Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Metabolically driven maturation of human-induced-pluripotent-stem-cell-derived cardiac microtissues on microfluidic chips

Abstract

The immature physiology of cardiomyocytes derived from human induced pluripotent stem cells (hiPSCs) limits their utility for drug screening and disease modelling. Here we show that suitable combinations of mechanical stimuli and metabolic cues can enhance the maturation of hiPSC-derived cardiomyocytes, and that the maturation-inducing cues have phenotype-dependent effects on the cells’ action-potential morphology and calcium handling. By using microfluidic chips that enhanced the alignment and extracellular-matrix production of cardiac microtissues derived from genetically distinct sources of hiPSC-derived cardiomyocytes, we identified fatty-acid-enriched maturation media that improved the cells’ mitochondrial structure and calcium handling, and observed divergent cell-source-dependent effects on action-potential duration (APD). Specifically, in the presence of maturation media, tissues with abnormally prolonged APDs exhibited shorter APDs, and tissues with aberrantly short APDs displayed prolonged APDs. Regardless of cell source, tissue maturation reduced variabilities in spontaneous beat rate and in APD, and led to converging cell phenotypes (with APDs within the 300–450 ms range characteristic of human left ventricular cardiomyocytes) that improved the modelling of the effects of pro-arrhythmic drugs on cardiac tissue.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Optimized hiPSC-CM MPS.
Fig. 2: Action-potential characterization of matured cardiac MPS.
Fig. 3: Mitochondrial morphology of MPS treated with MM.
Fig. 4: Inotropic response of MM-treated MPS.
Fig. 5: Gene expression analysis of monolayers and MPS treated with lipid-based MM.
Fig. 6: Mathematical modelling of the contribution of individual currents and calcium handling machinery to the action potential of monolayers and MPS.
Fig. 7: Pro-arrhythmia pharmacology of matured WTC cardiac MPS.
Fig. 8: Pro-arrhythmia pharmacology of matured SCVI20 cardiac MPS.

Similar content being viewed by others

Data availability

The main data supporting the results in this study are available within the paper and its Supplementary Information. All raw and analysed datasets generated during the study are available from the corresponding author on request. Source data are provided with this paper.

Code availability

The microphysiological systems and monolayer physiology were analysed with an open-source motion-tracking software available for download at huebschlab.wustl.edu. Calcium transients, action-potential waveforms and sarcomere regularity were analysed with in-house code that is available on request from the corresponding author.

References

  1. Scannell, J. W., Blanckley, A., Boldon, H. & Warrington, B. Diagnosing the decline in pharmaceutical R&D efficiency. Nat. Rev. Drug Discov. 11, 191–200 (2012).

    Article  CAS  PubMed  Google Scholar 

  2. Musunuru, K. et al. Induced pluripotent stem cells for cardiovascular disease modeling and precision medicine: a scientific statement from the American Heart Association. Circ-Genom Precis Me. 11, e000043 (2018).

    Google Scholar 

  3. Paci, M., Hyttinen, J., Rodriguez, B. & Severi, S. Human induced pluripotent stem cell-derived versus adult cardiomyocytes: an in silico electrophysiological study on effects of ionic current block. Br. J. Pharmacol. 172, 5147–5160 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Johnson, E. K. et al. Differential expression and remodeling of transient outward potassium currents in human left ventricles. Circ. Arrhyth. Electrophysiol. 11, e005914 (2018).

    Article  CAS  Google Scholar 

  5. Kuppusamy, K. T. et al. Let-7 family of microRNA is required for maturation and adult-like metabolism in stem cell-derived cardiomyocytes. Proc. Natl Acad. Sci. USA 112, E2785–E2794 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Lundy, S. D., Zhu, W. Z., Regnier, M. & Laflamme, M. A. Structural and functional maturation of cardiomyocytes derived from human pluripotent stem cells. Stem. Cells Dev. 22, 1991–2002 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Mills, R. J. et al. Functional screening in human cardiac organoids reveals a metabolic mechanism for cardiomyocyte cell cycle arrest. Proc. Natl Acad. Sci. USA, 114(40): E8373–E8381 (2017).

  8. Zhang, J. et al. Functional cardiomyocytes derived from human induced pluripotent stem cells. Circ. Res. 104, 30–41 (2009).

    Article  Google Scholar 

  9. Iseoka, H. et al. Pivotal role of non-cardiomyocytes in electromechanical and therapeutic potential of induced pluripotent stem cell-derived engineered cardiac tissue. Tissue Eng. Part A 24, 287–300 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Lemoine, M. D. et al. Human iPSC-derived cardiomyocytes cultured in 3D engineered heart tissue show physiological upstroke velocity and sodium current density. Sci. Rep. 7, Article number: 5464 (2017).

  11. Ma, J. et al. High purity human-induced pluripotent stem cell-derived cardiomyocytes: electrophysiological properties of action potentials and ionic currents. Am. J. Physiol. Heart Circ. Physiol. 301, H2006–H2017 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Spencer, C. I. et al. Calcium transients closely reflect prolonged action potentials in iPSC models of inherited cardiac arrhythmia. Stem Cell Rep. 3, 269–281 (2014).

    Article  CAS  Google Scholar 

  13. Liang, P. et al. Drug screening using a library of human induced pluripotent stem cell-derived cardiomyocytes reveals disease-specific patterns of cardiotoxicity. Circulation 127, 1677–1691 (2013).

    Article  CAS  PubMed  Google Scholar 

  14. Shadrin, I. Y. et al. Cardiopatch platform enables maturation and scale-up of human pluripotent stem cell-derived engineered heart tissues. Nat. Comm. https://doi.org/10.1038/s41467-017-01946-x (2017).

  15. Huebsch, N. et al. Miniaturized iPS-cell-derived cardiac muscles for physiologically relevant drug response analyses. Sci. Rep. 6, 1–12 (2016).

    Article  CAS  Google Scholar 

  16. Mannhardt, I. et al. Human engineered heart tissue: analysis of contractile force. Stem Cell Rep. 7, 29–42 (2016).

    Article  CAS  Google Scholar 

  17. Godier-Furnémont, A. F. G. et al. Physiologic force-frequency response in engineered heart muscle by electromechanical stimulation. Biomaterials 60, 82–91 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Hinson, J. T. et al. Titin mutations in iPS cells define sarcomere insufficiency as a cause of dilated cardiomyopathy. Science 349, 982–986 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Zhang, D. et al. Tissue-engineered cardiac patch for advanced functional maturation of human ESC-derived cardiomyocytes. Biomaterials 34, 5813–5820 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Nunes, S. S. et al. Biowire: a platform for maturation of human pluripotent stem cell-derived cardiomyocytes. Nat. Methods 10, 781–787 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Tulloch, N. L. et al. Growth of engineered human myocardium with mechanical loading and vascular coculture. Circ. Res. 109, 47–59 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Tiburcy, M. et al. Terminal differentiation, advanced organotypic maturation, and modeling of hypertrophic growth in engineered heart tissue. Circ. Res. 109, 1105–1114 (2011).

    Article  CAS  PubMed  Google Scholar 

  23. Zimmermann, W. H. et al. Tissue engineering of a differentiated cardiac muscle construct. Circ. Res. 90, 223–230 (2002).

    Article  CAS  PubMed  Google Scholar 

  24. Mathur, A. et al. Human iPSC-based cardiac microphysiological system for drug screening applications. Sci. Rep. https://doi.org/10.1038/srep08883 (2015).

  25. Charrez, B. et al. Heart muscle microphysiological system for cardiac liability prediction of repurposed COVID-19 therapeutics. Front. Pharmacol. https://doi.org/10.3389/fphar.2021.684252 (2021).

  26. Charrez, B. et al. In vitro safety “clinical trial” of the cardiac liability of drug polytherapy. Clin. Transl. Sci. 14, 1155–1165 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Jæger, K. H., Charwat, V., Wall, S., Healy, K. E. & Tveito, A. Identifying drug response by combining measurements of the membrane potential, the cytosolic calcium concentration, and the extracellular potential in microphysiological systems. Front. Pharmacol. https://doi.org/10.3389/fphar.2020.569489 (2021).

  28. Tveito, A. et al. Inversion and computational maturation of drug response using human stem cell derived cardiomyocytes in microphysiological systems. Sci. Rep. https://doi.org/10.1038/s41598-018-35858-7 (2018).

  29. Ronaldson-Bouchard, K. et al. Advanced maturation of human cardiac tissue grown from pluripotent stem cells. Nature 556, 239–243 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Ruan, J.-L. et al. Mechanical stress promotes maturation of human myocardium from pluripotent stem cell-derived progenitors. Stem Cells 33, 2148–2157 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Feyen, D. A. M. et al. Metabolic maturation media improve physiological function of human iPSC-derived cardiomyocytes. Cell Rep. 32, 107925 (2020).

    Article  CAS  PubMed  Google Scholar 

  32. Kadota, S., Pabon, L., Reinecke, H. & Murry, C. E. In vivo maturation of human induced pluripotent stem cell-derived cardiomyocytes in neonatal and adult rat hearts. Stem Cell Rep. 8, 278–289 (2017).

    Article  CAS  Google Scholar 

  33. Rupert, C. E. & Coulombe, K. L. K. IGF1 and NRG1 enhance proliferation, metabolic maturity, and the force-frequency response in hesc-derived engineered cardiac tissues. Stem Cells Int. https://doi.org/10.1155/2017/7648409 (2017).

  34. Tiburcy, M. et al. Defined engineered human myocardium with advanced maturation for applications in heart failure modeling and repair. Circulation 135, 1832–1847 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Fong, A. H. et al. Three-dimensional adult cardiac extracellular matrix promotes maturation of human induced pluripotent stem cell-derived cardiomyocytes. Tissue Eng. Part A 22, 1016–1025 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Yang, X. et al. Tri-iodo-l-thyronine promotes the maturation of human cardiomyocytes-derived from induced pluripotent stem cells. J. Mol. Cell. Cardiol. 72, 296–304 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Lopaschuk, G. D. & Jaswal, J. S. Energy metabolic phenotype of the cardiomyocyte during development, differentiation, and postnatal maturation. J. Cardiovasc. Pharmacol. 56, 130–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  38. Makinde, A.-O., Kantor, P. F. & Lopaschuk, G. D. in Molecular and Cellular Effects of Nutrition on Disease Processes (eds Pierce, G. N. et al.) 49–56 (Springer, 1998).

  39. Correia, C. et al. Distinct carbon sources affect structural and functional maturation of cardiomyocytes derived from human pluripotent stem cells. Sci. Rep. 7, 8590 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Rana, P., Anson, B., Engle, S. & Will, Y. Characterization of human-induced pluripotent stem cell-derived cardiomyocytes: bioenergetics and utilization in safety screening. Toxicol. Sci. 130, 117–131 (2012).

    Article  CAS  PubMed  Google Scholar 

  41. Matsa, E. et al. Transcriptome profiling of patient-specific human iPSC-cardiomyocytes predicts individual drug safety and efficacy responses in vitro. Cell Stem Cell 19, 311–325 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Huebsch, N. et al. Automated video-based analysis of contractility and calcium flux in human-induced pluripotent stem cell-derived cardiomyocytes cultured over different spatial scales. Tissue Eng. Part C 21, 467–479 (2015).

    Article  CAS  Google Scholar 

  43. Sun, N. et al. Patient-specific induced pluripotent stem cells as a model for familial dilated cardiomyopathy. Sci. Transl. Med. 4, 130ra147 (2012).

    Article  Google Scholar 

  44. Jha, A. K., Jackson, W. M. & Healy, K. E. Controlling osteogenic stem cell differentiation via soft bioinspired hydrogels. PLoS ONE 9, e98640 (2014).

  45. Stile, R. A., Barber, T. A., Castner, D. G. & Healy, K. E. Sequential robust design methodology and X-ray photoelectron spectroscopy to analyze the grafting of hyaluronic acid to glass substrates. J. Biomed. Mater. Res. 61, 391–398 (2002).

    Article  CAS  PubMed  Google Scholar 

  46. Taguchi, G. & Phadke, M. S. in Quality Control, Robust Design, and the Taguchi Method (ed. Dehnad, K.) 77–96 (Springer, 1989).

  47. Kitani, T. et al. Human-induced pluripotent stem cell model of trastuzumab-induced cardiac dysfunction in patients with breast cancer. Circulation 139, 2451–2465 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Zapata-Linares, N. et al. Generation and characterization of human iPSC line generated from mesenchymal stem cells derived from adipose tissue. Stem Cell Res. 16, 20–23 (2016).

    Article  CAS  PubMed  Google Scholar 

  49. Naito, H. et al. Optimizing engineered heart tissue for therapeutic applications as surrogate heart muscle. Circulation 114, 72–78 (2006).

    Article  Google Scholar 

  50. Wang, G. et al. Modeling the mitochondrial cardiomyopathy of Barth syndrome with induced pluripotent stem cell and heart-on-chip technologies. Nat. Med. 20, 616–623 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Naim, J. O., Ippolito, K. M. L., Lanzafame, R. J. & van Oss, C. J. The effect of molecular weight and gel preparation on humoral adjuvancy of silicone oils and silicone gels. Immunol. Investig. 24, 537–547 (1995).

    Article  CAS  Google Scholar 

  52. Park, E.-J., Lee, A. Y., Park, S., Kim, J.-H. & Cho, M.-H. Multiple pathways are involved in palmitic acid-induced toxicity. Food Chem. Toxicol. 67, 26–34 (2014).

    Article  CAS  PubMed  Google Scholar 

  53. Bodi, I., Mikala, G., Koch, S. E., Akhter, S. A. & Schwartz, A. The L-type calcium channel in the heart: the beat goes on. J. Clin. Investig. 115, 3306–3317 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Liu, R. et al. Palmitoylation regulates intracellular trafficking of β2 adrenergic receptor/arrestin/phosphodiesterase 4D complexes in cardiomyocytes. PLoS ONE 7, e42658 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Medda, L., Monduzzi, M. & Salis, A. The molecular motion of bovine serum albumin under physiological conditions is ion specific. Chem. Commun. 51, 6663–6666 (2015).

    Article  CAS  Google Scholar 

  56. Brandenburger, M. et al. Organotypic slice culture from human adult ventricular myocardium. Cardiovasc. Res. 93, 50–59 (2012).

    Article  CAS  PubMed  Google Scholar 

  57. Chen, T.-W. et al. Ultrasensitive fluorescent proteins for imaging neuronal activity. Nature 499, 295–300 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Paredes, R. M., Etzler, J. C., Watts, L. T., Zheng, W. & Lechleiter, J. D. Chemical calcium indicators. Methods 46, 143–151 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Mot, A. I., Liddell, J. R., White, A. R. & Crouch, P. J. Circumventing the Crabtree Effect: a method to induce lactate consumption and increase oxidative phosphorylation in cell culture. Int. J. Biochem. Cell Biol. 79, 128–138 (2016).

    Article  CAS  PubMed  Google Scholar 

  60. Anmann, T. et al. Formation of highly organized intracellular structure and energy metabolism in cardiac muscle cells during postnatal development of rat heart. Biochim. Biophys. Acta 1837, 1350–1361 (2014).

    Article  CAS  PubMed  Google Scholar 

  61. Ma, Z. et al. Contractile deficits in engineered cardiac microtissues as a result of MYBPC3 deficiency and mechanical overload. Nat. Biomed. Eng. 2, 955–967 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Lee, J. H., Protze, S. I., Laksman, Z., Backx, P. H. & Keller, G. M. Human pluripotent stem cell-derived atrial and ventricular cardiomyocytes develop from distinct mesoderm populations. Cell Stem Cell 21, 179–194.e4 (2017).

    Article  CAS  PubMed  Google Scholar 

  63. Zhang, J. Z. et al. A human iPSC double-reporter system enables purification of cardiac lineage subpopulations with distinct function and drug response profiles. Cell Stem Cell 24, 802–811.e5 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Campbell, K. L. & Dicke, A. A. Sarcolipin makes heat, but is it adaptive thermogenesis? Front. Physiol. https://doi.org/10.3389/fphys.2018.00714 (2018).

  65. Smith, W. S., Broadbridge, R., East, J. M. & Lee, A. G. Sarcolipin uncouples hydrolysis of ATP from accumulation of Ca2+ by the Ca2+-ATPase of skeletal-muscle sarcoplasmic reticulum. Biochem. J. 361, 277–286 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Mall, S. et al. The presence of sarcolipin results in increased heat production by Ca(2+)-ATPase. J. Biol. Chem. 281, 36597–36602 (2006).

    Article  CAS  PubMed  Google Scholar 

  67. Gorski, P. A., Ceholski, D. K. & Young, H. S. in Membrane Dynamics and Calcium Signaling (ed. Krebs, J.) 77–119 (Springer, 2017).

  68. Josowitz, R. et al. Identification and purification of human induced pluripotent stem cell-derived atrial-like cardiomyocytes based on sarcolipin expression. PLoS ONE 9, e101316 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  69. Schwach, V. et al. A COUP-TFII human embryonic stem cell reporter line to identify and select atrial cardiomyocytes. Stem Cell Rep. 9, 1765–1779 (2017).

    Article  CAS  Google Scholar 

  70. Venetucci, L., Denegri, M., Napolitano, C. & Priori, S. G. Inherited calcium channelopathies in the pathophysiology of arrhythmias. Nat. Rev. Cardiol. 9, 561–575 (2012).

    Article  CAS  PubMed  Google Scholar 

  71. Buraei, Z. & Yang, J. The β subunit of voltage-gated Ca2+ channels. Physiol. Rev. 90, 1461–1506 (2010).

    Article  CAS  PubMed  Google Scholar 

  72. Rees, C. M. et al. The Ca(2+) transient as a feedback sensor controlling cardiomyocyte ionic conductances in mouse populations. eLife https://doi.org/10.7554/eLife.36717 (2018).

  73. Sarkar, A. X., Christini, D. J. & Sobie, E. A. Exploiting mathematical models to illuminate electrophysiological variability between individuals. J. Physiol. 590, 2555–2567 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Jaeger, K. H., Wall, S. & Tveito, A. Detecting undetectables: can conductances of action potential models be changed without appreciable change in the transmembrane potential? Chaos 29, 073102 (2019).

    Article  PubMed  CAS  Google Scholar 

  75. Jaeger, K. H. et al. Improved computational identification of drug response using optical measurements of human stem cell derived cardiomyocytes in microphysiological systems. Front. Pharmacol. 10, 1648 (2019).

    Article  CAS  PubMed  Google Scholar 

  76. Jæger, K. H., Wall, S. & Tveito, A. Computational prediction of drug response in short QT syndrome type 1 based on measurements of compound effect in stem cell-derived cardiomyocytes. PLoS Comput. Biol. 17(2), (2021).

  77. Eisner, D. A., Caldwell, J. L., Kistamas, K. & Trafford, A. W. Calcium and excitation-contraction coupling in the heart. Circ. Res. 121, 181–195 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Parikh, S. S. et al. Thyroid and glucocorticoid hormones promote functional T-tubule development in human-induced pluripotent stem cell-derived cardiomyocytes. Circ. Res. 121, 1323–1330 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Redfern, W. S. et al. Relationships between preclinical cardiac electrophysiology, clinical QT interval prolongation and torsade de pointes for a broad range of drugs: evidence for a provisional safety margin in drug development. Cardiovasc. Res. 58, 32–45 (2003).

    Article  CAS  PubMed  Google Scholar 

  80. Navarrete, E. G. et al. Screening drug-induced arrhythmia events using human induced pluripotent stem cell-derived cardiomyocytes and low-impedance microelectrode arrays. Circulation https://doi.org/10.1161/CIRCULATIONAHA.112.000570 (2013).

  81. Aliot, E., Capucci, A., Crijns, H. J., Goette, A. & Tamargo, J. Twenty-five years in the making: flecainide is safe and effective for the management of atrial fibrillation. Europace 13, 161–173 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  82. Melgari, D., Zhang, Y., El Harchi, A., Dempsey, C. E. & Hancox, J. C. Molecular basis of hERG potassium channel blockade by the class Ic antiarrhythmic flecainide. J. Mol. Cell. Cardiol. 86, 42–53 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Bers, D. M. Excitation-Contraction Coupling and Cardiac Contractile Force (Kluwer Academic Publishers, 2001).

  84. Watanabe, H. et al. Flecainide prevents catecholaminergic polymorphic ventricular tachycardia in mice and humans. Nat. Med. 15, 380–383 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Hilliard, F. A. et al. Flecainide inhibits arrhythmogenic Ca2+ waves by open state block of ryanodine receptor Ca2+ release channels and reduction of Ca2+ spark mass. J. Mol. Cell. Cardiol. 48, 293–301 (2010).

    Article  CAS  PubMed  Google Scholar 

  86. Lacerda, A. E. et al. Alfuzosin delays cardiac repolarization by a novel mechanism. J. Pharmacol. Exp. Ther. 324, 427–433 (2007).

    Article  PubMed  CAS  Google Scholar 

  87. Kanzaki, Y. et al. Three-dimensional architecture of cardiomyocytes and connective tissue in human heart revealed by scanning electron microscopy. Circulation 122, 1973–1974 (2010).

    Article  PubMed  Google Scholar 

  88. Lian, X. et al. Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling. Proc. Natl Acad. Sci. USA 109, E1848–E1857 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Tohyama, S. et al. Distinct metabolic flow enables large-scale purification of mouse and human pluripotent stem cell-derived cardiomyocytes. Cell Stem Cell 12, 127–137 (2013).

    Article  CAS  PubMed  Google Scholar 

  90. Lian, X. et al. Efficient differentiation of human pluripotent stem cells to endothelial progenitors via small-molecule activation of WNT signaling. Stem Cell Rep. 3, 804–816 (2014).

    Article  CAS  Google Scholar 

  91. Komeya, M. et al. Pumpless microfluidic system driven by hydrostatic pressure induces and maintains mouse spermatogenesis in vitro. Sci. Rep. 7, 15459 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  92. Linkert, M. et al. Metadata matters: access to image data in the real world. J. Cell Biol. 189, 777–782 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Laughner, J. I., Ng, F. S., Sulkin, M. S., Arthur, R. M. & Efimov, I. R. Processing and analysis of cardiac optical mapping data obtained with potentiometric dyes. Am. J. Physiol. Heart Circ. Physiol. 303, H753–H765 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Fridericia, L. S. The duration of systole in an electrocardiogram in normal humans and in patients with heart disease. Ann. Noninvasive Electrocardiol. 8, 343–351 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Huang, Y. L., Walker, A. S. & Miller, E. W. A photostable silicon rhodamine platform for optical voltage sensing. J. Am. Chem. Soc. 137, 10767–10776 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Hough, P. V. C. Method and means for recognizing complex patterns. US patents, Ser. No. 17,715 6 Claims. (Cl. S40-146.3) (1960).

  97. Brunelli, R. Template Matching Techniques in Computer Vision: Theory and Practice (Wiley, 2009).

  98. Schocken, D. et al. Comparative analysis of media effects on human induced pluripotent stem cell-derived cardiomyocytes in proarrhythmia risk assessment. J. Pharmacol. Toxicol. Methods 90, 39–47 (2018).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was funded in part by the California Institute for Regenerative Medicine DISC2-10090 (K.E.H.), NIH-NHLBI HL130417 (K.E.H.), NIH-NIGMS R35GM1195855 (E.W.M.), NIH-NIGMS T32GM066698 (S.C.B.), the Research Council of Norway INTPART Project 249885, the SUURPh programme funded by the Norwegian Ministry of Education and Research, and the Peder Sather Center for Advanced Study (UC Berkeley). We thank M. West (UC Berkeley) for assistance with image analysis and flow cytometry; S. Weber (Technische Universität Dresden) and S. Renschler (Washington University in St. Louis) for helpful advice on RNA isolation, cDNA amplification and data analysis; Y. Rudy, J. Nerbonne, J. Silva and J. Cui (Washington University in St. Louis) for critical discussions on action potential acquisition, mathematical modelling and data analysis; B. Conklin (Gladstone Institutes, San Francisco, USA) for technical advice on the WTC iPSC line; J. Wu (Stanford University) and the Stanford University Cardiovascular BioBank for providing the SCVI20 and SCVI273 iPSC lines and providing technical advice regarding these lines; and the Barcelona Stem Cell Institute for providing the G15.AO line.

Author information

Authors and Affiliations

Authors

Contributions

N.H., B.C., G.N., A.G.E. and K.E.H. designed experimental studies. N.H., B.C., G.N., B.S., S.C.B., V.C., F.T.L.-M., N.C.J. and N.D. performed experimental studies and iPSC cultures. S.W., K.H.J., Å. Telle and A. Tveito designed and executed computational modelling studies to predict molecular changes in voltage and calcium handling underlying MM-induced physiology changes. B.C. and D.C. created software routines to automate analysis of the force produced by cardiac microtissues. J.S. and M.S. designed and assisted in gene expression analysis studies. K.E.H., A.G.E. and S.W. guided drug-probe-based studies on cardiomyocyte electrophysiology and calcium handling in MPS. E.W.M. and S.C.B. synthesized BeRST-1 dye for action potential analysis. A.S. advised on cell metabolism. K.E.H., N.H., A.G.E. and B.C. wrote the paper. K.E.H. supervised and funded the research.

Corresponding author

Correspondence to Kevin E. Healy.

Ethics declarations

Competing interests

K.E.H., A.E., N.H., S.W., B.S. and V.C. have a financial relationship with Organos Inc., and hence may benefit from the commercialization of the results of this research. The other authors declare no competing interests.

Peer review

Peer review information

Nature Biomedical Engineering thanks the anonymous reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Development and characterization of iPSC-stromal cells.

A) Differentiation tree depicting the lineage of iPSC-cardiomyocytes (hiPS-CM) and the isogenic hiPSC-derived stromal cell population (hiPSC-SC). Specific biomarkers (blue) were verified by qRT-PCR. B) Brewer plot identifying gene expression patterns over the course of differentiation of hiPSC into hiPSC-SC, with hiPSC-CM included on the plot for comparison. C) Immunofluorescence molecular characterization of hiPSC-SC. These hiPSC-SC were markedly positive for all stromal markers shown, while markers of smooth muscle (Calponin) and endothelial cells (CD31) were not detected. iPS-SC also produce key ECM proteins: Laminin, Fibronectin and Collagen IV, while substantial Collagen I was not detected. D) Representative fluorescence micrograph of cardiac MPS, which shows highly organized, aligned cardiomyocytes stained for Sarcomeric α-Actinin (ACTN2, green). Cells were counterstained for nuclei with DAPI (blue) in all fluorescence micrographs. Scale bars: C) 500 µm D) left panel, 20 µm and right panel, 10 µm.

Source data

Extended Data Fig. 2 Design-of-experiments (DoE) based screens identify maturation media for hiPSC-CM microphysiological systems.

A) Approach used for initial screen. Computational motion capture is performed on bright-field videos of contracting cardiac MPS, giving the beating interval (defined as the distance between peaks in motion speed for contraction and relaxation, which approximates the interval over which displacement occurs). The knock-in reporter, GCaMP6f, is used to monitor the timing (rate corrected Full-Width-Half-Maximum, FWHM and amplitude of calcium transients in MPS. B-F) Results from representative L9 Taguchi Array experiments, depicting B) beating interval, C) spontaneous beating frequency, and D) tissue beating prevalence, all obtained from motion tracking analysis, along with calcium transient E) FWHM and F) amplitude, obtained from analysis of GCaMP6f fluorescence. Based on 1-way ANOVA tests, glucose levels significantly impact beating interval (p < 0.05) along with spontaneous beat rate, beating prevalence at 1hz, and GCaMP amplitude at 1Hz (p < 0.01). BSA levels had a significant impact on beating interval (p < 0.05) and GCaMP FWHM at 1Hz (p < 0.01). MPS were cultured for ten days prior to analysis for the L9 experiments. G-H) Comparison of MPS cultured in standard media (red) to MPS cultured in glucose-free media during L9 studies (black), and MPS cultured in the final Maturation Media (blue) when assessed for (H) beating prevalence at 1hz or (I) beating interval. Data: B-F: plot of mean ± SEM, n = 9 MPS per group; G-H: all data points with mean ± SD, n = 12 for SM group, n = 10 for MM group, and n = 3-4 for all other groups, except for beating interval in media 8, which could only be calculated in n = 1 sample (no other samples cultured in this media exhibited either spontaneous or paced beating). p values reflect values obtained from post-hoc Holm Sidak test after confirming global significance across groups with 1-way ANOVA.

Source data

Extended Data Fig. 3 Additional information about the media screen and representative 2D traces of MM-treated monolayers.

A-B) Representative action-potential tracings for 2D monolayers of A) WTC and B) SCVI20 cell lines cultured for one week in Maturation Media (MM). C-D) Quantification of action potential time from 20% above baseline to peak (Upstroke80) in C) WTC MPS (n = 5 per group) and 2D monolayers (n = 21 per group) and D) SCVI20 MPS (n = 60-63 per group). E-F) Quantification of C) beat-rate corrected cAPD80 and (MM: n = 16, SM: n = 10 per group); D) background corrected calcium amplitude (F/F0) in SCVI273 MPS (MM: n = 20, SM: n = 25 per group). G-H) Analysis of changes in G) cAPD80 (MM: n = 13, SM: n = 12 per group) and H) contractile prevalence in MM-pre-treated MPS that resulted from modulating the levels of palmitate and albumin in MM (MM: n = 9, SM: n = 12 per group). Removing both palmitate and albumin from MM (M1) resulted in MPS with APD80 that were significantly higher than APD80 of MM-treated MPS, and which were no different from APD80 of SM-treated MPS. Removal of Palmitate alone (M2), or of albumin alone (M3) led to a new medium that exhibited APD80 significantly less than MM (p < 0.05). Compared to MM, M2 treated MPS exhibited slightly reduced beating prevalence, whereas this metric was enhanced for M3 treated MPS, although these changes were not statistically significant. Slight reduction of the albumin content of MM from 2.5% to 1% (M4) did not have significant effects on APD80 or prevalence of motion in MPS, compared to those treated with MM. All data: plot of all points with mean ± SD. p values from post-Hoc Holm-Bonferonni test after confirming global significance with 1-way ANOVA.

Source data

Extended Data Fig. 4 Action-potential duration and beat rate in MPS of four genotypes.

A) Randomly selected subset of data on rate-corrected APD80 (cAPD80) and B) Spontaneous beat rate for a representative set of MPS for all four genotypes, pre-treated for 10 days with either SM or MM. p values from t-test with Welch’s correction for non-constant standard deviation.

Source data

Extended Data Fig. 5 Extended mitochondrial and metabolic analysis of MPS and 2D iPSC-CM monolayers.

A-D) Representative images of A-B) SCVI20 and C-D) G15.AO MPS after treatment for 10 days with either (A,C) MM or (B,D) SM. MPS are stained for Mitotracker (red) or anti-mitochondrial antibodies (green) with Draq5 nuclear counterstain (blue). E) Representative Oxygen Consumption Rate (OCR) tracings of SCVI20 iPSC-CM monolayers after culture in MM (blue) or SM (red) for 10 days. F,G) Quantification of reserve OCR (change in OCR from baseline with FCCP treatment) and total ATP capacity indicate a shift toward ß-oxidation with MM in 2D monolayers (n = 4 independent wells per group). H-K) Representative images of 2D H,I) WTC and J,K) SCVI20 iPSC-CM monolayers stained with MitoTrackerRed (left; red) and anti-mitochondrial antibodies (right; green). Scale bars: 20μm.

Source data

Extended Data Fig. 6 Expression and localization of sarcomere proteins in MM-treated MPS.

A-B) Fourier domain-based quantification of sarcomeric order in MPS treated with SM versus MM for (A) WTC or (B) SCVI20 cell line treated with MM (blue) or SM (red) for 10 days (n = 9 independent MPS per group). C,D) Quantification of protein expression (antibody staining with analysis by a condition-blinded user) for C) MYH7 (n = 8 independent MPS per group; WTC, open squares and circles; SCVI20, closed squares and circles) and D) MLC-2v (n = 3 independent MPS per group; SCVI20) in MPS treated for ten days with MM (blue) or SM (red). E-G) Representative micrographs of E) WTC and F,G) SCVI20 MPS after staining for E,F) MYH7 and G) MLC-2v. Error bars represent mean ± SD. Antibody staining in green, with blue Draq5 nuclear counterstain. Scale bars: 20μm.

Source data

Extended Data Fig. 7 Representative action-potential changes in MPS exposed to verapamil, flecainide and alfuzosin after MM pre-treatment.

Representative, intensity normalized action-potential traces are depicted for (A,C,E) WTC and (B,D,F) SCVI MPS pre-treated with MM and exposed to escalating doses of (A,B) verapamil, (C,D) flecainide, or (E,F) alfuzosin.

Supplementary information

Supplementary Information

Supplementary methods, figures, tables and references.

Reporting Summary

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Huebsch, N., Charrez, B., Neiman, G. et al. Metabolically driven maturation of human-induced-pluripotent-stem-cell-derived cardiac microtissues on microfluidic chips. Nat. Biomed. Eng 6, 372–388 (2022). https://doi.org/10.1038/s41551-022-00884-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41551-022-00884-4

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing