Skip to main content

Advertisement

Log in

Two-Dimensional Culture Systems to Enable Mechanics-Based Assays for Stem Cell-Derived Cardiomyocytes

  • Published:
Experimental Mechanics Aims and scope Submit manuscript

A Correction to this article was published on 03 January 2020

This article has been updated

Abstract

Well-controlled 2D cell culture systems advance basic investigations in cell biology and provide innovative platforms for drug development, toxicity testing, and diagnostic assays. These cell culture systems have become more advanced in order to provide and to quantify the appropriate biomechanical and biochemical cues that mimic the milieu of conditions present in vivo. Here we present an innovative 2D cell culture system to investigate human stem cell-derived cardiomyocytes, the muscle cells of the heart responsible for pumping blood throughout the body. We designed our 2D cell culture platform to control intracellular features to produce adult-like cardiomyocyte organization with connectivity and anisotropic conduction comparable to the native heart, and combined it with optical microscopy to quantify cell-cell and cell-substrate mechanical interactions. We show the measurement of forces and displacements that occur within individual cells, between neighboring cells, and between cells and their surrounding matrix. This system has broad potential to expand our understanding of tissue physiology, with particular advantages for the study of the mechanically active heart. Furthermore, this technique should prove valuable in screening potential drugs for efficacy and testing for toxicity.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

Change history

  • 03 January 2020

    The authors would like to correct the sentence ���The mass ratio��������� in the subsection on Engineered Substrate System in MATERIALS AND METHODS, to read:

References

  1. Benjamin EJ, Blaha MJ, Chiuve SE, Cushman M, Das SR, Deo R, de Ferranti SD, Floyd J, Fornage M, Gillespie C, Isasi CR, Jiménez MC, Jordan LC, Judd SE, Lackland D, Lichtman JH, Lisabeth L, Liu S, Longenecker CT, Mackey RH, Matsushita K, Mozaffarian D, Mussolino ME, Nasir K, Neumar RW, Palaniappan L, Pandey DK, Thiagarajan RR, Reeves MJ, Ritchey M, Rodriguez CJ, Roth GA, Rosamond WD, Sasson C, Towfighi A, Tsao CW, Turner MB, Virani SS, Voeks JH, Willey JZ, Wilkins JT, Wu JH, Alger HM, Wong SS, Muntner P (2017) Heart disease and stroke statistics—2017 update: a report from the American Heart Association. Circulation. https://doi.org/10.1161/cir.0000000000000485

  2. Akhyari P, Kamiya H, Haverich A, Karck M, Lichtenberg A (2008) Myocardial tissue engineering: the extracellular matrix. Eur J Cardiothorac Surg 34(2):229–241

    Google Scholar 

  3. Xu J, Kochanek K, Murphy S, Tejada-Vera B (2010) Deaths: final data for 2007. Natl Vital Stat Rep 58(19):1–19

    Google Scholar 

  4. Xu J, Kochanek KD, Murphy SL, Tejada-Vera B (2016) Deaths: final data for 2014. Natl Vital Stat Rep 65:1-122.

  5. Michalopoulos GK, DeFrances MC (1997) Liver regeneration. Science 276(5309):60–66

    Google Scholar 

  6. Poss KD, Wilson LG, Keating MT (2002) Heart regeneration in zebrafish. Science 298(5601):2188–2190

    Google Scholar 

  7. Bergmann O, Bhardwaj RD, Bernard S, Zdunek S, Barnabé-Heider F, Walsh S, Zupicich J, Alkass K, Buchholz BA, Druid H (2009) Evidence for cardiomyocyte renewal in humans. Science 324(5923):98–102

    Google Scholar 

  8. Frangogiannis NG, Smith CW, Entman ML (2002) The inflammatory response in myocardial infarction. Cardiovasc Res 53(1):31–47

    Google Scholar 

  9. Holmes JW, Borg TK, Covell JW (2005) Structure and mechanics of healing myocardial infarcts. Annu Rev Biomed Eng 7:223–253

    Google Scholar 

  10. Bigger JT, Fleiss JL, Kleiger R, Miller JP, Rolnitzky LM (1984) The relationships among ventricular arrhythmias, left ventricular dysfunction, and mortality in the 2 years after myocardial infarction. Circulation 69(2):250–258

    Google Scholar 

  11. Wu R, Hu X, Wang J (2018) Concise review: optimized strategies for stem cell-based therapy in myocardial repair: clinical translatability and potential limitation. Stem Cells 36:482–500

    Google Scholar 

  12. Buikema JW, Wu SM (2017) Untangling the biology of genetic cardiomyopathies with pluripotent stem cell disease models. Curr Cardiol Rep 19(4):30

    Google Scholar 

  13. Mordwinkin NM, Burridge PW, Wu JC (2013) A review of human pluripotent stem cell-derived cardiomyocytes for high-throughput drug discovery, cardiotoxicity screening, and publication standards. J Cardiovasc Transl Res 6(1):22–30

    Google Scholar 

  14. Zhang BY, Xiao Y, Hsieh A, Thavandiran N, Radisic M (2011) Micro- and nanotechnology in cardiovascular tissue engineering. Nanotechnology 22(49):494003. https://doi.org/10.1088/0957-4484/22/49/494003

    Article  Google Scholar 

  15. Falconnet D, Csucs G, Grandin HM, Textor M (2006) Surface engineering approaches to micropattern surfaces for cell-based assays. Biomaterials 27(16):3044–3063. https://doi.org/10.1016/j.biomaterials.2005.12.024

    Article  Google Scholar 

  16. Flemming RG, Murphy CJ, Abrams GA, Goodman SL, Nealey PF (1999) Effects of synthetic micro- and nano-structured surfaces on cell behavior. Biomaterials 20(6):573–588. https://doi.org/10.1016/S0142-9612(98)00209-9

    Article  Google Scholar 

  17. Bettinger CJ, Langer R, Borenstein JT (2009) Engineering substrate topography at the micro- and nanoscale to control cell function. Angew Chem Int Ed 48(30):5406–5415. https://doi.org/10.1002/anie.200805179

    Article  Google Scholar 

  18. Lehnert D, Wehrle-Haller B, David C, Weiland U, Ballestrem C, Imhof BA, Bastmeyer M (2004) Cell behaviour on micropatterned substrata: limits of extracellular matrix geometry for spreading and adhesion. J Cell Sci 117(1):41–52. https://doi.org/10.1242/jcs.00836

    Article  Google Scholar 

  19. Parker KK, Brock AL, Brangwynne C, Mannix RJ, Wang N, Ostuni E, Geisse NA, Adams JC, Whitesides GM, Ingber DE (2002) Directional control of lamellipodia extension by constraining cell shape and orienting cell tractional forces. FASEB J 16(10):1195–1204. https://doi.org/10.1096/fj.02-0038com

    Article  Google Scholar 

  20. McBeath R, Pirone DM, Nelson CM, Bhadriraju K, Chen CS (2004) Cell shape, cytoskeletal tension, and RhoA regulate stem cell lineage commitment. Dev Cell 6(4):483–495. https://doi.org/10.1016/S1534-5807(04)00075-9

    Article  Google Scholar 

  21. Thomas CH, Collier JH, Sfeir CS, Healy KE (2002) Engineering gene expression and protein synthesis by modulation of nuclear shape. Proc Natl Acad Sci U S A 99(4):1972–1977. https://doi.org/10.1073/pnas.032668799

    Article  Google Scholar 

  22. Kilian KA, Bugarija B, Lahn BT, Mrksich M (2010) Geometric cues for directing the differentiation of mesenchymal stem cells. Proc Natl Acad Sci U S A 107(11):4872–4877. https://doi.org/10.1073/pnas.0903269107

    Article  Google Scholar 

  23. Kaji H, Takii Y, Nishizawa M, Matsue T (2003) Pharmacological characterization of micropatterned cardiac myocytes. Biomaterials 24(23):4239–4244

    Google Scholar 

  24. Chen CS, Mrksich M, Huang S, Whitesides GM, Ingber DE (1997) Geometric control of cell life and death. Science 276(5317):1425–1428

    Google Scholar 

  25. Legant WR, Pathak A, Yang MT, Deshpande VS, McMeeking RM, Chen CS (2009) Microfabricated tissue gauges to measure and manipulate forces from 3D microtissues. Proc Natl Acad Sci 106(25):10097–10102

    Google Scholar 

  26. Leonard A, Bertero A, Powers JD, Beussman KM, Bhandari S, Regnier M, Murry CE, Sniadecki NJ (2018) Afterload promotes maturation of human induced pluripotent stem cell derived cardiomyocytes in engineered heart tissues. J Mol Cell Cardiol 118:147–158

    Google Scholar 

  27. Schroer AK, Shotwell MS, Sidorov VY, Wikswo JP, Merryman WD (2017) I-wire heart-on-a-Chip II: biomechanical analysis of contractile, three-dimensional cardiomyocyte tissue constructs. Acta Biomater 48:79–87

    Google Scholar 

  28. de Lange WJ, Hegge LF, Grimes AC, Tong CW, Brost TM, Moss RL, Ralphe JC (2011) Neonatal mouse-derived engineered cardiac tissue: a novel model system for studying genetic heart disease. Circ Res 109(1):8–19. https://doi.org/10.1161/CIRCRESAHA.111.242354

    Article  Google Scholar 

  29. Shadrin IY, Allen BW, Qian Y, Jackman CP, Carlson AL, Juhas ME, Bursac N (2017) Cardiopatch platform enables maturation and scale-up of human pluripotent stem cell-derived engineered heart tissues. Nat Commun 8(1):1825

    Google Scholar 

  30. Huisken J, Swoger J, Del Bene F, Wittbrodt J, Stelzer EH (2004) Optical sectioning deep inside live embryos by selective plane illumination microscopy. Science 305(5686):1007–1009

    Google Scholar 

  31. Mertz J (2011) Optical sectioning microscopy with planar or structured illumination. Nat Methods 8(10):811

    Google Scholar 

  32. Chen F, Tillberg PW, Boyden ES (2015) Expansion microscopy. Science. 347(6221):543–548

    Google Scholar 

  33. Macosko EZ, Basu A, Satija R, Nemesh J, Shekhar K, Goldman M, Tirosh I, Bialas AR, Kamitaki N, Martersteck EM (2015) Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 161(5):1202–1214

    Google Scholar 

  34. Klein AM, Mazutis L, Akartuna I, Tallapragada N, Veres A, Li V, Peshkin L, Weitz DA, Kirschner MW (2015) Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells. Cell 161(5):1187–1201

    Google Scholar 

  35. Hughes S (2004) The pathology of hypertrophic cardiomyopathy. Histopathology 44(5):412–427

    Google Scholar 

  36. Hoskins AC, Jacques A, Bardswell SC, McKenna WJ, Tsang V, dos Remedios CG, Ehler E, Adams K, Jalilzadeh S, Avkiran M (2010) Normal passive viscoelasticity but abnormal myofibrillar force generation in human hypertrophic cardiomyopathy. J Mol Cell Cardiol 49(5):737–745

    Google Scholar 

  37. Fidzianska A, Glinka-Lindner Z, Religa G, Walczak E (2010) Usefulness of the ultrastructural and immunohistochemical analysis of cardiac biopsy in affected heart. Folia Neuropathol 48:57–63

    Google Scholar 

  38. Zhao Y-T, Valdivia CR, Gurrola GB, Hernández JJ, Valdivia HH (2015) Arrhythmogenic mechanisms in ryanodine receptor channelopathies. Sci China Life Sci 58(1):54–58

    Google Scholar 

  39. Novak A, Barad L, Zeevi-Levin N, Shick R, Shtrichman R, Lorber A, Itskovitz-Eldor J, Binah O (2012) Cardiomyocytes generated from CPVTD307H patients are arrhythmogenic in response to beta-adrenergic stimulation. J Cell Mol Med 16(3):468–482. https://doi.org/10.1111/j.1582-4934.2011.01476.x

    Article  Google Scholar 

  40. Halapas A, Papalois A, Stauropoulou A, Philippou A, Pissimissis N, Chatzigeorgiou A, Kamper E, Koutsilieris M (2008) In vivo models for heart failure research. In Vivo 22(6):767–780

    Google Scholar 

  41. Bers DM (2002) Cardiac excitation–contraction coupling. Nature 415(6868):198–205

    Google Scholar 

  42. Endoh M (2004) Force–frequency relationship in intact mammalian ventricular myocardium: physiological and pathophysiological relevance. Eur J Pharmacol 500(1):73–86

    Google Scholar 

  43. Dixon JA, Spinale FG (2009) Large animal models of heart failure a critical link in the translation of basic science to clinical practice. Circ Heart Fail 2(3):262–271

    Google Scholar 

  44. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones JM (1998) Embryonic stem cell lines derived from human blastocysts. science 282(5391):1145–1147

    Google Scholar 

  45. Laflamme MA, Chen KY, Naumova AV, Muskheli V, Fugate JA, Dupras SK, Reinecke H, Xu C, Hassanipour M, Police S (2007) Cardiomyocytes derived from human embryonic stem cells in pro-survival factors enhance function of infarcted rat hearts. Nat Biotechnol 25(9):1015–1024

    Google Scholar 

  46. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, Yamanaka S (2007) Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131(5):861–872

    Google Scholar 

  47. Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, Nie J, Jonsdottir GA, Ruotti V, Stewart R (2007) Induced pluripotent stem cell lines derived from human somatic cells. Science 318(5858):1917–1920

    Google Scholar 

  48. Lian X, Hsiao C, Wilson G, Zhu K, Hazeltine LB, Azarin SM, Raval KK, Zhang J, Kamp TJ, Palecek SP (2012) Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling. Proc Natl Acad Sci 109(27):E1848–E1857

    Google Scholar 

  49. Willems E, Spiering S, Davidovics H, Lanier M, Xia Z, Dawson M, Cashman J, Mercola M (2011) Small-molecule inhibitors of the Wnt pathway potently promote cardiomyocytes from human embryonic stem cell–derived mesoderm. Circ Res 109(4):360–364

    Google Scholar 

  50. Yang X, Pabon L, Murry CE (2014) Engineering adolescence: maturation of human pluripotent stem cell-derived cardiomyocytes. Circ Res 114(3):511–523. https://doi.org/10.1161/CIRCRESAHA.114.300558

    Article  Google Scholar 

  51. Cimetta E, Pizzato S, Bollini S, Serena E, De Coppi P, Elvassore N (2009) Production of arrays of cardiac and skeletal muscle myofibers by micropatterning techniques on a soft substrate. Biomed Microdevices 11(2):389–400. https://doi.org/10.1007/s10544-008-9245-9

    Article  Google Scholar 

  52. McDevitt TC, Angello JC, Whitney ML, Reinecke H, Hauschka SD, Murry CE, Stayton PS (2002) In vitro generation of differentiated cardiac myofibers on micropatterned laminin surfaces. J Biomed Mater Res 60(3):472–479

    Google Scholar 

  53. Feinberg AW, Alford PW, Jin H, Ripplinger CM, Werdich AA, Sheehy SP, Grosberg A, Parker KK (2012) Controlling the contractile strength of engineered cardiac muscle by hierarchal tissue architecture. Biomaterials 33(23):5732–5741. https://doi.org/10.1016/j.biomaterials.2012.04.043

    Article  Google Scholar 

  54. Bray MA, Sheehy SP, Parker KK (2008) Sarcomere alignment is regulated by myocyte shape. Cell Motil Cytoskeleton 65(8):641–651. https://doi.org/10.1002/cm.20290

    Article  Google Scholar 

  55. Chopra A, Patel A, Shieh AC, Janmey PA, Kresh JY (2012) Alpha-catenin localization and sarcomere self-organization on N-cadherin adhesive patterns are myocyte contractility driven. PLoS One 7(10):e47592. https://doi.org/10.1371/journal.pone.0047592

    Article  Google Scholar 

  56. Serena E, Cimetta E, Zatti S, Zaglia T, Zagallo M, Keller G, Elvassore N (2012) Micro-arrayed human embryonic stem cells-derived cardiomyocytes for in vitro functional assay. PLoS One 7(11):e48483. https://doi.org/10.1371/journal.pone.0048483

    Article  Google Scholar 

  57. Ribeiro AJ, Ang YS, Fu JD, Rivas RN, Mohamed TM, Higgs GC, Srivastava D, Pruitt BL (2015) Contractility of single cardiomyocytes differentiated from pluripotent stem cells depends on physiological shape and substrate stiffness. Proc Natl Acad Sci U S A 112(41):12705–12710. https://doi.org/10.1073/pnas.1508073112

    Article  Google Scholar 

  58. Engler AJ, Sen S, Sweeney HL, Discher DE (2006) Matrix elasticity directs stem cell lineage specification. Cell 126(4):677–689

    Google Scholar 

  59. Bhana B, Iyer RK, Chen WL, Zhao R, Sider KL, Likhitpanichkul M, Simmons CA, Radisic M (2010) Influence of substrate stiffness on the phenotype of heart cells. Biotechnol Bioeng 105(6):1148–1160. https://doi.org/10.1002/bit.22647

    Article  Google Scholar 

  60. Napiwocki BN, Salick MR, Ashton RS, Crone WC (2017) Controlling hESC-CM cell morphology on patterned substrates over a range of stiffness. In: Mechanics of biological systems and materials, volume 6. Springer, Cham, pp 161–168

    Google Scholar 

  61. Napiwocki BN, Salick MR, Ashton RS, Crone WC (2016) Polydimethylsiloxane lanes enhance sarcomere organization in human ESC-derived cardiomyocytes. In: Tekalur SA, Zavattieri P, Korach CS (eds) Mechanics of biological systems and materials, Vol 6. Conference proceedings of the society for experimental mechanics series. Springer, New York, pp 105–111. https://doi.org/10.1007/978-3-319-21455-9_12

    Chapter  Google Scholar 

  62. Notbohm J, Banerjee S, Utuje KJ, Gweon B, Jang H, Park Y, Shin J, Butler JP, Fredberg JJ, Marchetti MC (2016) Cellular contraction and polarization drive collective cellular motion. Biophys J 110(12):2729–2738

    Google Scholar 

  63. Napiwocki BN, Stempien A, Notbohm J, Ashton RS, Crone W (2018) Two-dimensional culture systems to investigate mechanical interactions of the cell. In: Mechanics of biological systems, materials and other topics in experimental and applied mechanics, volume 4. Springer, pp 37–39

  64. Pelham RJ, Wang YL (1997) Cell locomotion and focal adhesions are regulated by substrate flexibility. Proc Natl Acad Sci U S A 94(25):13661–13665

    Google Scholar 

  65. Yeung T, Georges PC, Flanagan LA, Marg B, Ortiz M, Funaki M, Zahir N, Ming WY, Weaver V, Janmey PA (2005) Effects of substrate stiffness on cell morphology, cytoskeletal structure, and adhesion. Cell Motil Cytoskeleton 60(1):24–34. https://doi.org/10.1002/cm.20041

    Article  Google Scholar 

  66. Tse JR, Engler AJ (2010) Preparation of hydrogel substrates with tunable mechanical properties. Curr Protoc Cell Biol Chapter 10:Unit 10 16. https://doi.org/10.1002/0471143030.cb1016s47

    Article  Google Scholar 

  67. Kong HJ, Wong E, Mooney DJ (2003) Independent control of rigidity and toughness of polymeric hydrogels. Macromolecules 36(12):4582–4588

    Google Scholar 

  68. Rosellini E, Cristallini C, Barbani N, Vozzi G, Giusti P (2009) Preparation and characterization of alginate/gelatin blend films for cardiac tissue engineering. J Biomed Mater Res A 91(2):447–453

    Google Scholar 

  69. Salick MR, Napiwocki BN, Sha J, Knight GT, Chindhy SA, Kamp TJ, Ashton RS, Crone WC (2014) Micropattern width dependent sarcomere development in human ESC-derived cardiomyocytes. Biomaterials 35(15):4454–4464. https://doi.org/10.1016/j.biomaterials.2014.02.001

    Article  Google Scholar 

  70. Dembo M, Wang YL (1999) Stresses at the cell-to-substrate interface during locomotion of fibroblasts. Biophys J 76(4):2307–2316

    Google Scholar 

  71. Butler JP, Tolic-Norrelykke IM, Fabry B, Fredberg JJ (2002) Traction fields, moments, and strain energy that cells exert on their surroundings. Am J Phys Cell Phys 282(3):C595–C605

    Google Scholar 

  72. Tohyama S, Hattori F, Sano M, Hishiki T, Nagahata Y, Matsuura T, Hashimoto H, Suzuki T, Yamashita H, Satoh Y (2013) Distinct metabolic flow enables large-scale purification of mouse and human pluripotent stem cell-derived cardiomyocytes. Cell Stem Cell 12(1):127–137

    Google Scholar 

  73. Palchesko RN, Zhang L, Sun Y, Feinberg AW (2012) Development of polydimethylsiloxane substrates with tunable elastic modulus to study cell mechanobiology in muscle and nerve. PLoS One 7(12):e51499

    Google Scholar 

  74. Schaefer JA, Tranquillo RT (2016) Tissue contraction force microscopy for optimization of engineered cardiac tissue. Tissue Eng Part C Methods 22(1):76–83. https://doi.org/10.1089/ten.TEC.2015.0220

    Article  Google Scholar 

  75. Soiné JR, Hersch N, Dreissen G, Hampe N, Hoffmann B, Merkel R, Schwarz US (2016) Measuring cellular traction forces on non-planar substrates. Interface Focus 6(5):20160024

    Google Scholar 

  76. Yu HY, Xiong SJ, Tay CY, Leong WS, Tan LP (2012) A novel and simple microcontact printing technique for tacky, soft substrates and/or complex surfaces in soft tissue engineering. Acta Biomater 8(3):1267–1272. https://doi.org/10.1016/j.actbio.2011.09.006

    Article  Google Scholar 

  77. Sutton MA, Orteu JJ, Schreier H (2009) Image correlation for shape, motion and deformation measurements: basic concepts, theory and applications. Springer Science & Business Media, New York

    Google Scholar 

  78. Blaber J, Adair B, Antoniou A (2015) Ncorr: open-source 2D digital image correlation Matlab software. Exp Mech 55(6):1105–1122. https://doi.org/10.1007/s11340-015-0009-1

    Article  Google Scholar 

  79. Bar-Kochba E, Toyjanova J, Andrews E, Kim KS, Franck C (2015) A fast iterative digital volume correlation algorithm for large deformations. Exp Mech 55(1):261–274. https://doi.org/10.1007/s11340-014-9874-2

    Article  Google Scholar 

  80. Sabass B, Gardel ML, Waterman CM, Schwarz US (2008) High resolution traction force microscopy based on experimental and computational advances. Biophys J 94(1):207–220. https://doi.org/10.1529/biophysj.107.113670

    Article  Google Scholar 

  81. Schwarz US, Balaban NQ, Riveline D, Bershadsky A, Geiger B, Safran SA (2002) Calculation of forces at focal adhesions from elastic substrate data: the effect of localized force and the need for regularization. Biophys J 83(3):1380–1394

    Google Scholar 

  82. del Alamo JC, Meili R, Alonso-Latorre B, Rodriguez-Rodriguez J, Aliseda A, Firtel RA, Lasheras JC (2007) Spatio-temporal analysis of eukaryotic cell motility by improved force cytometry. Proc Natl Acad Sci U S A 104(33):13343–13348. https://doi.org/10.1073/pnas.0705815104

    Article  Google Scholar 

  83. Trepat X, Wasserman MR, Angelini TE, Millet E, Weitz DA, Butler JP, Fredberg JJ (2009) Physical forces during collective cell migration. Nat Phys 5(6):426–430. https://doi.org/10.1038/nphys1269

    Article  Google Scholar 

  84. Gerdes AM, Kellerman SE, Moore JA, Muffly KE, Clark LC, Reaves PY, Malec KB, McKeown PP, Schocken DD (1992) Structural remodeling of cardiac myocytes in patients with ischemic cardiomyopathy. Circulation 86(2):426–430

    Google Scholar 

  85. Wrighton PJ, Klim JR, Hernandez BA, Koonce CH, Kamp TJ, Kiessling LL (2014) Signals from the surface modulate differentiation of human pluripotent stem cells through glycosaminoglycans and integrins. Proc Natl Acad Sci 111(51):18126–18131

    Google Scholar 

  86. Pillekamp F, Haustein M, Khalil M, Emmelheinz M, Nazzal R, Adelmann R, Nguemo F, Rubenchyk O, Pfannkuche K, Matzkies M (2012) Contractile properties of early human embryonic stem cell-derived cardiomyocytes: beta-adrenergic stimulation induces positive chronotropy and lusitropy but not inotropy. Stem Cells Dev 21(12):2111–2121

    Google Scholar 

  87. Maruthamuthu V, Sabass B, Schwarz US, Gardel ML (2011) Cell-ECM traction force modulates endogenous tension at cell-cell contacts. Proc Natl Acad Sci U S A 108(12):4708–4713. https://doi.org/10.1073/pnas.1011123108

    Article  Google Scholar 

  88. Tambe DT, Hardin CC, Angelini TE, Rajendran K, Park CY, Serra-Picamal X, Zhou EHH, Zaman MH, Butler JP, Weitz DA, Fredberg JJ, Trepat X (2011) Collective cell guidance by cooperative intercellular forces. Nat Mater 10(6):469–475. https://doi.org/10.1038/nmat3025

    Article  Google Scholar 

  89. Tambe DT, Croutelle U, Trepat X, Park CY, Kim JH, Millet E, Butler JP, Fredberg JJ (2013) Monolayer stress microscopy: limitations, artifacts, and accuracy of recovered intercellular stresses. PLoS One 8(2):e55172. https://doi.org/10.1371/journal.pone.0055172

    Article  Google Scholar 

Download references

Acknowledgements

Research reported in this publication was supported in part by the National Science Foundation under grant number 1660703 (JN) and by the National Heart, Lung, and Blood Institute (NHLBI) of the National Institutes of Health under award number R01 HL107367 (JCR). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health or the National Science Foundation. Support was also provided by the Karen Thompson Medhi Professorship (WCC), the Graduate School (WCC) and the Office of the Vice Chancellor for Research and Graduate Education (WCC, JN) at the University of Wisconsin-Madison. Additional thanks are given to Dr. Timothy Kamp of the University of Wisconsin-Madison for providing the cTnT H9 hESC line used in some of the experiments described above.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to J. Notbohm or W.C. Crone.

Additional information

Publisher’s Note

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

Electronic supplementary material

ESM 1

(AVI 4154 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Notbohm, J., Napiwocki, B., de Lange, W. et al. Two-Dimensional Culture Systems to Enable Mechanics-Based Assays for Stem Cell-Derived Cardiomyocytes. Exp Mech 59, 1235–1248 (2019). https://doi.org/10.1007/s11340-019-00473-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11340-019-00473-8

Keywords

Navigation