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.

Bioengineering approaches to treat the failing heart: from cell biology to 3D printing

Abstract

Successfully engineering a functional, human, myocardial pump would represent a therapeutic alternative for the millions of patients with end-stage heart disease and provide an alternative to animal-based preclinical models. Although the field of cardiac tissue engineering has made tremendous advances, major challenges remain, which, if properly resolved, might allow the clinical implementation of engineered, functional, complex 3D structures in the future. In this Review, we provide an overview of state-of-the-art studies, challenges that have not yet been overcome and perspectives on cardiac tissue engineering. We begin with the most clinically relevant cell sources used in this field and discuss the use of topological, biophysical and metabolic stimuli to obtain mature phenotypes of cardiomyocytes, particularly in relation to organized cytoskeletal and contractile intracellular structures. We then move from the cellular level to engineering planar cardiac patches and discuss the need for proper vascularization and the main strategies for obtaining it. Finally, we provide an overview of several different approaches for the engineering of volumetric organs and organ parts — from whole-heart decellularization and recellularization to advanced 3D printing technologies.

Key points

  • The successful engineering of functional, human, myocardial tissues would represent a therapeutic alternative for the millions of patients with end-stage heart disease.

  • The well-organized, characteristic structure of the heart across its multiple scales is crucial for its mechanical function; therefore, recapitulating these structure–function relationships in engineered myocardial tissues is necessary.

  • Developing integrative maturation protocols and quantitative tools to assess the maturation of human induced pluripotent stem cell-derived cardiomyocytes is needed to produce cells with a mature phenotype that can successfully integrate with the host tissue.

  • Although the engineering of anisotropic, thick, vascularized tissue patches has been demonstrated using various fabrication techniques, their clinical implementation is still hampered by their poor integration with host tissue.

  • Fabricating 3D, volumetric, myocardial tissues could address the urgent need of patients with end-stage heart failure for heart donation and is therefore an active and evolving field of research.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Cardiac tissue engineering timeline.
Fig. 2: Cardiac tissue engineering concepts.
Fig. 3: Maturation of stem cell-derived cardiomyocytes.
Fig. 4: Recapitulating the native heart matrix.
Fig. 5: Approaches for tissue vascularization.
Fig. 6: Whole heart bioengineering: how to build a heart.
Fig. 7: Bioengineering ventricular-like structures and whole hearts.

Similar content being viewed by others

References

  1. Walker, C. A. & Spinale, F. G. The structure and function of the cardiac myocyte: a review of fundamental concepts. J. Thorac. Cardiovasc. Surg. 118, 375–382 (1999).

    CAS  PubMed  Google Scholar 

  2. Chien, K. R., Domian, I. J. & Parker, K. K. Cardiogenesis and the complex biology of regenerative cardiovascular medicine. Science 322, 1494–1497 (2008).

    CAS  PubMed  Google Scholar 

  3. Hirose, K. et al. Evidence for hormonal control of heart regenerative capacity during endothermy acquisition. Science 364, 184–188 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Curfman, G. Stem cell therapy for heart failure: an unfulfilled promise? JAMA 321, 1186–1187 (2019).

    PubMed  Google Scholar 

  5. Miller, L., Birks, E., Guglin, M., Lamba, H. & Frazier, O. Use of ventricular assist devices and heart transplantation for advanced heart failure. Circ. Res. 124, 1658–1678 (2019).

    CAS  PubMed  Google Scholar 

  6. Benjamin, M. A. et al. Heart disease and stroke statistics — 2017 update: a report from the American Heart Association. Circulation 135, e146–e603 (2017).

    PubMed  PubMed Central  Google Scholar 

  7. Lawson, E. J. et al. 20-year trends in cause-specific heart failure outcomes by sex, socioeconomic status, and place of diagnosis: a population-based study. Lancet Public Health 4, e406–e420 (2019).

    PubMed  PubMed Central  Google Scholar 

  8. Mamas, M. A. et al. Do patients have worse outcomes in heart failure than in cancer? A primary care-based cohort study with 10-year follow-up in Scotland. Eur. J. Heart Fail. 19, 1095–1104 (2017).

    PubMed  Google Scholar 

  9. Bianco, P. et al. The meaning, the sense and the significance: translating the science of mesenchymal stem cells into medicine. Nat. Med. 19, 35–42 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Madonna, R. et al. Position paper of the European Society of Cardiology Working Group Cellular Biology of the Heart: cell-based therapies for myocardial repair and regeneration in ischemic heart disease and heart failure. Eur. Heart J. 37, 1789–1798 (2016).

    PubMed  PubMed Central  Google Scholar 

  11. Litviňuková, M. et al. Cells of the adult human heart. Nature 588, 466–472 (2020).

    PubMed  PubMed Central  Google Scholar 

  12. Karantalis, V. et al. Autologous mesenchymal stem cells produce concordant improvements in regional function, tissue perfusion, and fibrotic burden when administered to patients undergoing coronary artery bypass grafting: the Prospective Randomized Study of Mesenchymal Stem Cell Therapy in Patients Undergoing Cardiac Surgery (PROMETHEUS) trial. Circ. Res. 114, 1302–1310 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Heldman, A. W. et al. Transendocardial mesenchymal stem cells and mononuclear bone marrow cells for ischemic cardiomyopathy: the TAC-HFT randomized trial. JAMA 311, 62–73 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Makkar, R. R. et al. Intracoronary cardiosphere-derived cells for heart regeneration after myocardial infarction (CADUCEUS): a prospective, randomised phase 1 trial. Lancet 379, 895–904 (2012).

    PubMed  PubMed Central  Google Scholar 

  15. Gnecchi, M. et al. Paracrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cells. Nat. Med. 11, 367–368 (2005).

    CAS  PubMed  Google Scholar 

  16. Noiseux, N. et al. Mesenchymal stem cells overexpressing Akt dramatically repair infarcted myocardium and improve cardiac function despite infrequent cellular fusion or differentiation. Mol. Ther. 14, 840–850 (2006).

    CAS  PubMed  Google Scholar 

  17. Zeng, L. et al. Bioenergetic and functional consequences of bone marrow-derived multipotent progenitor cell transplantation in hearts with postinfarction left ventricular remodeling. Circulation 115, 1866–1875 (2007).

    PubMed  Google Scholar 

  18. Iso, Y. et al. Multipotent human stromal cells improve cardiac function after myocardial infarction in mice without long-term engraftment. Biochem. Biophys. Res. Commun. 354, 700–706 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Kehat, I. et al. Human embryonic stem cells can differentiate into myocytes with structural and functional properties of cardiomyocytes. J. Clin. Invest. 108, 407–414 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Shiba, Y. et al. Allogeneic transplantation of iPS cell-derived cardiomyocytes regenerates primate hearts. Nature 538, 388–391 (2016).

    CAS  PubMed  Google Scholar 

  21. Shiba, Y. et al. Human ES-cell-derived cardiomyocytes electrically couple and suppress arrhythmias in injured hearts. Nature 489, 322–325 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Chong, J. J. et al. Human embryonic-stem-cell-derived cardiomyocytes regenerate non-human primate hearts. Nature 510, 273–277 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Guo, Y. & Pu, W. T. Cardiomyocyte maturation: new phase in development. Circ. Res. 126, 1086–1106 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Nussbaum, J. et al. Transplantation of undifferentiated murine embryonic stem cells in the heart: teratoma formation and immune response. FASEB J. 21, 1345–1357 (2007).

    CAS  PubMed  Google Scholar 

  25. Takahashi, K. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861–872 (2007).

    CAS  PubMed  Google Scholar 

  26. Zhou, H. et al. Generation of induced pluripotent stem cells using recombinant proteins. Cell Stem Cell 4, 381–384 (2009).

    CAS  PubMed  Google Scholar 

  27. Li, X., Zhang, P., Wei, C. & Zhang, Y. Generation of pluripotent stem cells via protein transduction. Int. J. Dev. Biol. 58, 21–27 (2014).

    CAS  PubMed  Google Scholar 

  28. Zhou, Y. & Zeng, F. Integration-free methods for generating induced pluripotent stem cells. Genomics Proteom. Bioinforma. 11, 284–287 (2013).

    CAS  Google Scholar 

  29. Singh, V. K., Kumar, N., Kalsan, M., Saini, A. & Chandra, R. Mechanism of induction: induced pluripotent stem cells (iPSCs). J. Stem Cells 10, 43–62 (2015).

    CAS  PubMed  Google Scholar 

  30. Schlaeger, T. M. et al. A comparison of non-integrating reprogramming methods. Nat. Biotechnol. 33, 58–63 (2015).

    CAS  PubMed  Google Scholar 

  31. Higuchi, A. et al. Generation of pluripotent stem cells without the use of genetic material. Lab. Invest. 95, 26–42 (2015).

    CAS  PubMed  Google Scholar 

  32. Zhang, J. et al. Functional cardiomyocytes derived from human induced pluripotent stem cells. Circ. Res. 104, e30–e41 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Zhao, W. et al. Strategies for genetically engineering hypoimmunogenic Universal Pluripotent stem cells. iScience 23, 101162 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Palpant, N. J. et al. Generating high-purity cardiac and endothelial derivatives from patterned mesoderm using human pluripotent stem cells. Nat. Protoc. 12, 15–31 (2017).

    CAS  PubMed  Google Scholar 

  36. Lian, X. et al. Directed cardiomyocyte differentiation from human pluripotent stem cells by modulating Wnt/β-catenin signaling under fully defined conditions. Nat. Protoc. 8, 162–175 (2013).

    CAS  PubMed  Google Scholar 

  37. Chen, V. C. et al. Development of a scalable suspension culture for cardiac differentiation from human pluripotent stem cells. Stem Cell Res. 15, 365–375 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Jha, B. S., Farnoodian, M. & Bharti, K. Regulatory considerations for developing a phase I investigational new drug application for autologous induced pluripotent stem cells-based therapy product. Stem Cell Transl. Med. 10, 198–208 (2020).

    Google Scholar 

  39. Chen, Z. et al. Subtype-specific promoter-driven action potential imaging for precise disease modelling and drug testing in hiPSC-derived cardiomyocytes. Eur. Heart J. 38, 292–301 (2017).

    CAS  PubMed  Google Scholar 

  40. He, J.-Q., Ma, Y., Lee, Y., Thomson, J. A. & Kamp, T. J. Human embryonic stem cells develop into multiple types of cardiac myocytes: action potential characterization. Circ. Res. 93, 32–39 (2003).

    CAS  PubMed  Google Scholar 

  41. Cyganek, L. et al. Deep phenotyping of human induced pluripotent stem cell–derived atrial and ventricular cardiomyocytes. JCI Insight 3, e99941 (2018).

    PubMed Central  Google Scholar 

  42. Protze, S. I. et al. Sinoatrial node cardiomyocytes derived from human pluripotent cells function as a biological pacemaker. Nat. Biotechnol. 35, 56–68 (2017).

    CAS  PubMed  Google Scholar 

  43. Laksman, Z. et al. Modeling atrial fibrillation using human embryonic stem cell-derived atrial tissue. Sci. Rep. 7, 5268 (2017).

    PubMed  PubMed Central  Google Scholar 

  44. Bizy, A. et al. Myosin light chain 2-based selection of human iPSC-derived early ventricular cardiac myocytes. Stem Cell Res. 11, 1335–1347 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Goldfracht, I. et al. Generating ring-shaped engineered heart tissues from ventricular and atrial human pluripotent stem cell-derived cardiomyocytes. Nat. Commun. 11, 75 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Zhao, Y. et al. A platform for generation of chamber-specific cardiac tissues and disease modeling. Cell 176, 913–927 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Devalla, H. D. et al. Atrial-like cardiomyocytes from human pluripotent stem cells are a robust preclinical model for assessing atrial-selective pharmacology. EMBO Mol. Med. 7, 394–410 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Yang, X., Pabon, L. & Murry, C. E. Engineering adolescence: maturation of human pluripotent stem cell–derived cardiomyocytes. Circ. Res. 114, 511–523 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Heineke, J. & Molkentin, J. D. Regulation of cardiac hypertrophy by intracellular signalling pathways. Nat. Rev. Mol. Cell Biol. 7, 589–600 (2006).

    CAS  PubMed  Google Scholar 

  50. Mummery, C. L. et al. Differentiation of human embryonic stem cells and induced pluripotent stem cells to cardiomyocytes: a methods overview. Circ. Res. 111, 344–358 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Zwi, L. et al. Cardiomyocyte differentiation of human induced pluripotent stem cells. Circulation 120, 1513–1523 (2009).

    CAS  PubMed  Google Scholar 

  52. Sartiani, L. et al. Developmental changes in cardiomyocytes differentiated from human embryonic stem cells: a molecular and electrophysiological approach. Stem Cell 25, 1136–1144 (2007).

    CAS  Google Scholar 

  53. Dias, T. P. et al. Biophysical study of human induced pluripotent stem cell-derived cardiomyocyte structural maturation during long-term culture. Biochem. Biophys. Res. Commun. 499, 611–617 (2018).

    CAS  PubMed  Google Scholar 

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

    CAS  Google Scholar 

  55. Besser, R. R. et al. Engineered microenvironments for maturation of stem cell derived cardiac myocytes. Theranostics 8, 124–140 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Hazeltine, L. B. et al. Effects of substrate mechanics on contractility of cardiomyocytes generated from human pluripotent stem cells. Int. J. Cell Biol. 2012, 508294 (2012).

    PubMed  PubMed Central  Google Scholar 

  57. Xi, Y. et al. Substrate stiffness alters human induced pluripotent stem cell-derived cardiomyocyte differentiation and maturation. Circulation 140, A17119 (2019).

    Google Scholar 

  58. McCain, M. L., Yuan, H., Pasqualini, F. S., Campbell, P. H. & Parker, K. K. Matrix elasticity regulates the optimal cardiac myocyte shape for contractility. Am. J. Physiol. Heart Circ. Physiol. 306, H1525–H1539 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Bhana, B. et al. Influence of substrate stiffness on the phenotype of heart cells. Biotechnol. Bioeng. 105, 1148–1160 (2010).

    CAS  PubMed  Google Scholar 

  60. Radisic, M. et al. Functional assembly of engineered myocardium by electrical stimulation of cardiac myocytes cultured on scaffolds. Proc. Natl Acad. Sci. USA 101, 18129–18134 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Eng, G. et al. Autonomous beating rate adaptation in human stem cell-derived cardiomyocytes. Nat. Commun. 7, 10312 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Jackman, C. P., Carlson, A. L. & Bursac, N. Dynamic culture yields engineered myocardium with near-adult functional output. Biomaterials 111, 66–79 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Zuppinger, C. 3D culture for cardiac cells. Biochim. Biophys. Acta 1863, 1873–1881 (2016).

    CAS  PubMed  Google Scholar 

  67. Bargehr, J. et al. Epicardial cells derived from human embryonic stem cells augment cardiomyocyte-driven heart regeneration. Nat. Biotechnol. 37, 895–906 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Ravenscroft, S. M., Pointon, A., Williams, A. W., Cross, M. J. & Sidaway, J. E. Cardiac non-myocyte cells show enhanced pharmacological function suggestive of contractile maturity in stem cell derived cardiomyocyte microtissues. Toxicol. Sci. 152, 99–112 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Giacomelli, E. et al. Three-dimensional cardiac microtissues composed of cardiomyocytes and endothelial cells co-differentiated from human pluripotent stem cells. Development 144, 1008–1017 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. 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).

    PubMed  PubMed Central  Google Scholar 

  71. Nakano, H. et al. Glucose inhibits cardiac muscle maturation through nucleotide biosynthesis. Elife 6, e29330 (2017).

    PubMed  PubMed Central  Google Scholar 

  72. 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, E8372–E8381 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Yang, X. et al. Fatty acids enhance the maturation of cardiomyocytes derived from human pluripotent stem cells. Stem Cell Rep. 13, 657–668 (2019).

    CAS  Google Scholar 

  74. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Földes, G. et al. Modulation of human embryonic stem cell-derived cardiomyocyte growth: a testbed for studying human cardiac hypertrophy? J. Mol. Cell. Cardiol. 50, 367–376 (2011).

    PubMed  PubMed Central  Google Scholar 

  77. Wu, L. et al. Angiotensin II promotes cardiac differentiation of embryonic stem cells via angiotensin type 1 receptor. Differentiation 86, 23–29 (2013).

    CAS  PubMed  Google Scholar 

  78. Rupert, C. E. & Coulombe, K. L. IGF1 and NRG1 enhance proliferation, metabolic maturity, and the force-frequency response in hESC-derived engineered cardiac tissues. Stem Cell Int. 2017, 7648409 (2017).

    Google Scholar 

  79. Sheehy, S. P. et al. Quality metrics for stem cell-derived cardiac myocytes. Stem Cell Rep. 2, 282–294 (2014).

    CAS  Google Scholar 

  80. Karbassi, E. et al. Cardiomyocyte maturation: advances in knowledge and implications for regenerative medicine. Nat. Rev. Cardiol. 17, 341–359 (2020).

    PubMed  PubMed Central  Google Scholar 

  81. Reinecke, H., Zhang, M., Bartosek, T. & Murry, C. E. Survival, integration, and differentiation of cardiomyocyte grafts: a study in normal and injured rat hearts. Circulation 100, 193–202 (1999).

    CAS  PubMed  Google Scholar 

  82. Kuo, P.-L. et al. Myocyte shape regulates lateral registry of sarcomeres and contractility. Am. J. Pathol. 181, 2030–2037 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Clark, K. A., McElhinny, A. S., Beckerle, M. C. & Gregorio, C. C. Striated muscle cytoarchitecture: an intricate web of form and function. Annu. Rev. Cell Dev. Biol. 18, 637–706 (2002).

    CAS  PubMed  Google Scholar 

  84. LeDuc, P. P. & Bellin, R. R. Nanoscale intracellular organization and functional architecture mediating cellular behavior. Ann. Biomed. Eng. 34, 102–113 (2006).

    PubMed  Google Scholar 

  85. Geisse, N. A., Sheehy, S. P. & Parker, K. K. Control of myocyte remodeling in vitro with engineered substrates. In Vitro Cell. Dev. Biol. Anim. 45, 343–350 (2009).

    PubMed  Google Scholar 

  86. Ribeiro, A. J. et al. Contractility of single cardiomyocytes differentiated from pluripotent stem cells depends on physiological shape and substrate stiffness. Proc. Natl Acad. Sci. USA 112, 12705–12710 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. McCain, M. L., Sheehy, S. P., Grosberg, A., Goss, J. A. & Parker, K. K. Recapitulating maladaptive, multiscale remodeling of failing myocardium on a chip. Proc. Natl Acad. Sci. USA 110, 9770–9775 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Schroer, A., Pardon, G., Castillo, E., Blair, C. & Pruitt, B. Engineering hiPSC cardiomyocyte in vitro model systems for functional and structural assessment. Prog. Biophys. Mol. Biol. 144, 3–15 (2019).

    CAS  PubMed  Google Scholar 

  89. Yuan, H., Marzban, B. & Kit Parker, K. Myofibrils in cardiomyocytes tend to assemble along the maximal principle stress directions. J. Biomech. Eng. 139, 1210101–1210108 (2017).

    PubMed Central  Google Scholar 

  90. Zhao, G., Bao, X., Huang, G., Xu, F. & Zhang, X. Differential effects of directional cyclic stretching on the functionalities of engineered cardiac tissues. ACS Appl. Bio. Mater. 2, 3508–3519 (2019).

    CAS  Google Scholar 

  91. Parker, K. K., Tan, J., Chen, C. S. & Tung, L. Myofibrillar architecture in engineered cardiac myocytes. Circ. Res. 103, 340–342 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Matsuda, T. et al. N-cadherin-mediated cell adhesion determines the plasticity for cell alignment in response to mechanical stretch in cultured cardiomyocytes. Biochem. Biophys. Res. Commun. 326, 228–232 (2005).

    CAS  PubMed  Google Scholar 

  93. Kada, K. et al. Orientation change of cardiocytes induced by cyclic stretch stimulation: time dependency and involvement of protein kinases. J. Mol. Cell. Cardiol. 31, 247–259 (1999).

    CAS  PubMed  Google Scholar 

  94. Badie, N. & Bursac, N. Novel micropatterned cardiac cell cultures with realistic ventricular microstructure. Biophys. J. 96, 3873–3885 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Fleischer, S. et al. Albumin fiber scaffolds for engineering functional cardiac tissues. Biotechnol. Bioeng. 111, 1246–1257 (2014).

    CAS  PubMed  Google Scholar 

  96. Orlova, Y., Magome, N., Liu, L., Chen, Y. & Agladze, K. Electrospun nanofibers as a tool for architecture control in engineered cardiac tissue. Biomaterials 32, 5615–5624 (2011).

    CAS  PubMed  Google Scholar 

  97. Nawroth, J. C. et al. Automated fabrication of photopatterned gelatin hydrogels for organ-on-chips applications. Biofabrication 10, 025004 (2018).

    PubMed  PubMed Central  Google Scholar 

  98. Bursac, N., Loo, Y., Leong, K. & Tung, L. Novel anisotropic engineered cardiac tissues: studies of electrical propagation. Biochem. Biophys. Res. Commun. 361, 847–853 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Bursac, N., Parker, K., Iravanian, S. & Tung, L. Cardiomyocyte cultures with controlled macroscopic anisotropy: a model for functional electrophysiological studies of cardiac muscle. Circ. Res. 91, e45–e54 (2002).

    CAS  PubMed  Google Scholar 

  100. Benam, K. H. et al. Engineered in vitro disease models. Annu. Rev. Pathol. 10, 195–262 (2015).

    CAS  PubMed  Google Scholar 

  101. Saleem, U. et al. Blinded, multicenter evaluation of drug-induced changes in contractility using human-induced pluripotent stem cell-derived cardiomyocytes. Toxicol. Sci. 176, 103–123 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Yadid, M. et al. Endothelial extracellular vesicles contain protective proteins and rescue ischemia-reperfusion injury in a human heart-on-chip. Sci. Transl. Med. 12, eaax8005 (2020).

    CAS  PubMed  Google Scholar 

  103. Stein, J. M., Mummery, C. L. & Bellin, M. Engineered models of the human heart: directions and challenges. Stem Cell Rep. 3, S2213–S6711 (2020).

    Google Scholar 

  104. Ogle, B. M. et al. Distilling complexity to advance cardiac tissue engineering. Sci. Trans. Med. 8, 342ps313 (2016).

    Google Scholar 

  105. Naveed, M. et al. Cardio-supportive devices (VRD & DCC device) and patches for advanced heart failure: a review, summary of state of the art and future directions. Biomed. Pharmacother. 102, 41–54 (2018).

    PubMed  Google Scholar 

  106. Chachques, J. C. et al. Myocardial assistance by grafting a new bioartificial upgraded myocardium (MAGNUM trial): clinical feasibility study. Ann. Thorac. Surg. 85, 901–908 (2008).

    PubMed  Google Scholar 

  107. Menasché, P. et al. Human embryonic stem cell-derived cardiac progenitors for heart failure. one-year results of the ESCORT Trial. Circulation 136, A14798 (2017).

    Google Scholar 

  108. Menasche, P. et al. Human embryonic stem cell-derived cardiac progenitors for severe heart failure treatment: first clinical case report. Eur. Heart J. 36, 2011–2017 (2015).

    PubMed  Google Scholar 

  109. Riegler, J. et al. Human engineered heart muscles engraft and survive long term in a rodent myocardial infarction model. Circ. Res. 117, 720–730 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Weinberger, F. et al. Cardiac repair in guinea pigs with human engineered heart tissue from induced pluripotent stem cells. Sci. Transl. Med. 8, 363ra148 (2016).

    PubMed  Google Scholar 

  111. Capulli, A., MacQueen, L., Sheehy, S. P. & Parker, K. Fibrous scaffolds for building hearts and heart parts. Adv. Drug Deliv. Rev. 96, 83–102 (2016).

    CAS  PubMed  Google Scholar 

  112. Fleischer, S., Miller, J., Hurowitz, H., Shapira, A. & Dvir, T. Effect of fiber diameter on the assembly of functional 3D cardiac patches. Nanotechnology 26, 291002 (2015).

    PubMed  Google Scholar 

  113. Fleischer, S., Shapira, A., Feiner, R. & Dvir, T. Modular assembly of thick multifunctional cardiac patches. Proc. Natl. Acad. Sci. USA 114, 1898–1903 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Adadi, N. et al. Electrospun fibrous PVDF-TrFe scaffolds for cardiac tissue engineering, differentiation, and maturation. Adv. Mater. Technol. 5, 1900820 (2020).

    CAS  Google Scholar 

  115. Tan, G. Z. & Zhou, Y. Electrospinning of biomimetic fibrous scaffolds for tissue engineering: a review. Int. J. Polymeric Mater. Polymeric Biomater. 69, 947–960 (2020).

    CAS  Google Scholar 

  116. Armstrong, J. P. et al. Engineering anisotropic muscle tissue using acoustic cell patterning. Adv. Mater. 30, e1802649 (2018).

    PubMed  Google Scholar 

  117. Naseer, S. M. et al. Surface acoustic waves induced micropatterning of cells in gelatin methacryloyl (GelMA) hydrogels. Biofabrication 9, 015020 (2017).

    PubMed  PubMed Central  Google Scholar 

  118. Serpooshan, V. et al. Bioacoustic-enabled patterning of human iPSC-derived cardiomyocytes into 3D cardiac tissue. Biomaterials 131, 47–57 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Zwi-Dantsis, L. et al. Remote magnetic nanoparticle manipulation enables the dynamic patterning of cardiac tissues. Adv. Mater. 32, e1904598 (2020).

    PubMed  Google Scholar 

  120. Gao, L. et al. Myocardial tissue engineering with cells derived from human-induced pluripotent stem cells and a native-like, high-resolution, 3-dimensionally printed scaffold. Circ. Res. 120, 1318–1325 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Kuetemeyer, K. et al. Two-photon induced collagen cross-linking in bioartificial cardiac tissue. Opt. Express 19, 15996–16007 (2011).

    CAS  PubMed  Google Scholar 

  122. Ma, Z. et al. Three-dimensional filamentous human diseased cardiac tissue model. Biomaterials 35, 1367–1377 (2014).

    CAS  PubMed  Google Scholar 

  123. Castilho, M. et al. Melt electrospinning writing of poly-Hydroxymethylglycolide-co-ε-Caprolactone-based scaffolds for cardiac tissue engineering. Adv. Healthc. Mater. https://doi.org/10.1002/adhm.201700311 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  124. Mao, M., He, J., Li, Z., Han, K. & Li, D. Multi-directional cellular alignment in 3D guided by electrohydrodynamically-printed microlattices. Acta Biomater. 101, 141–151 (2020).

    CAS  PubMed  Google Scholar 

  125. Liau, B., Christoforou, N., Leong, K. W. & Bursac, N. Pluripotent stem cell-derived cardiac tissue patch with advanced structure and function. Biomaterials 32, 9180–9187 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Huang, N. F. et al. Big bottlenecks in cardiovascular tissue engineering. Commun. Biol. 1, 199 (2018).

    PubMed  PubMed Central  Google Scholar 

  127. Dvir, T. et al. Nanowired three-dimensional cardiac patches. Nat. Nanotechnol. 6, 720–725 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Shevach, M., Maoz, B. M., Feiner, R., Shapira, A. & Dvir, T. Nanoengineering gold particle composite fibers for cardiac tissue engineering. J. Mater. Chem. B. 1, 5210–5217 (2013).

    CAS  PubMed  Google Scholar 

  129. Martins, A. M. et al. Electrically conductive chitosan/carbon scaffolds for cardiac tissue engineering. Biomacromolecules 15, 635–643 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Liang, S. et al. Paintable and rapidly bondable conductive hydrogels as therapeutic cardiac patches. Adv. Mater. 30, e1704235 (2018).

    PubMed  Google Scholar 

  131. Navaei, A. et al. Gold nanorod-incorporated gelatin-based conductive hydrogels for engineering cardiac tissue constructs. Acta Biomater. 41, 133–146 (2016).

    CAS  PubMed  Google Scholar 

  132. Shin, S. R. et al. Reduced graphene oxide-gelMA hybrid hydrogels as scaffolds for cardiac tissue engineering. Small 12, 3677–3689 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Roshanbinfar, K. et al. Carbon nanotube doped pericardial matrix derived electroconductive biohybrid hydrogel for cardiac tissue engineering. Biomater. Sci. 7, 3906–3917 (2019).

    CAS  PubMed  Google Scholar 

  134. Yadid, M., Feiner, R. & Dvir, T. Gold nanoparticle-integrated scaffolds for tissue engineering and regenerative medicine. Nano Lett. 19, 2198–2206 (2019).

    CAS  PubMed  Google Scholar 

  135. Zhao, R., Boudou, T., Wang, W. G., Chen, C. S. & Reich, D. H. Decoupling cell and matrix mechanics in engineered microtissues using magnetically actuated microcantilevers. Adv. Mater. 25, 1699–1705 (2013).

    CAS  PubMed  Google Scholar 

  136. Malki, M., Fleischer, S., Shapira, A. & Dvir, T. Gold nanorod-based engineered cardiac patch for suture-free engraftment by near IR. Nano Lett. 18, 4069–4073 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Lin, X. et al. A viscoelastic adhesive epicardial patch for treating myocardial infarction. Nat. Biomed. Eng. 3, 632–643 (2019).

    CAS  PubMed  Google Scholar 

  138. Walker, B. W. et al. Engineering a naturally-derived adhesive and conductive cardiopatch. Biomaterials 207, 89–101 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Feiner, R. et al. Engineered hybrid cardiac patches with multifunctional electronics for online monitoring and regulation of tissue function. Nat. Mater. 15, 679–685 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Feiner, R. et al. A stretchable and flexible cardiac tissue–electronics hybrid enabling multiple drug release, sensing, and stimulation. Small 15, 1805526 (2019).

    Google Scholar 

  141. Xu, L. et al. 3D multifunctional integumentary membranes for spatiotemporal cardiac measurements and stimulation across the entire epicardium. Nat. Commun. 5, 3329 (2014).

    PubMed  Google Scholar 

  142. Zheng, G., Patolsky, F., Cui, Y., Wang, W. U. & Lieber, C. M. Multiplexed electrical detection of cancer markers with nanowire sensor arrays. Nat. Biotechnol. 23, 1294–1301 (2005).

    CAS  PubMed  Google Scholar 

  143. Andorko, J. I. & Jewell, C. M. Designing biomaterials with immunomodulatory properties for tissue engineering and regenerative medicine. Bioeng. Transl. Med. 2, 139–155 (2017).

    PubMed  PubMed Central  Google Scholar 

  144. Edri, R. et al. Personalized hydrogels for engineering diverse fully autologous tissue implants. Adv. Mater. 31, e1803895 (2019).

    PubMed  Google Scholar 

  145. Shevach, M. et al. Omentum ECM-based hydrogel as a platform for cardiac cell delivery. Biomed. Mater. 10, 034106 (2015).

    PubMed  Google Scholar 

  146. Chan, V., Raman, R., Cvetkovic, C. & Bashir, R. Enabling microscale and nanoscale approaches for bioengineered cardiac tissue. ACS Nano 7, 1830–1837 (2013).

    CAS  PubMed  Google Scholar 

  147. Chang, W. G. & Niklason, L. E. A short discourse on vascular tissue engineering. NPJ Regen. Med. 2, 7 (2017).

    PubMed  PubMed Central  Google Scholar 

  148. Masumoto, H. et al. Human iPS cell-engineered cardiac tissue sheets with cardiomyocytes and vascular cells for cardiac regeneration. Sci. Rep. 4, 6716 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Masumoto, H. et al. Pluripotent stem cell-engineered cell sheets reassembled with defined cardiovascular populations ameliorate reduction in infarct heart function through cardiomyocyte-mediated neovascularization. Stem Cell 30, 1196–1205 (2012).

    CAS  Google Scholar 

  150. Shimizu, T. et al. Polysurgery of cell sheet grafts overcomes diffusion limits to produce thick, vascularized myocardial tissues. FASEB J. 20, 708–710 (2006).

    CAS  PubMed  Google Scholar 

  151. Rakusan, K., Flanagan, M. F., Geva, T., Southern, J. & Van Praagh, R. Morphometry of human coronary capillaries during normal growth and the effect of age in left ventricular pressure-overload hypertrophy. Circulation 86, 38–46 (1992).

    CAS  PubMed  Google Scholar 

  152. Rouwkema, J., Rivron, N. C. & van Blitterswijk, C. A. Vascularization in tissue engineering. Trends Biotechnol. 26, 434–441 (2008).

    CAS  PubMed  Google Scholar 

  153. Dvir, T. et al. Prevascularization of cardiac patch on the omentum improves its therapeutic outcome. Proc. Natl Acad. Sci. USA 106, 14990–14995 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Anderson, E. M. et al. VEGF and IGF delivered from alginate hydrogels promote stable perfusion recovery in ischemic hind limbs of aged mice and young rabbits. J. Vasc. Res. 54, 288–298 (2017).

    CAS  PubMed  Google Scholar 

  155. Miyagi, Y. et al. Biodegradable collagen patch with covalently immobilized VEGF for myocardial repair. Biomaterials 32, 1280–1290 (2011).

    CAS  PubMed  Google Scholar 

  156. Sekine, H. et al. Endothelial cell coculture within tissue-engineered cardiomyocyte sheets enhances neovascularization and improves cardiac function of ischemic hearts. Circulation 118 (Suppl. 14), S145–S152 (2008).

    CAS  PubMed  Google Scholar 

  157. Davies, N. H., Schmidt, C., Bezuidenhout, D. & Zilla, P. Sustaining neovascularization of a scaffold through staged release of vascular endothelial growth factor-A and platelet-derived growth factor-BB. Tissue Eng. Part A 18, 26–34 (2012).

    CAS  PubMed  Google Scholar 

  158. Lesman, A., Gepstein, L. & Levenberg, S. in Cardiac Tissue Engineering Ch. 12 (eds Radisic, M. & Black, L. D.) 131–137 (Springer, 2014).

  159. Levenberg, S. et al. Engineering vascularized skeletal muscle tissue. Nat. Biotechnol. 23, 879–884 (2005).

    CAS  PubMed  Google Scholar 

  160. Caspi, O. et al. Tissue engineering of vascularized cardiac muscle from human embryonic stem cells. Circ. Res. 100, 263–272 (2007).

    CAS  PubMed  Google Scholar 

  161. Lesman, A. et al. Transplantation of a tissue-engineered human vascularized cardiac muscle. Tissue Eng. Part A 16, 115–125 (2010).

    CAS  PubMed  Google Scholar 

  162. Gao, L. et al. Large cardiac muscle patches engineered from human induced-pluripotent stem cell–derived cardiac cells improve recovery from myocardial infarction in swine. Circulation 137, 1712–1730 (2018).

    PubMed  Google Scholar 

  163. Streeter, D. D. Jr., Spotnitz, H. M., Patel, D. P., Ross, J. Jr. & Sonnenblick, E. H. Fiber orientation in the canine left ventricle during diastole and systole. Circ. Res. 24, 339–347 (1969).

    PubMed  Google Scholar 

  164. Rohmer, D., Sitek, A. & Gullberg, G. T. Reconstruction and visualization of fiber and laminar structure in the normal human heart from ex vivo diffusion tensor magnetic resonance imaging (DTMRI) data. Invest. Radiol. 42, 777–789 (2007).

    PubMed  Google Scholar 

  165. Lee, J. M., Sing, S. L., Tan, E. Y. S. & Yeong, W. Y. Bioprinting in cardiovascular tissue engineering: a review. Int. J. Bioprinting 2, 27–36 (2016).

    CAS  Google Scholar 

  166. Cui, H. et al. 4D physiologically adaptable cardiac patch: a 4-month in vivo study for the treatment of myocardial infarction. Sci. Adv. 6, eabb5067 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  167. Jain, R. K. Molecular regulation of vessel maturation. Nat. Med. 9, 685–693 (2003).

    CAS  PubMed  Google Scholar 

  168. Kolesky, D. B., Homan, K. A., Skylar-Scott, M. A. & Lewis, J. A. Three-dimensional bioprinting of thick vascularized tissues. Proc. Natl Acad. Sci. USA 113, 3179–3184 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  169. Noor, N. et al. 3D printing of personalized thick and perfusable cardiac patches and hearts. Adv. Sci. 6, 1900344 (2019).

    Google Scholar 

  170. Zhu, W. et al. Direct 3D bioprinting of prevascularized tissue constructs with complex microarchitecture. Biomaterials 124, 106–115 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  171. Chaturvedi, R. R. et al. Patterning vascular networks in vivo for tissue engineering applications. Tissue Eng. C. Methods 21, 509–517 (2015).

    CAS  Google Scholar 

  172. Raghavan, S., Nelson, C. M., Baranski, J. D., Lim, E. & Chen, C. S. Geometrically controlled endothelial tubulogenesis in micropatterned gels. Tissue Eng. Part A 16, 2255–2263 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Arvatz, S., Wertheim, L., Fleischer, S., Shapira, A. & Dvir, T. Channeled ECM-based nanofibrous hydrogel for engineering vascularized cardiac tissues. Nanomaterials 9, 689 (2019).

    CAS  PubMed Central  Google Scholar 

  174. Miri, A. K. et al. Microfluidics-enabled multimaterial maskless stereolithographic bioprinting. Adv. Mater. 30, e1800242 (2018).

    PubMed  Google Scholar 

  175. Chen, L., Kenkel, S. M., Hsieh, P. H., Gryka, M. C. & Bhargava, R. Freeform three-dimensionally printed microchannels via surface-initiated photopolymerization combined with sacrificial molding. ACS Appl. Mater. Interfaces 12, 50105–50112 (2020).

    CAS  PubMed  Google Scholar 

  176. Grigoryan, B. et al. Multivascular networks and functional intravascular topologies within biocompatible hydrogels. Science 364, 458–464 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  177. Skylar-Scott, M. A. et al. Biomanufacturing of organ-specific tissues with high cellular density and embedded vascular channels. Sci. Adv. 5, eaaw2459 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  178. Miller, J. S. et al. Rapid casting of patterned vascular networks for perfusable engineered three-dimensional tissues. Nat. Mater. 11, 768–774 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  179. Ye, X. et al. A biodegradable microvessel scaffold as a framework to enable vascular support of engineered tissues. Biomaterials 34, 10007–10015 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  180. Bettinger, C. J. et al. Silk fibroin microfluidic devices. Adv. Mater. 19, 2847–2850 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  181. Zhang, B. et al. Biodegradable scaffold with built-in vasculature for organ-on-a-chip engineering and direct surgical anastomosis. Nat. Mater. 15, 669–678 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  182. Asulin, M., Michael, I., Shapira, A. & Dvir, T. One-step 3D printing of heart patches with built-in electronics for performance regulation. Adv. Sci. 8, 2004205 (2021).

    Google Scholar 

  183. Zimmermann, W.-H. et al. Engineered heart tissue grafts improve systolic and diastolic function in infarcted rat hearts. Nat. Med. 12, 452–458 (2006).

    CAS  PubMed  Google Scholar 

  184. Didié, M. et al. Parthenogenetic stem cells for tissue-engineered heart repair. J. Clin. Invest. 123, 1285–1298 (2013).

    PubMed  PubMed Central  Google Scholar 

  185. Li, R.-K. et al. Survival and function of bioengineered cardiac grafts. Circulation 100 (Suppl. 19), II63–II69 (1999).

    CAS  PubMed  Google Scholar 

  186. Shudo, Y. et al. Novel regenerative therapy using cell-sheet covered with omentum flap delivers a huge number of cells in a porcine myocardial infarction model. J. Thorac. Cardiovasc. Surg. 142, 1188–1196 (2011).

    PubMed  Google Scholar 

  187. Fujita, B. & Zimmermann, W.-H. Myocardial tissue engineering strategies for heart repair: current state of the art. Interact. Cardiovasc. Thorac. Surg. 27, 916–920 (2018).

    PubMed  Google Scholar 

  188. Nguyen, P. K., Neofytou, E., Rhee, J.-W. & Wu, J. C. Potential strategies to address the major clinical barriers facing stem cell regenerative therapy for cardiovascular disease: a review. JAMA Cardiol. 1, 953–962 (2016).

    PubMed  PubMed Central  Google Scholar 

  189. Stapleton, L., Zhu, Y., Woo, Y.-P. J. & Appel, E. Engineered biomaterials for heart disease. Curr. Opin. Biotechnol. 66, 246–254 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  190. Moroni, L. & Elisseeff, J. H. Biomaterials engineered for integration. Mater. Today 11, 44–51 (2008).

    CAS  Google Scholar 

  191. Orlando, G., Soker, S. & Stratta, R. J. Organ bioengineering and regeneration as the new Holy Grail for organ transplantation. Ann. Surg. 258, 221–232 (2013).

    PubMed  Google Scholar 

  192. Ott, H. C. et al. Perfusion-decellularized matrix: using nature’s platform to engineer a bioartificial heart. Nat. Med. 14, 213–221 (2008).

    CAS  PubMed  Google Scholar 

  193. Lu, T.-Y. et al. Repopulation of decellularized mouse heart with human induced pluripotent stem cell-derived cardiovascular progenitor cells. Nat. Commun. 4, 2307 (2013).

    PubMed  Google Scholar 

  194. Guyette, J. P. et al. Bioengineering human myocardium on native extracellular matrix. Circ. Res. 118, 56–72 (2016).

    CAS  PubMed  Google Scholar 

  195. MacQueen, L. A. et al. A tissue-engineered scale model of the heart ventricle. Nat. Biomed. Eng. 2, 930–941 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  196. Corbett, D. C., Olszewski, E. & Stevens, K. A FRESH take on resolution in 3D bioprinting. Trends Biotechnol. 37, 1153–1155 (2019).

    CAS  PubMed  Google Scholar 

  197. Lee, A. et al. 3D bioprinting of collagen to rebuild components of the human heart. Science 365, 482–487 (2019).

    CAS  PubMed  Google Scholar 

  198. Hinton, T. J. et al. Three-dimensional printing of complex biological structures by freeform reversible embedding of suspended hydrogels. Sci. Adv. 1, e1500758 (2015).

    PubMed  PubMed Central  Google Scholar 

  199. Mirdamadi, E., Tashman, J. W., Shiwarski, D. J., Palchesko, R. N. & Feinberg, A. W. FRESH 3D bioprinting a full-size model of the human heart. ACS Biomater. Sci. Eng. 6, 6453–6459 (2020).

    CAS  PubMed  Google Scholar 

  200. Shapira, A., Noor, N., Asulin, M. & Dvir, T. Stabilization strategies in extrusion-based 3D bioprinting for tissue engineering. Appl. Phys. Rev. 5, 041112 (2018).

    Google Scholar 

  201. Shapira, A., Noor, N., Oved, H. & Dvir, T. Transparent support media for high resolution 3D printing of volumetric cell-containing ECM structures. Biomed. Mater. 15, 045018 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  202. Kupfer, M. E. et al. In situ expansion, differentiation and electromechanical coupling of human cardiac muscle in a 3D bioprinted, chambered organoid. Circ. Res. 127, 207–224 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  203. Romagnuolo, R. et al. Human embryonic stem cell-derived cardiomyocytes regenerate the infarcted pig heart but induce ventricular tachyarrhythmias. Stem Cell Rep. 12, 967–981 (2019).

    Google Scholar 

  204. Liu, Y.-W. et al. Human embryonic stem cell–derived cardiomyocytes restore function in infarcted hearts of non-human primates. Nat. Biotechnol. 36, 597–605 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  205. Menasché, P. et al. Transplantation of human embryonic stem cell-derived cardiovascular progenitors for severe ischemic left ventricular dysfunction. J. Am. Coll. Cardiol. 71, 429–438 (2018).

    PubMed  Google Scholar 

  206. Cyranoski, D. ‘Reprogrammed’ stem cells approved to mend human hearts for the first time. Nature 557, 619–620 (2018).

    CAS  PubMed  Google Scholar 

  207. Shapira, A. & Dvir, T. 3D tissue and organ printing — hope and reality. Adv. Sci. 8, 2003751 (2021).

    CAS  Google Scholar 

  208. Yadid, M., Sela, G., Pavlov, D. A. & Landesberg, A. Adaptive control of cardiac contraction to changes in loading: from theory of sarcomere dynamics to whole-heart function. Pflügers Arch. 462, 49–60 (2011).

    CAS  PubMed  Google Scholar 

  209. Leor, J. et al. Bioengineered cardiac grafts: a new approach to repair the infarcted myocardium? Circulation 102, III56–III61 (2000).

    CAS  PubMed  Google Scholar 

  210. Leor, J. et al. Intracoronary injection of in situ forming alginate hydrogel reverses left ventricular remodeling after myocardial infarction in Swine. J. Am. Coll. Cardiol. 54, 1014–1023 (2009).

    PubMed  Google Scholar 

  211. Zhang, S. et al. A self-assembly pathway to aligned monodomain gels. Nat. Mater. 9, 594–601 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  212. Montgomery, M. et al. Flexible shape-memory scaffold for minimally invasive delivery of functional tissues. Nat. Mater. 16, 1038–1046 (2017).

    CAS  PubMed  Google Scholar 

  213. Sapir, Y., Polyak, B. & Cohen, S. Cardiac tissue engineering in magnetically actuated scaffolds. Nanotechnology 25, 014009 (2013).

    PubMed  PubMed Central  Google Scholar 

  214. Shevach, M., Fleischer, S., Shapira, A. & Dvir, T. Gold nanoparticle-decellularized matrix hybrids for cardiac tissue engineering. Nano Lett. 14, 5792–5796 (2014).

    CAS  PubMed  Google Scholar 

  215. Eschenhagen, T. et al. Three-dimensional reconstitution of embryonic cardiomyocytes in a collagen matrix: a new heart muscle model system. FASEB J. 11, 683–694 (1997).

    CAS  PubMed  Google Scholar 

  216. Bayer, J. D., Blake, R. C., Plank, G. & Trayanova, N. A. A novel rule-based algorithm for assigning myocardial fiber orientation to computational heart models. Ann. Biomed. Eng. 40, 2243–2254 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  217. Wilson, A. J. et al. Myocardial Laminar Organization Is Retained in Angiotensin-Converting Enzyme Inhibitor Treated SHRs. Exp. Mech. 61, 31–40 (2021).

    CAS  Google Scholar 

  218. Fleischer, S. et al. Spring-like fibers for cardiac tissue engineering. Biomaterials 34, 8599–8606 (2013).

    CAS  PubMed  Google Scholar 

  219. Macchiarelli, G. et al. A micro-anatomical model of the distribution of myocardial endomysial collagen. Histol. Histopathol. 17, 699–706 (2002).

    CAS  PubMed  Google Scholar 

  220. Hasegawa, T., Visovatti, S. H., Hyman, M. C., Hayasaki, T. & Pinsky, D. J. Heterotopic vascularized murine cardiac transplantation to study graft arteriopathy. Nat. Protoc. 2, 471–480 (2007).

    CAS  PubMed  Google Scholar 

  221. Gloschat, C. et al. RHYTHM: an open source imaging toolkit for cardiac panoramic optical mapping. Sci. Rep. 8, 2921 (2018).

    PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Contributions

All the authors researched data for the article, provided substantial contributions to discussions of its content, wrote the article, and undertook review and/or editing of the manuscript before submission.

Corresponding author

Correspondence to Tal Dvir.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Cardiology thanks N. Hibino, J. Kluin, T. Shimizu and W. Zimmermann for their contribution to the peer review of this work.

Publisher’s note

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

Glossary

Anisotropy

A property of a material that allows it to show different characteristics in different directions, as opposed to isotropy. In this Review, we refer to the longitudinal alignment of the cells in one direction.

Micropatterning

Creating microscale patterns in a material or on its surface.

Surface functionalization

Altering the surface properties of a material to achieve specific function or activity, such as by binding cells or molecules.

Cyclic stretch

The application of periodic stretch–release cycles to cell-containing material to simulate the physiological mechanical strains that cells in the native heart experience.

Hydrogels

A crosslinked hydrophilic polymer that does not dissolve in water.

Melt electrospinning writing

A fabrication technique that uses polymer melts for controlled deposition of an electrospun fibre.

Conductive nanoparticles

Nano-scale particles possessing the physical property of being electrically conductive.

3D bioprinting

An additive manufacturing technique that uses cells and biocompatible materials as bio-inks to print living structures.

Photopolymerization

The polymerization or solidification of solubilized monomers by exposure to light, usually ultraviolet.

Stereolithography

A layer-by-layer 3D printing process whereby a photopolymer is selectively cured by a moving laser beam, causing a controlled, local crosslinking of the material.

Sacrificial writing

The process of printing structures using fugitive materials that will later dissolve, leaving empty spaces in the printed structure, as used for the bioprinting of blood vessels.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yadid, M., Oved, H., Silberman, E. et al. Bioengineering approaches to treat the failing heart: from cell biology to 3D printing. Nat Rev Cardiol 19, 83–99 (2022). https://doi.org/10.1038/s41569-021-00603-7

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41569-021-00603-7

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research