qureshi A, Pandey A, Chouhan RS, Gurbuz Y, Niazi JH. Whole-cell based label-free capacitive biosensor for rapid nanosize-dependent toxicity detection. Biosens Bioelectron. 2015; 67: 100–106.
- 105Smirnov MN, Smirnov VN, Budowsky EI, Inge-Vechtomov SG, Serebrjakov NG. Red pigment of adenine-deficient yeast Saccharomyces cerevisiae. Biochem Biophys Res Commun. 1967; 27: 299–304.
- 106Wang T, Hu N, Cao J, Wu J, Su K, Wang P. A cardiomyocyte-based biosensor for antiarrhythmic drug evaluation by simultaneously monitoring cell growth and beating. Biosens Bioelectron. 2013; 49: 9–13.
- 107Hamill OP, Marty A, Neher E, Sakmann B, Sigworth FJ. Improved patch-clamp techniques for high-resolution current recording from cells and cell-free membrane patches. Pflugers Arch. 1981; 391: 85–100.
- 108Monyer H, Lambolez B. Molecular biology and physiology at the single-cell level. Curr Opin Neurobiol. 1995; 5: 382–387.
- 109Dixon AK, Richardson PJ, Pinnock RD, Lee K. Gene-expression analysis at the single-cell level. Trends Pharmacol Sci. 2000; 21: 65–70.
- 110Neher E, Sakmann B, Steinbach JH. The extracellular patch clamp: A method for resolving currents through individual open channels in biological membranes. Eur J Physiol. 1978; 228: 219–228.
- 111Scheel O, Frech S, Amuzescu B, Eisfeld J, Lin KH, Knott T. Action potential characterization of human induced pluripotent stem cell– derived cardiomyocytes using automated patch-clamp technology. Assay Drug Dev Technol. 2014; 12: 457–469.
- 112Cao F, Wagner RA, Wilson KD, et al. Transcriptional and functional profiling of human embryonic stem cell-derived cardiomyocytes. PLoS One. 2008; 3:e3474.
- 113Holt GR, Koch C. Electrical interactions via the extracellular potential near cell bodies. J Comput Neurosci. 1999; 6: 169–184.
- 114Stett A, Egert U, Guenther E, et al. Biological application of microelectrode arrays in drug discovery and basic research. Anal Bioanal Chem. 2003; 377: 486–495.
- 115Frega M, Tedesco M, Massobrio P, Pesce M, Martinoia S. Network dynamics of 3D engineered neuronal cultures: A new experimental model for in-vitro electrophysiology. Sci Rep. 2014; 4: 5489.
- 116Banach K, Halbach MD, Hu P, Hescheler J, Egert U. Development of electrical activity in cardiac myocyte aggregates derived from mouse embryonic stem cells. Am J Physiol Heart Circ Physiol. 2003; 284: H2114–H2123.
- 117Besl B, Fromherz P. Transistor array with an organotypic brain slice: field potential records and synaptic currents. Eur J Neurosci. 2002; 15: 999–1005.
- 118Kanagasabapathi TT, Massobrio P, Barone RA, et al. Functional connectivity and dynamics of cortical-thalamic networks co-cultured in a dual compartment device. J Neural Eng. 2012; 9: 36010.
- 119Sigg DC, Iaizzo PA, Xiao Y-F, He B. Cardiac Electrophysiology Methods and Models. Boston, MA, US: Springer Science + Business Media; 2010. doi: 10.1007/978-1-4419-6658-2
- 120Saric T, Halbach M, Khalil M, Er F. Induced pluripotent stem cells as cardiac arrhythmic in vitro models and the impact for drug discovery. Expert Opin Drug Discovery. 2014; 9: 55–76.
- 121Navarrete EG, Liang P, Lan F, et al. Screening drug-induced arrhythmia [corrected] using human induced pluripotent stem cell-derived cardiomyocytes and low-impedance microelectrode arrays. Circulation. 2013; 128: S3–S13.
- 122Asai Y, Tada M, Otsuji TG, Nakatsuji N. Combination of functional cardiomyocytes derived from human stem cells and a highly-efficient microelectrode array system: an ideal hybrid model assay for drug development. Curr Stem Cell Res Ther. 2010; 5: 227–232.
- 123Giaever I, Keese CR. Monitoring fibroblast behavior in tissue culture with an applied electric field. Proc Natl Acad Sci U S A. 1984; 81: 3761–3764.
- 124Qiu Y, Liao R, Zhang X. Real-time monitoring primary cardiomyocyte adhesion based on electrochemical impedance spectroscopy and electrical cell-substrate impedance sensing. Anal Chem. 2008; 80: 990–996.
- 125Doherty KR, Talbert DR, Trusk PB, Moran DM, Shell SA, Bacus S. Structural and functional screening in human induced-pluripotent stem cell-derived cardiomyocytes accurately identifies cardiotoxicity of multiple drug types. Toxicol Appl Pharmacol. 2015; 285: 51–60.
- 126Guo L, Abrams RM, Babiarz JE, et al. Estimating the risk of druginduced proarrhythmia using human induced pluripotent stem cell derived cardiomyocytes. Toxicol Sci. 2011; 4077: 1–29.
- 127Scott CW, Zhang X, Abi-Gerges N, Lamore SD, Abassi YA, Peters MF. An impedance-based cellular assay using human iPSC-derived cardiomyocytes to quantify modulators of cardiac contractility. Toxicol Sci. 2014; 142: 331–338.
- 128Efimov IR, Nikolski VP, Salama G. Optical imaging of the heart. Circ Res. 2004; 95: 21–33.
- 129Radaszkiewicz KA, Sýkorová D, Karas P, et al. Simple non-invasive analysis of embryonic stem cell-derived cardiomyocytes beating in vitro. Rev Sci Instrum. 2016; 87: 024301.
- 130Peterson P, Kalda M, Vendelin M. Real-time determination of sarcomere length of a single cardiomyocyte during contraction. Am J Physiol Cell Physiol. 2013; 304: C519–C531.
- 131Kamgoué A, Ohayon J, Usson Y, Riou L, Tracqui P. Quantification of cardiomyocyte contraction based on image correlation analysis. Cytom Part A. 2009; 75: 298–308.
- 132London B, Krueger JW. Contraction in voltage-clamped, internally perfused single heart cells. J Gen Physiol. 1986; 88: 475–505.
- 133Odstrcilik J, Cmiel V, Kolar R, et al. Computer analysis of isolated cardiomyocyte contraction process via advanced image processing techniques. 2015Computing Cardiol Confer (CinC). 2015; 453–456. doi: 10.1109/CIC.2015.7408684
- 134Herron TJ, Lee P, Jalife J. Optical imaging of voltage and calcium in cardiac cells & tissues. Circ Res. 2012; 110: 609–623.
- 135Hayakawa T, Kunihiro T, Ando T, et al. 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: 178–191.
- 136Rappaz B, Moon I, Yi F, Javidi B, Marquet P, Turcatti G. Automated multi-parameter measurement of cardiomyocytes dynamics with digital holographic microscopy. Opt Express. 2015; 23: 13333–13347.
- 137Grienberger C, Konnerth A. Imaging calcium in neurons. Neuron. 2012; 73: 862–885.
- 138Melzer W, Herrmann-Frank A, Lüttgau HC. The role of Ca2+ ions in excitation-contraction coupling of skeletal muscle fibres. Biochim Biophys Acta—Rev Biomembr. 1995; 1241: 59–116.
- 139Roderick HL, Berridge MJ, Bootman MD. Calcium-induced calcium release. Curr Biol. 2003; 13: R425.
- 140Swandulla D, Hans M, Zipser K, Augustine GJ. Role of residual calcium in synaptic depression and posttetanic potentiation: Fast and slow calcium signaling in nerve terminals. Neuron. 1991; 7: 915–926.
- 141Lasser-Ross N, Miyakawa H, Lev-Ram V, Young SR, Ross WN. High time resolution fluorescence imaging with a CCD camera. J Neurosci Methods. 1991; 36: 253–261.
- 142Bigas M, Cabruja E, Forest J, Salvi J. Review of CMOS image sensors. Microchem J. 2006; 37: 433–451.
- 143Sauer H, Theben T, Hescheler J, Lindner M, Brandt MC, Wartenberg M. Characteristics of calcium sparks in cardiomyocytes derived from embryonic stem cells. Am J Physiol Heart Circ Physiol. 2001; 281: H411–H421.
- 144Fabiato A. Calcium-induced release of calcium from the cardiac sarcoplasmic reticulum. Am J Physiol. 1983; 245: C1–C14.
- 145Lieu DK, Liu J, Siu CW, et al. Absence of transverse tubules contributes to non-uniform Ca2+ wavefronts in mouse and human embryonic stem cell–derived cardiomyocytes. Stem Cells Dev. 2009; 18: 1493–1500.
- 146Satin J, Itzhaki I, Rapoport S, et al. Calcium handling in human embryonic stem cell–derived cardiomyocytes. Stem Cells. 2008; 26: 1961–1972.
- 147Lee Y-K, Ng KM, Lai WH, et al. Calcium homeostasis in human induced pluripotent stem cell–derived cardiomyocytes. Stem Cell Rev. 2011; 7: 976–986.
- 148Dempsey GT, Chaudhary KW, Atwater N, et al. Cardiotoxicity screening with simultaneous optogenetic pacing, voltage imaging and calcium imaging. J Pharmacol Toxicol Methods. 2011; doi: 10.1016/j. vascn.2016.05.003
- 149Bräuner T, Hülser DF, Strasser RJ. Comparative measurements of membrane potentials with microelectrodes and voltage-sensitive dyes. Biochim Biophys Acta—Biomembr. 1984; 771: 208–216.
- 150Xue T, Cho HC, Akar FG, et al. Functional integration of electrically active cardiac derivatives from genetically engineered human embryonic stem cells with quiescent recipient ventricular cardiomyocytes: Insights into the development of cell-based pacemakers. Circulation. 2005; 111: 11–20.
- 151Zhang J, Klos M, Wilson GF, et al. Extracellular matrix promotes highly efficient cardiac differentiation of human pluripotent stem cells: The matrix sandwich method. Circ Res. 2012; 111: 1125–1136.
- 152Karakikes I, Senyei GD, Hansen J, et al. Small molecule–mediated directed differentiation of human embryonic stem cells toward ventricular cardiomyocytes. Stem Cells Transl Med. 2014; 3: 18–31.
- 153Zapletalová H, Přibyl J, Ambrož M, Vůjtek M, Skládal P, Kolářová H. Improved method for Mica functionalization used in single molecule imaging of DNA with atomic force microscopy. Mediterr J Chem. 2016; 5: 589–598.
10.13171/mjc55/01607041620/zapletalova Google Scholar
- 154Vahabi S, Nazemi Salman B, Javanmard A. Atomic force microscopy application in biological research: A review study. Iran J Med Sci. 2013; 38: 76–83.
- 155Sundararajan S, Bhushan B. Development of AFM-based techniques to measure mechanical properties of nanoscale structures. Sensors Actuators A Phys. 2002; 101: 338–351.
- 156Wang C, Kim J, Zhu Y, et al. An aptameric graphene nanosensor for label-free detection of small-molecule biomarkers. Biosens Bioelectron. 2015; 71: 222–229.
- 157Ossola D, Amarouch M-Y, Behr P, Vörös J, Abriel H, Zambelli T. Forcecontrolled patch clamp of beating cardiac cells. Nano Lett. 2015; 15: 1743–1750.
- 158Obataya I, Nakamura C, Han S, Nakamura N, Miyake J. Mechanical sensing of the penetration of various nanoneedles into a living cell using atomic force microscopy. 2005; 20: 1652–1655.
- 159Wang J, Wan Z, Liu W, et al. Atomic force microscope study of tumor cell membranes following treatment with anti-cancer drugs. Biosens Bioelectron. 2009; 25: 721–727.
- 160Liu J, Sun N, Bruce MA, Wu JC, Butte MJ. Atomic force mechanobiology of pluripotent stem cell–derived cardiomyocytes. PLoS One. 2012; 7:e37559.
- 161Pesl M, Acimovic I, Pribyl J, et al. Molecular and functional characterization of uniform-sized beating embryoid bodies and cardiomyocytes from human embryonic and induced pluripotent stem cells. Biophys J. 2014; 106: 565A–565A.
- 162Cogollo JFS, Tedesco M, Martinoia S, Raiteri R. A new integrated system combining atomic force microscopy and microelectrode array for measuring the mechanical properties of living cardiac myocytes. 2011; 613–621. doi: 10.1007/s10544-011-9531-9
- 163Raiteri R, Caluori G, Tedesco M, Andrade Caicedo HH, Ward CW. A novel AFM-based technique for measuring coupled phenomena in cardiac myocytes. Biophys J. 2015; 108–168a.
- 164Tan Y, Sun D, Cheng SH, Li RA. Robotic cell manipulation with optical tweezers for biomechanical characterization. in 2011 IEEE International Conference on Robotics and Automation (ICRA). 2011; 4104–4109. doi: 10.1109/ICRA.2011.5979577
10.1109/ICRA.2011.5979577 Google Scholar
- 165Laurent VM, Hénon S, Planus E, et al. Assessment of mechanical properties of adherent living cells by bead micromanipulation: Comparison of magnetic twisting cytometry vs optical tweezers. J Biomech Eng. 2002; 124: 408–421.
- 166Tan Y, Kong CW, Chen S, Cheng SH, Li RA, Sun D. Probing the mechanobiological properties of human embryonic stem cells in cardiac differentiation by optical tweezers. J Biomech. 2012; 45: 123–128.
- 167Rodriguez ML, Graham BT, Pabon LM, Han SJ, Murry CE, Sniadecki NJ. Measuring the contractile forces of human induced pluripotent stem cell-derived cardiomyocytes with arrays of microposts. J Biomech Eng. 2014; 136: 051005.
- 168Pribyl J, Skladal P. Quartz crystal biosensor for detection of sugars and glycated hemoglobin. Anal Chim Acta. 2005; 530: 75–84.
- 169Fohlerová Z, Skládal P, Turánek J. Adhesion of eukaryotic cell lines on the gold surface modified with extracellular matrix proteins monitored by the piezoelectric sensor. Biosens Bioelectron. 2007; 22: 1896–1901.
- 170Fohlerová Z, Turánek J, Skládal P. The cell adhesion and cytotoxicity effects of the derivate of vitamin E compared for two cell lines using a piezoelectric biosensor. Sens Actuators B. 2012; 174: 153–157.
- 171Fredriksson C, Khilman S, Kasemo B, Steel DM. In vitro real-time characterization of cell attachment and spreading. J Mater Sci Mater Med. 1998; 9: 785–788.
- 172Tymchenko N, Nilebäck E, Voinova MV, Gold J, Kasemo B, Svedhem S. Reversible changes in cell morphology due to cytoskeletal rearrangements measured in real-time by QCM-D. Biointerphases. 2012; 7: 43.
- 173Elmlund L, Käck C, Aastrup T, Nicholls IA. Study of the interaction of trastuzumab and SKOV3 epithelial cancer cells using a quartz crystal microbalance sensor. Sensors. 2015; 15: 5884–5894.
- 174Wang G, Dewilde AH, Zhang J, et al. A living cell quartz crystal microbalance biosensor for continuous monitoring of cytotoxic responses of macrophages to single-walled carbon nanotubes. Part Fibre Toxicol. 2011; 8: 4.
- 175Tymchenko N, Kunze A, Dahlenborg K, Svedhem S, Steel D. Acoustical sensing of cardiomyocyte cluster beating. Biochem Biophys Res Commun. 2013; 435: 520–525.
- 176Otsuji TG, Minami I, Kurose Y, Yamauchi K, Tada M, Nakatsuji N. Progressive maturation in contracting cardiomyocytes derived from human embryonic stem cells: Qualitative effects on electrophysiological responses to drugs. Stem Cell Res. 2010; 4: 201–213.
- 177Andersson H, Steel D, Asp J, et al. Assaying cardiac biomarkers for toxicity testing using biosensing and cardiomyocytes derived from human embryonic stem cells. J Biotechnol. 2010; 150: 175–181.
- 178Singal PK, Iliskovic N. Doxorubicin-induced cardiomyopathy. N Engl J Med. 1998; 339: 900–905.