Open access peer-reviewed chapter

Human-Induced Pluripotent Stem Cell-Derived Engineered Cardiac Tissues

Written By

Takeichiro Nakane, Hidetoshi Masumoto and Bradley B. Keller

Submitted: 05 September 2017 Reviewed: 11 October 2017 Published: 20 December 2017

DOI: 10.5772/intechopen.71621

From the Edited Volume

Stem Cells in Clinical Practice and Tissue Engineering

Edited by Rakesh Sharma

Chapter metrics overview

1,320 Chapter Downloads

View Full Metrics

Abstract

By combining tissue engineering techniques with human-induced pluripotent stem cell (hiPSC) technology, human-derived engineered cardiac tissues (ECTs) have been developed using several cell lineage compositions and 3-dimensional geometries. Although hiPSC ECTs are relatively immature compared with native adult heart tissues, they have promising potential as a platform technology for drug-screening and disease modeling, and as grafts for hiPSC-based regenerative heart therapy. This chapter provides the focused overview of the current status of cardiac tissue engineering technology and its possible application.

Keywords

  • iPS cell
  • engineered cardiac tissue
  • tissue engineering
  • drug screening
  • disease modeling
  • cardiac regeneration

1. Introduction

With the progress of tissue engineering technology in the last decade, many kinds of engineered cardiac tissues (ECTs) have been developed and reported. These tissues possess striated myofibers which recapitulate unique contractile function of heart tissues. Takahashi, Yamanaka, and colleagues developed iPSCs from mice in 2006 and from human next year [1, 2]. Human iPS cells (hiPSCs) have the potential to differentiate into cardiomyocytes and other cardiac lineage cells. Recently, the efficiency of cardiac differentiation has rapidly improved, which makes it possible to robustly induce cardiac lineage cells [3, 4, 5, 6]. By using cardiac cells derived from hiPSCs as a cell source for ECT generation, the potential of ECTs has expanded. The present chapter overviews the current status of ECT technology and its possible application.

Advertisement

2. Methods for generating engineered cardiac tissues

2.1. Cell sheet technology

Okano, Shimizu and colleagues have developed a culture surface grafted with temperature-responsive polymer, poly(N-isopropyl acrylamide) (PIPAAm) [7]. This system enables confluent cells to detach themselves from the surface while maintaining a sheet structure simply by reducing the culture temperature. The cell sheets are fabricated without the use of exogenous matrices. It has endogenous matrix layer at the bottom side, and it secures biological attachment to the recipient heart surface within 30 min [8]. This scaffold-free tissue is a thin sheet, and three-dimensional thicker structure can be obtained by stacking several sheets. However, stacking more than three layers (approximately 80 μm thickness) triggers cell death at the center due to hypoxia [9]. At our lab, we overcame this limit by stacking cell sheets with gelatin hydrogel microspheres (GHM) [10]. Five-layered cell sheets with GHM displayed better cell viability in vitro compared with the control cell sheet without GHM. Furthermore, cell sheet modification with GHM improved the retention of grafted cell sheets on the rat heart epicardial surface after myocardial infarction. We succeeded in generating over 1 mm thick constructs from 15 sheets using this technology.

2.2. Biomaterials

Cardiomyocytes and other cells are embedded in biomaterials, such as collagen I, matrigel, and/or fibrin in a casting mold. Exogenous gel matrix promotes self-assembly of cells leading to form a tissue structure. The first successful cardiac tissue fabrication by this method was reported in the year 1997 [11]. Fixed anchors in these molds generate static strain, which enhances the cell alignment and contractile function. The geometry of mold controls the final tissue architecture, and a variety of tissue shapes have been formulated, including linear [12, 13, 14, 15, 16], circular [17], and mesh structures (Figure 1) [18, 19, 20]. By applying engineering manufacturing methods, it may be possible to automate the process of generating small-sized ECTs, leading to the development of high throughput in vitro analysis systems [21, 22]. In fact, recently, 3D bio-printing technology has been applied and provided the possibility of creating more complicated structures reproducibly by printing both scaffold matrix and living cells [23].

Figure 1.

Generation of a mesh ECT by biomaterial-based technique. (a) Schematic diagram for a mesh ECT generation. Cardiomyocytes (CM), endothelial cells (EC), and mural cells (MC) are induced from human iPS cells (h-iPSCs) using two different protocols. Cells and collagen-based matrix were combined and poured into custom polydimethylsiloxane (PDMS) molds at day 0 and cultured for 14 days. (b) Representative image of a PDMS mold (left) and a mesh ECT (right). Scale bar: 10 mm. (Reproduced from Ref. [20] with permission).

2.3. Prefabricated matrix

Biomaterials provide a ‘soft’ environment for the cells to grow in. On the other hand, synthetic microporous (‘spongy’) scaffolds made from alginate, collagen, and gelatin or other stiffer materials, such as polystyrene, PLLA (Poly-L-Lactide Acid), PLGA (Poly(lactic-co-glycolic acid)) and PCL (Polycaprolactone), have also been tested [24, 25]. The advantage of this method is that these rigid scaffolds contribute to engineering any desired structure and size with mechanical stability. However, the maturation of various mesodermal cells is matrix stiffness dependent, and therefore both the biomaterial gel and the supporting mold may impact cell maturation and survival [26].

2.4. Decellularized tissue

Deccllurization of a whole heart is achieved by the treatment with sodium dodecyl sulfate (SDS) and Triton X-100 under Langendorf perfusion [27]. The decellularized tissue maintains most of the tissue contents and function of the extracellular matrix. Hence, decellularized tissue provides a native scaffold which can support repopulation by mesoderm lineage cells. These decellularized tissues may be advantageous in the generation of in vitro cardiac tissues [28, 29].

Advertisement

3. Maturation of the tissue

3.1. Cellular contents

A heart tissue is composed of multiple cell types including cardiomyocytes, fibroblasts, endothelial cells, and smooth muscle cells. In human hearts, cardiomyocytes account for 25-50% of total cells, occupying around 70% of the whole volume [30]. Many reports have demonstrated that non-myocytes support the maturation of cardiomyocytes and play a significant role in both myocardial structure and function [13, 16, 19].

We induced cardiomyocytes (CM), endothelial cells (EC), and vascular mural cells (MC) from hiPSCs, and generated ECTs from several formulations of cardiomyocytes and non-myocytes, such as CM+EC, CM+MC, and CM+EC+MC [16]. According to in vitro force measurement, CM+EC+MC ECTs showed most advanced electrophysiological properties. Furthermore, histological analysis revealed CM+EC+MC possessed more unidirectionally aligned myofiber with mature sarcomeric structures (Figure 2).

Figure 2.

Electromechanical properties and structural maturation of hiPSC-ECTs. (a) Schematic diagrams for generationg 3 types of ECTs containing cardiomyocytes (CM), endothelial cells (EC), and/or mural cells (MC) (upper) and the proportions of each cell type (lower) [n = 8 (CM+ EC), 7 (CM+ MC), and 12 (CM+ EC+ MC). (b–d) Results of contractile force measurements [n = 8 (CM+ EC), 7 (CM+ MC), and 12 (CM+ EC+ MC)]. (b) Maximum capture rate (left), relaxation time (center) and excitation threshold voltage (right) (c) Relationship between active force and pacing frequency (2Hz to 3.5Hz). (d) Young’s modulus of ECTs. NS, not significant; *P < 0.05,**P < 0.01, ***P < 0.001. (e) Representative alignment analysis using cTnT-stained images after 2-dimensional (2D) and 3-dimensional (3D) culture for 3 types of ECTs (left). Calculated concentration parameter (κ ) (right) [n =3 (2D) and 5 (each 3D)]. Larger values of κ represent greater alignment to a single direction. cTnT, cardiac troponin T; Deg, degree. (f) Representative transmission electron microscopic images for each type of ECT cultured for 4 weeks. Arrows indicate myofibers. N, nucleus; Mt, mitochondria; I, I-band; A, A-band; Z, Z-line. (Reproduced from Ref. [16] with permission).

The mechanism for the improvement of tissue maturation as the result of a more complex lineage mixture is still unknown. Evidence is emerging that cardiomyocytes and fibroblasts are electrically coupled, which may modulate electrophysiologic function [31]. Direct interaction of different cell types or paracrine effects can be considered. It is reported that extracellular matrix derived from cardiac fibroblast supported the proliferation in vitro and indicated the usefulness of the coculture [32].

3.2. Effects of extended culture duration on CM and ECT maturation and function

Cardiomyocytes derived from pluripotent stem cells mature early in culture but are arrested at the late embryonic stage under 2-dimensional (2D) culture condition [33]. Meanwhile, it is widely recognized that cardiomyocytes in a 3-dimensional (3D) cardiac tissue acquire a more mature phenotype than those in 2D culture [25, 34].

In our study, we prolonged culture duration of mesh ECTs from 14 to 28days [20]. Long-cultured constructs showed the tendency to augment the active contractile force along with the increase of beating frequency from 1.5Hz to 2.5Hz and maintained greater force compared to 14-day constructs. The shift from a negative to a neutral force-frequency relationship in 28-day constructs represents sustained functional maturation as well as more rapid force generation and relaxation cycle, and the ability to capture higher pacing frequency. Additional culture duration enhanced myofiber alignment along with the expression of several genes related to ion channels and gap junction (Figure 3).

Figure 3.

Effects of extended culture duration on mesh ECT maturation. (a) Averaged normalized active force-time curves for mesh ECT cultured for either 14 (blue, D14) or 28 (red, D28) (n = 6) days under 2 Hz electrical stimulation. Contraction time (CT) or relaxation time (RT) represents the time of 90% increase or decrease of force respectively. Comparison of (b) contraction time 90% (n = 6; ***P < 0.01), (c) relaxation time 90% (***P < 0.001) at 2 Hz pacing, (d) maximum capture rate (n = 6; ***P < 0.001), (e) force-frequency relationship (n = 6; *P < 0.05 to ***P < 0.001 vs. D14), and (f) cardiomyocyte alignment concentration (κ), [n = 4 (D14) and n = 3 (D28); ***P < 0.001]. (g), (h) Q-PCR analysis (n = 3; *P < 0.05 to ***P < 0.001). cTnT level was lower in D28, and other genes were normalized to the value of cTnT expression. (Reproduced from Ref. [20] with permission).

3.3. Impact of culture condition on ECT maturation and function

In order to expand the dimension of ECTs, it is necessary to improve the distribution of oxygen and nutrient throughout tissues. Direct perfusion of culture medium reduces the gradients associated with diffusional mass transport between the construct surfaces and the inner phase and improves the microenvironmental conditions within ECTs. [35, 36, 37]. In addition to the increase of cellularity at the center of ECTs, perfusion system contributes to the maturation of cardiomyocytes and the tissue structure. It is reported that even the simple rocking dynamic culture yields engineered myocardium with near-adult functional output [38]. The system accelerates force generation and conduction velocity probably due to the activation of mTOR signaling.

For generating ECTs, especially from biomaterial-based technique, materials from other sources, such as collagen derived from rat or matrigel, have been widely used with xeno-serum containing culture medium. Tiburcy and colleagues modified their methods toward clinical application and developed a formulation for generating ECTs by using medical grade bovine collagen and maintaining them under serum-free defined condition [19]. This modification provided a proof-of-concept for a universally applicable technology for the engineering cardiac tissue. In addition, ECTs generated by this condition displayed advanced cardiomyocyte maturation compared to the conventional methods [19].

3.4. Role of mechanical loading on ECT maturation and function.

Several in vitro ECT studies revealed the impact of uniaxial mechanical loading on the alignment of cardiomyocytes and functional maturation. The simplest system for that is the two fixed anchors which generate static strain on the tissue between them. Furthermore, cyclic stress conditioning markedly increases cardiomyocyte hypertrophy and proliferation rates versus unconditioned constructs [13]. We demonstrated that ECTs displayed a broad spectrum of altered gene expression in response to cyclic stretch, reflecting a complex regulation of proliferation, differentiation, and architectural alignment of cardiomyocytes and non-cardiomyocytes within ECTs [39].

Electrical stimulation is considered to be another important cue for further maturation of tissues [40]. Nunes and colleagues subjected their engineered tissues, named biowires, on electrical stimulation at up to 6 Hz [41]. Biowires subjected to electrical stimulation had markedly increased myofibril ultrastructural organization, elevated conduction velocity and improved both electrophysiological and Ca2+ handling properties compared to non-stimulated controls. These changes were in agreement with cardiomyocyte maturation and were dependent on the stimulation frequency.

Another group created a system which applied combined electromechanical stimulation mimicking isovolumic contraction and confirmed the improvement of functional properties over electrical or mechanical stimulation alone. In the report, the timing of the combined stimulation greatly affected the electrophysiological properties of ECTs [42].

Advertisement

4. Applications of ECTs

4.1. Drug screening

Cardiotoxicity is one of the main causes of drug withdrawal from the market [43, 44]. To evaluate the safety and effectiveness of drugs, several kinds of pre-clinical studies are performed using non-human models. However, these models occasionally show incorrect results, such as pseudo-negative, due to the difference of electrophysiology in other species [45]. By using hiPSC technology, effects to humans can be examined from the early stage [46]. Moreover, hiPSC-derived ECTs are expected to be more specific and sensitive platforms for drug screening [14].

We have developed a 3D cardiac tissue model which reproduces Torsade de Pointes (TdP) showing both a typical polymorphic extracellular field potential and meandering spiral wave re-entry upon treatment with IKr channel blockers [47]. It is generated from hiPSC-derived cardiomyocytes and mesenchymal cells using the cell sheet technology. The appearance of TdP-like waveforms was significantly higher in this 3D model compared with 2D monolayer conditions indicating that the multilayered 3D structure is an essential factor for this arrhythmia along with the coexistence of non-cardiomyocytes.

Many groups have developed various high throughput screening formats, such as strip format [48] and heart-on-chip [21, 49], and confirmed the similarity of hiPSC-derived engineered tissues with native heart tissues [14, 50, 51, 52]. Video optical recording system set up in a usual CO2 incubator is also useful to continuously monitor contractile abilities of tissues [48].

Cardiomyocytes from hiPSCs are at the premature stage, and even ECTs generated to date remain immature compared to native adult myocardium. Hence, further cues for maturation as described above is necessary to imitate native tissues more precisely [51].

4.2. Disease modeling

iPS cells derived from a patient with a known genetic mutation for the disease can provide the disease model, which may offer a useful strategy for understanding the mechanism of the disease and exploring a new treatment modality.

Cardio-facio-cutaneous syndrome (CFCS) is one of the RASopathies, and cardiac abnormalities are the most common findings among CFCS, including hypertrophic cardiomyopathy (HCM) in approximately 40% of patients. Nearly 75% of patients with CFCS exhibit mutations in BRAF, which encodes a serine/threonine kinase and a direct effector of Ras. Cashman and colleagues created 3D human engineered cardiac tissue model of HCM using human cardiomyocytes yielded by directed differentiation of iPSCs established from a patient with CFCS carrying an activated BRAF mutation [53].

Chronic catecholamine overstimulation contributes to heart failure progression. Overstimulation of ECTs with norepinephrine provides a simulation of a human heart failure phenotype [19]. Tissues responded to chronic catecholamine toxicity with contractile dysfunction, cardiomyocyte hypertrophy, cardiomyocyte death, and NT-proBNP release, which are classical hallmarks of heart failure. Notably, the pathological phenotype could be partially or fully prevented by β1- or α1-adrenoreceptor blockade, demonstrating the applicability of ECTs in the in vitro simulation of heart failure and its prevention by pharmacological means.

4.3. Transplantation therapy

HiPSCs are now one of the most promising cell sources for cardiac regenerative cell therapy [54, 55, 56]. There are major methods of cell delivery, including intracoronary or intramuscular injection of dispersed cells and epicardial transplantation of engineered tissues [57]. It is possible to deliver a large number of differentiated cells with organized architecture by ECT implantation. The grafted tissues survive and support the heart wall, which overcomes the problem of poor retention rate following cell injection [34].

A variety of studies has revealed the efficacy of ECT implantation for myocardial structural and functional recovery in injured hearts of several animal models [6, 10, 58, 59]. We implanted ECTs in an athymic nude rat myocardial infarction model. ECTs with vascular cells displayed the invasion of vasculature from the host heart to the tissue and their perfusion. Survived ECTs replaced the ventricular wall in the injured area and prevented the scar formation after myocardial infarction and improved cardiac function (Figure 4). ECTs survived during 4-week follow-up period [16, 20]. However, further work is required to identify the underlying mechanism for the functional recovery. Meanwhile, the first transplantation of cardiac progenitor patch derived from human embryonic stem cells in a severe heart failure patient was performed in France, offering an encouraging result [60].

Figure 4.

Therapeutic effects of hiPSC-mesh ECT implantation in a rat myocardial infarction model. (a) Schematic timeline of surgery. A mesh ECT matured in vitro for 14 days (or sham suture) is implanted in a nude rat (week 0) 1 week after the induction of myocardial infarction by ligating left anterior descending artery (LAD). Echocardiogram (Echo) is performed prior to LAD ligation at week-1 (W-1), prior to implantation at week 0 (W0), then week 2 (W2) and week 4 (W4). (b) Grafted mesh ECT on the heart surface covering infarction site. (c) Representative Masson’s trichrome staining images of sham treated (left) and mesh ECT implanted (right) rat hearts at W4. Scale bar: 2 mm. Red dotted line indicates engrafted area. (d) Comparison of scar area (% of LV area) at W4 (n=5, *P<0.05 Implant versus Sham). (e-g) Results of echocardiogram [n=5 (Implant, red solid line) and 5 (Sham, blue dotted line)]. (e) Left ventricular end diastolic area (LVAd; mm2), (f) ejection fraction, EF (%), and (g) cardiac index, CI (mL/min/kg) (*P<0.05 Implant versus Sham at W4). (Reproduced from Ref. [20] with permission).

Advertisement

5. Conclusion

In this chapter, we have reviewed several aspects of current cardiac tissue engineering technologies and presented the possible applications of these tissues for in vitro drug toxicity testing, human disease modeling, and paradigms for myocardial recovery with muscle replacement following injury. This rapidly evolving new field is now incorporating manufacturing process to expand the scalability and reduce the cost of generating these novel engineered in vitro myocardial tissues.

Advertisement

Conflict of interest

None.

References

  1. 1. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663-676
  2. 2. Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861-872
  3. 3. Laflamme MA, Chen KY, Naumova AV, et al. Cardiomyocytes derived from human embryonic stem cells in pro-survival factors enhance function of infarcted rat hearts. Nature Biotechnology. 2007;25:1015-1024
  4. 4. Kattman SJ, Witty AD, Gagliardi M, et al. Stage-specific optimization of activin/nodal and BMP signaling promotes cardiac differentiation of mouse and human pluripotent stem cell lines. Cell Stem Cell. 2011;8:228-240
  5. 5. Uosaki H, Fukushima H, Takeuchi A, et al. Efficient and scalable purification of cardiomyocytes from human embryonic and induced pluripotent stem cells by VCAM1 surface expression. PLoS One. 2011;6:e23657
  6. 6. Masumoto H, Ikuno T, Takeda M, et al. Human iPS cell-engineered cardiac tissue sheets with cardiomyocytes and vascular cells for cardiac regeneration. Scientific Reports. 2014;4:6716
  7. 7. Shimizu T, Yamato M, Isoi Y, et al. Fabrication of pulsatile cardiac tissue grafts using a novel 3-dimensional cell sheet manipulation technique and temperature-responsive cell culture surfaces. Circulation Research. 2002;90:40-48
  8. 8. Chang D, Shimizu T, Haraguchi Y, et al. Time course of cell sheet adhesion to porcine heart tissue after transplantation. PLoS One. 2015;10:1-14
  9. 9. Shimizu T, Sekine H, Yang J, et al. Polysurgery of cell sheet grafts overcomes diffusion limits to produce thick, vascularized myocardial tissues. The FASEB Journal. 2006;20:1-20
  10. 10. Matsuo T, Masumoto H, Tajima S, et al. Efficient long-term survival of cell grafts after myocardial infarction with thick viable cardiac tissue entirely from pluripotent stem cells. Scientific Reports. 2015;5:16842
  11. 11. Eschenhagen T, Fink C, Remmers U, et al. Three-dimensional reconstitution of embryonic cardiomyocytes in a collagen matrix: A new heart muscle model system. The FASEB Journal. 1997;11:683-694
  12. 12. Tobita K, Liu LJ, Janczewski AM, et al. Engineered early embryonic cardiac tissue retains proliferative and contractile properties of developing embryonic myocardium. American Journal of Physiology. Heart and Circulatory Physiology. 2006;291:H1829-H1837
  13. 13. Tulloch NL, Muskheli V, Razumova MV, et al. Growth of engineered human myocardium with mechanical loading and vascular coculture. Circulation Research. 2011;109:47-59
  14. 14. Schaaf S, Shibamiya A, Mewe M, et al. Human engineered heart tissue as a versatile tool in basic research and preclinical toxicology. PLoS One. 2011;6(10):e26397. DOI: 10.1371/journal.pone.0026397
  15. 15. Kensah G, Lara AR, Dahlmann J, et al. Murine and human pluripotent stem cell-derived cardiac bodies form contractile myocardial tissue in vitro. European Heart Journal. 2013;34:1134-1146
  16. 16. Masumoto H, Nakane T, Tinney JP, et al. The myocardial regenerative potential of three-dimensional engineered cardiac tissues composed of multiple human iPS cell-derived cardiovascular cell lineages. Scientific Reports. 2016;6:29933
  17. 17. Zimmermann W-H, Melnychenko I, Wasmeier G, et al. Engineered heart tissue grafts improve systolic and diastolic function in infarcted rat hearts. Nature Medicine. 2006;12:452-458
  18. 18. Bian W, Liau B, Badie N, et al. Mesoscopic hydrogel molding to control the 3D geometry of bioartificial muscle tissues. Nature Protocols. 2009;4:1522-1534
  19. 19. Tiburcy M, Hudson JE, Balfanz P, et al. Defined engineered human myocardium with advanced maturation for applications in heart failure modeling and repair clinical perspective. Circulation. 2017;135:1832-1847
  20. 20. Nakane T, Masumoto H, Tinney JP, et al. Impact of cell composition and geometry on human induced pluripotent stem cells-derived engineered cardiac tissue. Scientific Reports. 2017;7:45641
  21. 21. Sidorov VY, Samson PC, Sidorova TN, et al. I-Wire Heart-on-a-Chip I: Three-dimensional cardiac tissue constructs for physiology and pharmacology. Acta Biomaterialia. 2017;48:68-78
  22. 22. Schroer AK, Shotwell MS, Sidorov VY, et al. I-Wire Heart-on-a-Chip II: Biomechanical analysis of contractile, three-dimensional cardiomyocyte tissue constructs. Acta Bio-materialia. 2017;48:79-87
  23. 23. Gaetani R, Feyen DAM, Verhage V, et al. Epicardial application of cardiac progenitor cells in a 3D-printed gelatin/hyaluronic acid patch preserves cardiac function after myocardial infarction. Biomaterials. 2015;61:339-348
  24. 24. Asthana A, Kisaalita WS. Biophysical microenvironment and 3D culture physiological relevance. Drug Discovery Today. 2013;18:533-540
  25. 25. Hirt MN, Hansen A, Eschenhagen T. Cardiac tissue engineering: State of the art. Circulation Research. 2014;114:354-367
  26. 26. Young JL, Engler AJ. Hydrogles with time-dependent material properties enhance cardiomyocyte differentiation in-vitro. Biomaterials. 2011;32:1002-1009
  27. 27. Ott HC, Matthiesen TS, Goh S-K, et al. Perfusion-decellularized matrix: using nature’s platform to engineer a bioartificial heart. Nature Medicine. 2008;14:213-221
  28. 28. Guyette JP, Charest J, Mills RW, et al. Bioengineering human myocardium on native extracellular matrix. Circulation Research. 2015 CIRCRESAHA.115.306874
  29. 29. Blazeski A, Kostecki GM, Tung L. Engineered heart slices for electrophysiological and contractile studies. Biomaterials. 2015;55:119-128
  30. 30. Jugdutt BI. Ventricular remodeling after infarction and the extracellular collagen matrix: When is enough enough? Circulation. 2003;108:1395-1403
  31. 31. Quinn TA, Camelliti P, Rog-Zielinska EA, et al. Electrotonic coupling of excitable and nonexcitable cells in the heart revealed by optogenetics. Proceedings of the National Academy of Sciences. 2016;113:14852-14857
  32. 32. Zeng Q, Guo Y, Liu L, et al. Cardiac fibroblast-derived extracellular matrix produced in vitro stimulates growth and metabolism of cultured ventricular cells. International Heart Journal. 2013;54:40-44
  33. 33. Uosaki H, Cahan P, Lee DI, et al. Transcriptional landscape of cardiomyocyte maturation. Cell Reports. 2015;13:1705-1716
  34. 34. Pecha S, Eschenhagen T, Reichenspurner H. Myocardial tissue engineering for cardiac repair. The Journal of Heart and Lung Transplantation. 2016;35:294-298
  35. 35. Radisic M, Yang L, Boublik J, et al. Medium perfusion enables engineering of compact and contractile cardiac tissue. American Journal of Physiology. Heart and Circulatory Physiology. 2004;286:H507-H516
  36. 36. Carrier RL, Rupnick M, Langer R, et al. Effects of oxygen on engineered cardiac muscle. Biotechnology and Bioengineering. 2002;78:617-625
  37. 37. Carrier RL, Rupnick M, Langer R, et al. Perfusion improves tissue architecture of engineered cardiac muscle. Tissue Engineering. 2002;8:175-188
  38. 38. Jackman CP, Carlson AL, Bursac N. Dynamic culture yields engineered myocardium with near-adult functional output. Biomaterials. 2016;111:66-79
  39. 39. Ye F, Yuan F, Li X, et al. Gene expression profiles in engineered cardiac tissues respond to mechanical loading and inhibition of tyrosine kinases. Physiological Reports. 2013;1:e00078
  40. 40. Korolj A, Wang EY, Civitarese RA, et al. Biophysical stimulation for in vitro engineering of functional cardiac tissues. Clinical Science. 2017;131http://www.clinsci.org/content/131/13/1393 [Accessed 5 September 2017]
  41. 41. Nunes SS, Miklas JW, Liu J, et al. Biowire: A platform for maturation of human pluripotent stem cell-derived cardiomyocytes. Nature Methods. 2013;10:781-787
  42. 42. Morgan KY, Black LD. Mimicking isovolumic contraction with combined electromechanical stimulation improves the development of engineered cardiac constructs. Tissue Engineering. Part A. 2014;20:1654-1667
  43. 43. Ferri N, Siegl P, Corsini A, et al. Drug attrition during pre-clinical and clinical development: Understanding and managing drug-induced cardiotoxicity. Pharmacology & Therapeutics. 2013;138:470-484
  44. 44. Li X, Zhang R, Zhao B, et al. Cardiotoxicity screening: A review of rapid-throughput in vitro approaches. Archives of Toxicology. 2016;90:1803-1816
  45. 45. Gintant G. An evaluation of hERG current assay performance: Translating preclinical safety studies to clinical QT prolongation. Pharmacology & Therapeutics. 2011;129:109-119
  46. 46. Navarrete EG, Liang P, Lan F, et al. Screening drug-induced arrhythmia events using human induced pluripotent stem cell-derived cardiomyocytes and low-impedance microelectrode arrays. Circulation. 2013;128:S3-13. DOI: 10.1161/CIRCULATIONAHA.112.000570
  47. 47. Kawatou M, Masumoto H, Fukushima H, et al. Modelling Torsade de Pointes arrhythmias in vitro in 3D human iPS cell-engineered heart tissue. Nature Communications. 2017;8:1078
  48. 48. Hansen A, Eder A, Bönstrup M, et al. Development of a drug screening platform based on engineered heart tissue. Circulation Research. 2010;107:35-44
  49. 49. Grosberg A, Alford PW, McCain ML, et al. Ensembles of engineered cardiac tissues for physiological and pharmacological study: Heart on a chip. Lab on a Chip. 2011;11:4165-4173
  50. 50. Tzatzalos E, Abilez OJ, Shukla P, et al. Engineered heart tissues and induced pluripotent stem cells: Macro- and microstructures for disease modeling, drug screening, and translational studies. Advanced Drug Delivery Reviews. 2015:1-11
  51. 51. Feric NT, Radisic M. Towards adult-like human engineered cardiac tissue: Maturing human pluripotent stem cell-derived cardiomyocytes in human engineered cardiac tissues. Advanced drug delivery reviews. 2016;96:110-134. DOI: 10.1016/j.addr.2015.04.019
  52. 52. Mannhardt I, Breckwoldt K, Letuffe-Brenière D, et al. Human engineered heart tissue: Analysis of contractile force. Stem Cell Reports. 2016;7:29-42
  53. 53. Cashman TJ, Josowitz R, Johnson BV, et al. Human engineered cardiac tissues created using induced pluripotent stem cells reveal functional characteristics of BRAF-mediated hypertrophic cardiomyopathy. PLoS One. 2016;11:1-17
  54. 54. Yamashita JK. ES and iPS cell research for cardiovascular regeneration. Experimental Cell Research. 2010;316:2555-2559
  55. 55. Masumoto H, Sakata R. Cardiovascular surgery for realization of regenerative medicine. General Thoracic and Cardiovascular Surgery. 2012;60:744-755
  56. 56. Masumoto H, Yamashita JK. Pluripotent Stem Cells for Cardiac Cell Therapy: The Application of Cell Sheet Technology, Pluripotent Stem Cells, Dr. Deepa Bhartiya (Ed.), InTech; 2013. DOI: 10.5772/56326. Available from: https://www.intechopen.com/books/pluripotent-stem-cells/pluripotent-stem-cells-for-cardiac-cell-therapy-the-application-of-cell-sheet-technology
  57. 57. Lalit PA, Hei DJ, Raval AN, et al. Induced pluripotent stem cells for post-myocardial infarction repair: Remarkable opportunities and challenges. Circulation Research. 2014;114:1328-1345
  58. 58. Kawamura M, Miyagawa S, Miki K, et al. Feasibility, safety, and therapeutic efficacy of human induced pluripotent stem cell-derived cardiomyocyte sheets in a porcine ischemic cardiomyopathy model. Circulation. 2012;126:29-37
  59. 59. Weinberger F, Breckwoldt K, Pecha S, et al. Cardiac repair in guinea pigs with human engineered heart tissue from induced pluripotent stem cells. Science Translational Medicine. 2016;8:363ra148-363ra148
  60. 60. Menasché P, Vanneaux V, Hagège A, et al. Human embryonic stem cell-derived cardiac progenitors for severe heart failure treatment: First clinical case report: Figure 1. European Heart Journal. 2015;36:2011-2017

Written By

Takeichiro Nakane, Hidetoshi Masumoto and Bradley B. Keller

Submitted: 05 September 2017 Reviewed: 11 October 2017 Published: 20 December 2017