Skip to main content

Tissue-Engineered Stem Cell Models of Cardiovascular Diseases

  • Chapter
  • First Online:
Cardiovascular Regenerative Medicine

Abstract

In vitro disease modeling can provide compelling insights into the molecular underpinnings of various pathological conditions. Traditionally, researchers have used two-dimensional (2D) cell culture methods to elucidate the molecular mechanisms that drive diseases. However, this method of cell culture fails to recapitulate the complex, three-dimensional (3D) microenvironment that is present in vivo. Recently, tissue-engineered models have been generated to create culture systems that better recapitulate the complex 3D microenvironments found in living tissue. By using these engineered models, more relevant data may be produced to unravel the molecular underpinnings of disease states and potentially translate into new therapeutic strategies.

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

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 79.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 99.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 109.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Works Cited

  1. Thomson J, et al. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282:1145–7.

    Article  CAS  Google Scholar 

  2. Itskovitz-Eldor J, et al. Differentiation of human embryonic stem cells into embryoid bodies compromising the three embryonic germ layers. Mol Med. 2000;6:88–95.

    Article  CAS  Google Scholar 

  3. Mclaren A. Ethical and social considerations of stem cell research. Nature. 2001;414:129–31.

    Article  CAS  Google Scholar 

  4. Takahashi K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861–72. https://doi.org/10.1016/j.cell.2007.11.019.

    Article  CAS  Google Scholar 

  5. Später D, Hansson EM, Zangi L, Chien KR. How to make a cardiomyocyte. Development. 2014;141:4418–31.

    Article  Google Scholar 

  6. Lee TM, et al. Pediatric cardiomyopathies. Circ Res. 2017;121:855–73.

    Article  CAS  Google Scholar 

  7. Hansen A, et al. Development of a drug screening platform based on engineered heart tissue. Circ Res. 2010;107:35–44.

    Article  CAS  Google Scholar 

  8. Schwan J, et al. Anisotropic engineered heart tissue made from laser-cut decellularized myocardium. Sci Rep. 2016;6:32068.

    Article  CAS  Google Scholar 

  9. Haraguchi Y, et al. Fabrication of functional three-dimensional tissues by stacking cell sheets in vitro. Nat Protoc. 2012;7:850–8.

    Article  CAS  Google Scholar 

  10. Jackman CP, Carlson AL, Bursac N. Dynamic culture yields engineered myocardium with near-adult functional output. Biomaterials. 2016;111:66–79.

    Article  CAS  Google Scholar 

  11. Bian W, Badie N, Himel HD, Bursac N. Robust T-tubulation and maturation of cardiomyocytes using tissue-engineered epicardial mimetics. Biomaterials. 2014;35:3819–28.

    Article  CAS  Google Scholar 

  12. Tiburcy M, et al. Defined engineered human myocardium with advanced maturation for applications in heart failure modelling and repair. Circulation. 2017;116:024145. https://doi.org/10.1161/CIRCULATIONAHA.116.024145.

    Article  CAS  Google Scholar 

  13. MacQueen LA, et al. A tissue-engineered scale model of the heart ventricle. Nat Biomed Eng. 2018;2:930.

    Article  CAS  Google Scholar 

  14. Wu B, et al. Developmental mechanisms of aortic valve malformation and disease. Annu Rev Physiol. 2017;79:21–41.

    Article  CAS  Google Scholar 

  15. Arevalos CA, et al. Valve interstitial cells act in a Pericyte manner promoting angiogensis and invasion by valve endothelial cells. Ann Biomed Eng. 2016;44:1–17.

    Article  Google Scholar 

  16. Varzideh F, et al. Human cardiomyocytes undergo enhanced maturation in embryonic stem cell-derived organoid transplants. Biomaterials. 2018;192:537–50.

    Article  Google Scholar 

  17. Voges HK, et al. Development of a human cardiac organoid injury model reveals innate regenerative potential. Development. 2017;144:1118–27.

    Article  CAS  Google Scholar 

  18. Mills RJ, et al. Functional screening in human cardiac organoids reveals a metabolic mechanism for cardiomyocyte cell cycle arrest. Proc Natl Acad Sci. 2017;114:E8372. https://doi.org/10.1073/pnas.1707316114.

    Article  CAS  PubMed  Google Scholar 

  19. Feric NT, Radisic M. Maturing human pluripotent stem cell-derived cardiomyocytes in human engineered cardiac tissues. Adv Drug Deliv Rev. 2016;96:110–34.

    Article  CAS  Google Scholar 

  20. Robertson C, Tran D, George S. Concise review: maturation phases of human pluripotent stem cell-derived cardiomyocytes. Stem Cells. 2013;31:1–17.

    Article  Google Scholar 

  21. French A, et al. Enabling consistency in pluripotent stem cell-derived products for research and development and clinical applications through material standards. Stem Cells Transl Med. 2015;4:217–23.

    Article  CAS  Google Scholar 

  22. McMullen JR, Jennings GL. Differences between pathological and physiological cardiac hypertrophy: novel therapeutic strategies to treat heart failure. Clin Exp Pharmacol Physiol. 2007;34:255–62.

    Article  CAS  Google Scholar 

  23. Feinberg AW, et al. Controlling the contractile strength of engineered cardiac muscle by hierarchal tissue architecture. Biomaterials. 2012;33:5732–41.

    Article  CAS  Google Scholar 

  24. Ruan J-L, et al. Mechanical stress conditioning and electrical stimulation promote contractility and force maturation of induced pluripotent stem cell-derived human cardiac tissue clinical perspective. Circulation. 2016;134:1557–67.

    Article  CAS  Google Scholar 

  25. Schwan J, Campbell SG. Prospects for in vitro myofilament maturation in stem cell-derived cardiac myocytes. Biomark Insights. 2015;2015:91–103.

    Google Scholar 

  26. Chong JJH, et al. Human embryonic-stem-cell-derived cardiomyocytes regenerate non-human primate hearts. Nature. 2014;510:273–7.

    Article  CAS  Google Scholar 

  27. Hirt MN, et al. Functional improvement and maturation of rat and human engineered heart tissue by chronic electrical stimulation. J Mol Cell Cardiol. 2014;74:151–61.

    Article  CAS  Google Scholar 

  28. Mihic A, et al. The effect of cyclic stretch on maturation and 3D tissue formation of human embryonic stem cell-derived cardiomyocytes. Biomaterials. 2014;35:2798–808.

    Article  CAS  Google Scholar 

  29. Ronaldson-Bouchard K, et al. Advanced maturation of human cardiac tissue grown from pluripotent stem cells. Nature. 2018;556:239–43.

    Article  CAS  Google Scholar 

  30. van Mil A, et al. Modelling inherited cardiac disease using human induced pluripotent stem cell-derived cardiomyocytes: progress, pitfalls, and potential. Cardiovasc Res. 2018;114(14):1828–42. https://doi.org/10.1093/cvr/cvy208.

  31. Eschenhagen T, Carrier L. Cardiomyopathy phenotypes in human-induced pluripotent stem cell-derived cardiomyocytes—a systematic review. Pflugers Arch. 2018; https://doi.org/10.1093/cvr/cvy208.

  32. Mestroni L, Brun F, Spezzacatene A, Sinagra G, MRG T. Genetic causes of dilated cardiomyopathy. Prog Pediatr Cardiol. 2015;37:13–8.

    Article  Google Scholar 

  33. Hinson JT, et al. Titin mutations in iPS cells define sarcomere insufficiency as a cause of dilated cardiomyopathy. Science. 2015;349:982–6.

    Article  CAS  Google Scholar 

  34. Boudou T, et al. A microfabricated platform to measure and manipulate the mechanics of engineered cardiac microtissues. Tissue Eng Part A. 2012;18:910–9.

    Article  CAS  Google Scholar 

  35. Tiburcy M, Meyer T, Soong PL, Zimmermann W-H. Collagen-based engineered heart muscle. Methods Mol Biol. 2014;1181:167–76.

    Article  Google Scholar 

  36. Streckfuss-Bömeke K, et al. Severe DCM phenotype of patient harboring RBM20 mutation S635A can be modeled by patient-specific induced pluripotent stem cell-derived cardiomyocytes. J Mol Cell Cardiol. 2017;113:9–21.

    Article  Google Scholar 

  37. Marian AJ, Braunwald E. Hypertrophic cardiomyopathy: genetics, pathogenesis, clinical manifestations, diagnosis, and therapy. Circ Res. 2017;121:749–70.

    Article  CAS  Google Scholar 

  38. Mosqueira D, et al. CRISPR/Cas9 editing in human pluripotent stem cell-cardiomyocytes highlights arrhythmias, hypocontractility, and energy depletion as potential therapeutic targets for hypertrophic cardiomyopathy. Eur Heart J. 2018;39:3879–92.

    Article  Google Scholar 

  39. Smith JGW, et al. Isogenic pairs of hiPSC-CMs with hypertrophic cardiomyopathy/LVNC-associated ACTC1 E99K mutation unveil differential functional deficits. Stem Cell Reports. 2018;11:1226–43.

    Article  CAS  Google Scholar 

  40. Daw EW, et al. Genome-wide mapping of modifier chromosomal loci for human hypertrophic cardiomyopathy. Hum Mol Genet. 2007;16:2463–71.

    Article  CAS  Google Scholar 

  41. Hinson JT, et al. Integrative analysis of PRKAG2 cardiomyopathy iPS and microtissue models identifies AMPK as a regulator of metabolism, survival, and fibrosis. Cell Rep. 2016;17:3292–304.

    Article  CAS  Google Scholar 

  42. Marcelo KL, Goldie LC, Hirschi KK. Regulation of endothelial cell differentiation and specification. Circ Res. 2013;112:1272–87.

    Article  CAS  Google Scholar 

  43. Masumura T, Yamamoto K, Shimizu N, Obi S, Ando J. Shear stress increases expression of the arterial endothelial marker ephrinB2 in murine ES cells via the VEGF-notch signaling pathways. Arterioscler Thromb Vasc Biol. 2009;29:2125–31.

    Article  CAS  Google Scholar 

  44. Sivarapatna A, et al. Arterial specification of endothelial cells derived from human induced pluripotent stem cells in a biomimetic flow bioreactor. Biomaterials. 2015;53:621–33.

    Article  CAS  Google Scholar 

  45. Yoder MC. Endothelial stem and progenitor cells (stem cells): (2017 Grover Conference Series). Pulm Circ. 2018;8 https://doi.org/10.1177/2045893217743950.

    Article  Google Scholar 

  46. Lee SJ, Kim KH, Yoon YS. Generation of human pluripotent stem cell-derived endothelial cells and their therapeutic utility. Curr Cardiol Rep. 2018;20(45):45.

    Article  Google Scholar 

  47. Lian X, et al. Efficient differentiation of human pluripotent stem cells to endothelial progenitors via small-molecule activation of WNT signaling. Stem Cell Reports. 2014;3:804–16.

    Article  CAS  Google Scholar 

  48. Patsch C, et al. Generation of vascular endothelial and smooth muscle cells from human pluripotent stem cells. Nat Cell Biol. 2015;17:994–1003.

    Article  CAS  Google Scholar 

  49. Prasain N, et al. Differentiation of human pluripotent stem cells to cells similar to cord-blood endothelial colony-forming cells. Nat Biotechnol. 2014;32:1151–7.

    Article  CAS  Google Scholar 

  50. Ellis MW, Luo J, Qyang Y. Modeling elastin-associated vasculopathy with patient induced pluripotent stem cells and tissue engineering. Cell Mol Life Sci. 2018. https://doi.org/10.1007/s00018-018-2969-7 [pii].

    Article  Google Scholar 

  51. Majesky MW, Mummery CL. Smooth muscle diversity from human pluripotent cells. Nat Biotechnol. 2012;30:152–4.

    Article  CAS  Google Scholar 

  52. Cheung C, Bernardo AS, Trotter MW, Pedersen RA, Sinha S. Generation of human vascular smooth muscle subtypes provides insight into embryological origin-dependent disease susceptibility. Nat Biotechnol. 2012;30:165–73.

    Article  CAS  Google Scholar 

  53. Xie C, Ritchie RP, Huang H, Zhang J, Chen YE. Smooth muscle cell differentiation in vitro: models and underlying molecular mechanisms. Arterioscler Thromb Vasc Biol. 2011;31:1485–94.

    Article  CAS  Google Scholar 

  54. Rensen SS, Doevendans PA, van Eys GJ. Regulation and characteristics of vascular smooth muscle cell phenotypic diversity. Neth Hear J. 2007;15:100–8.

    Article  CAS  Google Scholar 

  55. Xie CQ, et al. A highly efficient method to differentiate smooth muscle cells from human embryonic stem cells. Arterioscler Thromb Vasc Biol. 2007;27:e311–2.

    Article  CAS  Google Scholar 

  56. Wanjare M, Kuo F, Gerecht S. Derivation and maturation of synthetic and contractile vascular smooth muscle cells from human pluripotent stem cells. Cardiovasc Res. 2013;97:321–30.

    Article  CAS  Google Scholar 

  57. Wanjare M, Kusuma S, Gerecht S. Defining differences among perivascular cells derived from human pluripotent stem cells. Stem Cell Reports. 2014;2:746.

    Article  CAS  Google Scholar 

  58. Wanjare M, Agarwal N, Gerecht S. Biomechanical strain induces elastin and collagen production in human pluripotent stem cell-derived vascular smooth muscle cells. Am J Physiol Cell Physiol. 2015;309:C271–81.

    Article  CAS  Google Scholar 

  59. Eoh JH, et al. Enhanced elastin synthesis and maturation in human vascular smooth muscle tissue derived from induced-pluripotent stem cells. Acta Biomater. 2017;52:49–59.

    Article  CAS  Google Scholar 

  60. Niklason LE, et al. Functional arteries grown in vitro. Science. 1999;284:489–93.

    Article  CAS  Google Scholar 

  61. Syedain ZH, et al. A completely biological ‘off-the-shelf’ arteriovenous graft that recellularizes in baboons. Sci Transl Med. 2017;9:pii: eaan4209.

    Article  Google Scholar 

  62. Atchison L, Zhang H, Cao K, Truskey GA. A tissue engineered blood vessel model of Hutchinson-Gilford Progeria Syndrome using human iPSC-derived smooth muscle cells. Sci Rep. 2017;7(8168):8168.

    Article  Google Scholar 

  63. Gui L, et al. Implantable tissue-engineered blood vessels from human induced pluripotent stem cells. Biomaterials. 2016;102:120–9.

    Article  CAS  Google Scholar 

  64. Fernandez CE, et al. Human vascular microphysiological system for in vitro drug screening. Sci Rep. 2016;6:21579.

    Article  CAS  Google Scholar 

  65. Strobel HA, Calamari EL, Alphonse B, Hookway TA, Rolle MW. Fabrication of custom agarose wells for cell seeding and tissue ring self-assembly using 3D-printed molds. J Vis Exp. 2018; https://doi.org/10.3791/56618.

  66. Dash BC, et al. Tissue-engineered vascular rings from human iPSC-derived smooth muscle cells. Stem Cell Reports. 2016;7:19–28.

    Article  CAS  Google Scholar 

  67. Ren Y, et al. Small molecule Wnt inhibitors enhance the efficiency of BMP-4-directed cardiac differentiation of human pluripotent stem cells. J Mol Cell Cardiol. 2011;51:280–7.

    Article  CAS  Google Scholar 

  68. Gui L, et al. Biomaterials implantable tissue-engineered blood vessels from human induced pluripotent stem cells. Biomaterials. 2016;102:120–9.

    Article  CAS  Google Scholar 

  69. Zhang D, et al. Tissue-engineered cardiac patch for advanced functional maturation of human ESC-derived cardiomyocytes. Biomaterials. 2013;34:5813–20.

    Article  CAS  Google Scholar 

  70. Abilez OJ, et al. Passive stretch induces structural and functional maturation of engineered heart muscle as predicted by computational modeling. Stem Cells. 2018;36:265–77.

    Article  CAS  Google Scholar 

  71. Khan M, et al. Evaluation of changes in morphology and function of human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) cultured on an aligned-nanofiber cardiac patch. PLoS One. 2015;10:1–19.

    Google Scholar 

Download references

Funding Acknowledgments

This work was supported by 1R01HL116705, 1R01HL131940, DOD 11959515, Connecticut’s Regenerative Medicine Research Fund (CRMRF) (all to YQ), P.D. Soros Fellowship for New Americans and a NIH/NIGMS Medical Scientist Training Program Grant (T32GM007205) (both to LRS), NIH 1F31HL143928-01 (to CWA), and 1R01HL136590 (to SC).

Competing Interests L.E.N. is a founder and shareholder in Humacyte, Inc., which is a regenerative medicine company. Humacyte produces engineered blood vessels from allogeneic smooth muscle cells for vascular surgery. L.E.N.’s spouse has equity in Humacyte, and L.E.N. serves on Humacyte’s Board of Directors. L.E.N. is an inventor on patents that are licensed to Humacyte and that produce royalties for L.E.N. L.E.N. has received an unrestricted research gift to support research in her laboratory at Yale. Humacyte did not fund this review, and Humacyte did not influence the writing of this review.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Yibing Qyang .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2019 Springer Nature Switzerland AG

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Anderson, C.W. et al. (2019). Tissue-Engineered Stem Cell Models of Cardiovascular Diseases. In: Serpooshan, V., Wu, S. (eds) Cardiovascular Regenerative Medicine. Springer, Cham. https://doi.org/10.1007/978-3-030-20047-3_1

Download citation

  • DOI: https://doi.org/10.1007/978-3-030-20047-3_1

  • Published:

  • Publisher Name: Springer, Cham

  • Print ISBN: 978-3-030-20046-6

  • Online ISBN: 978-3-030-20047-3

  • eBook Packages: MedicineMedicine (R0)

Publish with us

Policies and ethics