Progress in Bioengineering Strategies for Heart Regenerative Medicine
Abstract
:1. Introduction
2. Stem Cell Engineering and Cardiomyocyte Maturation
2.1. Cell Sources for Cardiac Bioengineering
2.2. Cardiomyocyte Maturation Strategies
3. Development of Functional Biomaterials for Cardiac Tissue Engineering
3.1. Hydrogels
3.2. Decellularized Bioscaffolds
4. Implementation of Biofabrication Tools for Cardiac Tissue Engineering
4.1. Microfabrication Technologies for Generating Engineered Heart Tissues
4.2. Bioprinting
5. Clinical Applications of Cardiac Tissue Engineering-Based Technologies
5.1. Drug Screening and Disease Modeling
5.2. Heart Regenerative Therapy
6. Future Perspectives of Cardiac Tissue Engineering
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Roth, G.A.; Johnson, C.; Abajobir, A.; Abd-Allah, F.; Abera, S.F.; Abyu, G.; Ahmed, M.; Aksut, B.; Alam, T.; Alam, K.; et al. Global, regional, and national burden of cardiovascular diseases for 10 causes, 1990 to 2015. J. Am. Coll. Cardiol. 2017, 70, 1–25. [Google Scholar] [CrossRef] [PubMed]
- Dai, H.; Zhang, Q.; Much, A.A.; Maor, E.; Segev, A.; Beinart, R.; Adawi, S.; Lu, Y.; Bragazzi, N.L.; Wu, J. Global, regional, and national prevalence, incidence, mortality, and risk factors for atrial fibrillation, 1990–2017: Results from the Global Burden of Disease Study 2017. Eur. Heart J. Qual. Care Clin. Outcomes 2021, 7, 574–582. [Google Scholar] [CrossRef] [PubMed]
- Bergmann, O.; Zdunek, S.; Felker, A.; Salehpour, M.; Alkass, K.; Bernard, S.; Sjostrom, S.L.; Szewczykowska, M.; Jackowska, T.; Dos Remedios, C.; et al. Dynamics of cell generation and turnover in the human heart. Cell 2015, 161, 1566–1575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tajabadi, M.; Goran Orimi, H.; Ramzgouyan, M.R.; Nemati, A.; Deravi, N.; Beheshtizadeh, N.; Azami, M. Regenerative strategies for the consequences of myocardial infarction: Chronological indication and upcoming visions. Biomed. Pharmacother. 2022, 146, 112584. [Google Scholar] [CrossRef] [PubMed]
- Witman, N.; Zhou, C.; Beverborg, N.G.; Sahara, M.; Chien, K.R. Cardiac progenitors and paracrine mediators in cardiogenesis and heart regeneration. Semin. Cell Dev. Biol. 2020, 100, 29–51. [Google Scholar] [CrossRef] [PubMed]
- Florian, W.; Ingra, M.; Thomas, E. Engineering cardiac muscle tissue: A maturating field of research. Circ. Res. 2017, 120, 1487–1500. [Google Scholar]
- Sheikh, A.Y.; Lin, S.A.; Cao, F.; Cao, Y.; van der Bogt, K.E.; Chu, P.; Chang, C.P.; Contag, C.H.; Robbins, R.C.; Wu, J.C. Molecular imaging of bone marrow mononuclear cell homing and engraftment in ischemic myocardium. Stem Cells 2007, 25, 2677–2684. [Google Scholar] [CrossRef] [Green Version]
- van den Akker, F.; Feyen, D.A.; van den Hoogen, P.; van Laake, L.W.; van Eeuwijk, E.C.; Hoefer, I.; Pasterkamp, G.; Chamuleau, S.A.; Grundeman, P.F.; Doevendans, P.A.; et al. Intramyocardial stem cell injection: Go(ne) with the flow. Eur. Heart J. 2017, 38, 184–186. [Google Scholar] [CrossRef] [Green Version]
- Madonna, R.; Van Laake, L.W.; Davidson, S.M.; Engel, F.B.; Hausenloy, D.J.; Lecour, S.; Leor, J.; Perrino, C.; Schulz, R.; Ytrehus, K.; 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. 2016, 37, 1789–1798. [Google Scholar] [CrossRef]
- Gaetani, R.; Rizzitelli, G.; Chimenti, I.; Barile, L.; Forte, E.; Ionta, V.; Angelini, F.; Sluijter, J.P.; Barbetta, A.; Messina, E.; et al. Cardiospheres and tissue engineering for myocardial regeneration: Potential for clinical application. J. Cell Mol. Med. 2010, 14, 1071–1077. [Google Scholar] [CrossRef] [Green Version]
- Kwon, S.G.; Kwon, Y.W.; Lee, T.W.; Park, G.T.; Kim, J.H. Recent advances in stem cell therapeutics and tissue engineering strategies. Biomater. Res. 2018, 22, 36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Berry, J.L.; Zhu, W.; Tang, Y.L.; Krishnamurthy, P.; Ge, Y.; Cooke, J.P.; Chen, Y.; Garry, D.J.; Yang, H.T.; Rajasekaran, N.S.; et al. Convergences of life sciences and engineering in understanding and treating heart failure. Circ. Res. 2019, 124, 161–169. [Google Scholar] [CrossRef] [PubMed]
- Thomson, J.A.; Itskovitz-Eldor, J.; Shapiro, S.S.; Waknitz, M.A.; Swiergiel, J.J.; Marshall, V.S.; Jones, J.M. Embryonic stem cell lines derived from human blastocysts. Science 1998, 282, 1145–1147. [Google Scholar] [CrossRef] [Green Version]
- Takahashi, K.; Tanabe, K.; Ohnuki, M.; Narita, M.; Ichisaka, T.; Tomoda, K.; Yamanaka, S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007, 131, 861–872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Burridge, P.W.; Matsa, E.; Shukla, P.; Lin, Z.C.; Churko, J.M.; Ebert, A.D.; Lan, F.; Diecke, S.; Huber, B.; Mordwinkin, N.M.; et al. Chemically defined generation of human cardiomyocytes. Nat. Methods 2014, 11, 855–860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lian, X.; Zhang, J.; Azarin, S.M.; Zhu, K.; Hazeltine, L.B.; Bao, X.; Hsiao, C.; Kamp, T.J.; Palecek, S.P. Directed cardiomyocyte differentiation from human pluripotent stem cells by modulating Wnt/β-catenin signaling under fully defined conditions. Nat. Protoc. 2013, 8, 162–175. [Google Scholar] [CrossRef] [Green Version]
- James, D.; Nam, H.S.; Seandel, M.; Nolan, D.; Janovitz, T.; Tomishima, M.; Studer, L.; Lee, G.; Lyden, D.; Benezra, R.; et al. Expansion and maintenance of human embryonic stem cell-derived endothelial cells by TGFbeta inhibition is Id1 dependent. Nat. Biotechnol. 2010, 28, 161–166. [Google Scholar] [CrossRef]
- Sahara, M.; Hansson, E.M.; Wernet, O.; Lui, K.O.; Später, D.; Chien, K.R. Manipulation of a VEGF-Notch signaling circuit drives formation of functional vascular endothelial progenitors from human pluripotent stem cells. Cell Res. 2014, 24, 820–841. [Google Scholar] [CrossRef] [Green Version]
- Patsch, C.; Challet-Meylan, L.; Thoma, E.C.; Urich, E.; Heckel, T.; O’Sullivan, J.F.; Grainger, S.J.; Kapp, F.G.; Sun, L.; Christensen, K.; et al. Generation of vascular endothelial and smooth muscle cells from human pluripotent stem cells. Nat. Cell Biol. 2015, 17, 994–1003. [Google Scholar] [CrossRef]
- Gong, J.; Zhou, D.; Jiang, L.; Qiu, P.; Milewicz, D.M.; Chen, Y.E.; Yang, B. In vitro lineage-specific differentiation of vascular smooth muscle cells in response to SMAD3 deficiency: Implications for SMAD3-related thoracic aortic aneurysm. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 1651–1663. [Google Scholar] [CrossRef]
- Chen, I.Y.; Matsa, E.; Wu, J.C. Induced pluripotent stem cells: At the heart of cardiovascular precision medicine. Nat. Rev. Cardiol. 2016, 13, 333–349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sahara, M.; Santoro, F.; Chien, K.R. Programming and reprogramming a human heart cell. EMBO J. 2015, 34, 710–738. [Google Scholar] [CrossRef] [PubMed]
- Tenreiro, M.F.; Louro, A.F.; Alves, P.M.; Serra, M. Next generation of heart regenerative therapies: Progress and promise of cardiac tissue engineering. NPJ Regen. Med. 2021, 6, 30. [Google Scholar] [CrossRef] [PubMed]
- Palpant, N.J.; Pabon, L.; Friedman, C.E.; Roberts, M.; Hadland, B.; Zaunbrecher, R.J.; Bernstein, I.; Zheng, Y.; Murry, C.E. Generating high-purity cardiac and endothelial derivatives from patterned mesoderm using human pluripotent stem cells. Nat. Protoc. 2017, 12, 15–31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, V.C.; Ye, J.; Shukla, P.; Hua, G.; Chen, D.; Lin, Z.; Liu, J.C.; Chai, J.; Gold, J.; Wu, J.; et al. Development of a scalable suspension culture for cardiac differentiation from human pluripotent stem cells. Stem Cell Res. 2015, 15, 365–375. [Google Scholar] [CrossRef] [PubMed]
- Santoro, F.; Chien, K.R.; Sahara, M. Isolation of human ESC-derived cardiac derivatives and embryonic heart cells for population and single-cell RNA-seq analysis. STAR Protoc. 2021, 2, 100339. [Google Scholar] [CrossRef]
- Yang, X.; Pabon, L.; Murry, C.E. Engineering adolescence: Maturation of human pluripotent stem cell-derived cardiomyocytes. Circ. Res. 2014, 114, 511–523. [Google Scholar] [CrossRef] [Green Version]
- Veerman, C.C.; Kosmidis, G.; Mummery, C.L.; Casini, S.; Verkerk, A.O.; Bellin, M. Immaturity of human stem-cell-derived cardiomyocytes in culture: Fatal flaw or soluble problem? Stem Cells Dev. 2015, 24, 1035–1052. [Google Scholar] [CrossRef] [Green Version]
- Zuppinger, C.; Gibbons, G.; Dutta-Passecker, P.; Segiser, A.; Most, H.; Suter, T.M. Characterization of cytoskeleton features and maturation status of cultured human iPSC-derived cardiomyocytes. Eur. J. Histochem. 2017, 61, 2763. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.W.; Chen, B.; Yang, X.; Fugate, J.A.; Kalucki, F.A.; Futakuchi-Tsuchida, A.; Couture, L.; Vogel, K.W.; Astley, C.A.; Baldessari, A.; et al. Human embryonic stem cell-derived cardiomyocytes restore function in infarcted hearts of non-human primates. Nat. Biotechnol. 2018, 36, 597–605. [Google Scholar] [CrossRef]
- Romagnuolo, R.; Masoudpour, H.; Porta-Sánchez, A.; Qiang, B.; Barry, J.; Laskary, A.; Qi, X.; Massé, S.; Magtibay, K.; Kawajiri, H.; et al. Human embryonic stem cell-derived cardiomyocytes regenerate the infarcted pig heart but induce ventricular tachyarrhythmias. Stem Cell Rep. 2019, 12, 967–981. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Karbassi, E.; Fenix, A.; Marchiano, S.; Muraoka, N.; Nakamura, K.; Yang, X.; Murry, C.E. Cardiomyocyte maturation: Advances in knowledge and implications for regenerative medicine. Nat. Rev. Cardiol. 2020, 17, 341–359. [Google Scholar] [CrossRef] [PubMed]
- 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 1999, 100, 193–202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- de Soysa, T.Y.; Ranade, S.S.; Okawa, S.; Ravichandran, S.; Huang, Y.; Salunga, H.T.; Schricker, A.; Del Sol, A.; Gifford, C.A.; Srivastava, D. Single-cell analysis of cardiogenesis reveals basis for organ-level developmental defects. Nature 2019, 572, 120–124. [Google Scholar] [CrossRef]
- Cui, Y.; Zheng, Y.; Liu, X.; Yan, L.; Fan, X.; Yong, J.; Hu, Y.; Dong, J.; Li, Q.; Wu, X.; et al. Single-cell transcriptome analysis maps the developmental track of the human heart. Cell Rep. 2019, 26, 1934–1950.e5. [Google Scholar] [CrossRef] [Green Version]
- Litviňuková, M.; Talavera-López, C.; Maatz, H.; Reichart, D.; Worth, C.L.; Lindberg, E.L.; Kanda, M.; Polanski, K.; Heinig, M.; Lee, M.; et al. Cells of the adult human heart. Nature 2020, 588, 466–472. [Google Scholar] [CrossRef]
- Sahara, M.; Santoro, F.; Sohlmér, J.; Zhou, C.; Witman, N.; Leung, C.Y.; Mononen, M.; Bylund, K.; Gruber, P.; Chien, K.R. Population and single-cell analysis of human cardiogenesis reveals unique LGR5 ventricular progenitors in embryonic outflow tract. Dev. Cell 2019, 48, 475–490. [Google Scholar] [CrossRef] [Green Version]
- Feric, N.T.; Radisic, M. Maturing human pluripotent stem cell-derived cardiomyocytes in human engineered cardiac tissues. Adv. Drug Deliv. Rev. 2016, 96, 110–134. [Google Scholar] [CrossRef] [Green Version]
- Lundy, S.D.; Zhu, W.Z.; Regnier, M.; Laflamme, M.A. Structural and functional maturation of cardiomyocytes derived from human pluripotent stem cells. Stem Cells Dev. 2013, 22, 1991–2002. [Google Scholar] [CrossRef] [Green Version]
- Zimmermann, W.H.; Schneiderbanger, K.; Schubert, P.; Didié, M.; Münzel, F.; Heubach, J.F.; Kostin, S.; Neuhuber, W.L.; Eschenhagen, T. Tissue engineering of a differentiated cardiac muscle construct. Circ. Res. 2002, 90, 223–230. [Google Scholar] [CrossRef] [Green Version]
- Hansen, A.; Eder, A.; Bönstrup, M.; Flato, M.; Mewe, M.; Schaaf, S.; Aksehirlioglu, B.; Schwoerer, A.P.; Uebeler, J.; Eschenhagen, T. Development of a drug screening platform based on engineered heart tissue. Circ. Res. 2010, 107, 35–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ronaldson-Bouchard, K.; Ma, S.P.; Yeager, K.; Chen, T.; Song, L.; Sirabella, D.; Morikawa, K.; Teles, D.; Yazawa, M.; Vunjak-Novakovic, G. Advanced maturation of human cardiac tissue grown from pluripotent stem cells. Nature 2018, 556, 239–243. [Google Scholar] [CrossRef] [PubMed]
- Jackman, C.P.; Carlson, A.L.; Bursac, N. Dynamic culture yields engineered myocardium with near-adult functional output. Biomaterials 2016, 111, 66–79. [Google Scholar] [CrossRef] [Green Version]
- Kroll, K.; Chabria, M.; Wang, K.; Häusermann, F.; Schuler, F.; Polonchuk, L. Electro-mechanical conditioning of human iPSC-derived cardiomyocytes for translational research. Prog. Biophys. Mol. Biol. 2017, 130, 212–222. [Google Scholar] [CrossRef] [PubMed]
- Nunes, S.S.; Miklas, J.W.; Liu, J.; Aschar-Sobbi, R.; Xiao, Y.; Zhang, B.; Jiang, J.; Massé, S.; Gagliardi, M.; Hsieh, A.; et al. Biowire: A platform for maturation of human pluripotent stem cell-derived cardiomyocytes. Nat. Methods 2013, 10, 781–787. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, D.; Shadrin, I.Y.; Lam, J.; Xian, H.Q.; Snodgrass, H.R.; Bursac, N. Tissue-engineered cardiac patch for advanced functional maturation of human ESC-derived cardiomyocytes. Biomaterials 2013, 34, 5813–5820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hirt, M.N.; Hansen, A.; Eschenhagen, T. Cardiac tissue engineering: State of the art. Circ. Res. 2014, 114, 354–367. [Google Scholar] [CrossRef] [Green Version]
- Tiburcy, M.; Hudson, J.E.; Balfanz, P.; Schlick, S.; Meyer, T.; Chang Liao, M.L.; Levent, E.; Raad, F.; Zeidler, S.; Wingender, E.; et al. Defined Engineered human myocardium with advanced maturation for applications in heart failure modeling and repair. Circulation 2017, 135, 1832–1847. [Google Scholar] [CrossRef]
- Ulmer, B.M.; Stoehr, A.; Schulze, M.L.; Patel, S.; Gucek, M.; Mannhardt, I.; Funcke, S.; Murphy, E.; Eschenhagen, T.; Hansen, A. Contractile work contributes to maturation of energy metabolism in hiPSC-derived cardiomyocytes. Stem Cell Rep. 2018, 10, 834–847. [Google Scholar] [CrossRef] [Green Version]
- Frey, O.; Misun, P.M.; Fluri, D.A.; Hengstler, J.G.; Hierlemann, A. Reconfigurable microfluidic hanging drop network for multi-tissue interaction and analysis. Nat. Commun. 2014, 5, 4250. [Google Scholar] [CrossRef] [Green Version]
- Correia, C.; Koshkin, A.; Duarte, P.; Hu, D.; Carido, M.; Sebastião, M.J.; Gomes-Alves, P.; Elliott, D.A.; Domian, I.J.; Teixeira, A.P.; et al. 3D aggregate culture improves metabolic maturation of human pluripotent stem cell derived cardiomyocytes. Biotechnol. Bioeng. 2018, 115, 630–644. [Google Scholar] [CrossRef] [PubMed]
- Abecasis, B.; Gomes-Alves, P.; Rosa, S.; Gouveia, P.J.; Ferreira, L.; Serra, M.; Alves, P.M. Unveiling the molecular crosstalk in a human induced pluripotent stem cell-derived cardiac model. Biotechnol. Bioeng. 2019, 116, 1245–1252. [Google Scholar] [CrossRef] [PubMed]
- Dunn, K.K.; Reichardt, I.M.; Simmons, A.D.; Jin, G.; Floy, M.E.; Hoon, K.M.; Palecek, S.P. Coculture of endothelial cells with human pluripotent stem cell-derived cardiac progenitors reveals a differentiation stage-specific enhancement of cardiomyocyte maturation. Biotechnol. J. 2019, 14, e1800725. [Google Scholar] [CrossRef]
- Bargehr, J.; Ong, L.P.; Colzani, M.; Davaapil, H.; Hofsteen, P.; Bhandari, S.; Gambardella, L.; Le Novère, N.; Iyer, D.; Sampaziotis, F.; et al. Epicardial cells derived from human embryonic stem cells augment cardiomyocyte-driven heart regeneration. Nat. Biotechnol. 2019, 37, 895–906. [Google Scholar] [CrossRef] [PubMed]
- Brutsaert, D.L. Cardiac endothelial-myocardial signaling: Its role in cardiac growth, contractile performance, and rhythmicity. Physiol. Rev. 2003, 83, 59–115. [Google Scholar] [CrossRef] [PubMed]
- Tirziu, D.; Giordano, F.J.; Simons, M. Cell communications in the heart. Circulation 2010, 122, 928–937. [Google Scholar] [CrossRef] [Green Version]
- Pinto, A.R.; Ilinykh, A.; Ivey, M.J.; Kuwabara, J.T.; D’Antoni, M.L.; Debuque, R.; Chandran, A.; Wang, L.; Arora, K.; Rosenthal, N.A.; et al. Revisiting cardiac cellular composition. Circ. Res. 2016, 118, 400–409. [Google Scholar] [CrossRef] [Green Version]
- Fountoulaki, K.; Dagres, N.; Iliodromitis, E.K. Cellular communications in the heart. Card. Fail. Rev. 2015, 1, 64–68. [Google Scholar] [CrossRef]
- Hu, D.; Linders, A.; Yamak, A.; Correia, C.; Kijlstra, J.D.; Garakani, A.; Xiao, L.; Milan, D.J.; van der Meer, P.; Serra, M.; et al. Metabolic maturation of human pluripotent stem cell-derived cardiomyocytes by inhibition of HIF1α and LDHA. Circ. Res. 2018, 123, 1066–1079. [Google Scholar] [CrossRef] [Green Version]
- Yang, X.; Rodriguez, M.L.; Leonard, A.; Sun, L.; Fischer, K.A.; Wang, Y.; Ritterhoff, J.; Zhao, L.; Kolwicz, S.C., Jr.; Pabon, L.; et al. Fatty acids enhance the maturation of cardiomyocytes derived from human pluripotent stem cells. Stem Cell Rep. 2019, 13, 657–668. [Google Scholar] [CrossRef] [Green Version]
- Yang, X.; Rodriguez, M.; Pabon, L.; Fischer, K.A.; Reinecke, H.; Regnier, M.; Sniadecki, N.J.; Ruohola-Baker, H.; Murry, C.E. Tri-iodo-l-thyronine promotes the maturation of human cardiomyocytes-derived from induced pluripotent stem cells. J. Mol. Cell Cardiol. 2014, 72, 296–304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, M.; Iismaa, S.E.; Naqvi, N.; Nicks, A.; Husain, A.; Graham, R.M. Thyroid hormone action in postnatal heart development. Stem Cell Res. 2014, 13, 582–591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rog-Zielinska, E.A.; Thomson, A.; Kenyon, C.J.; Brownstein, D.G.; Moran, C.M.; Szumska, D.; Michailidou, Z.; Richardson, J.; Owen, E.; Watt, A.; et al. Glucocorticoid receptor is required for foetal heart maturation. Hum. Mol. Genet. 2013, 22, 3269–3282. [Google Scholar] [CrossRef] [Green Version]
- Ferrini, A.; Stevens, M.M.; Sattler, S.; Rosenthal, N. Toward regeneration of the heart: Bioengineering strategies for immunomodulation. Front. Cardiovasc. Med. 2019, 6, 26. [Google Scholar] [CrossRef] [Green Version]
- Gomez-Garcia, M.J.; Quesnel, E.; Al-Attar, R.; Laskary, A.R.; Laflamme, M.A. Maturation of human pluripotent stem cell derived cardiomyocytes in vitro and in vivo. Semin. Cell Dev. Biol. 2021, 118, 163–171. [Google Scholar] [CrossRef]
- Chung, S.; Dzeja, P.P.; Faustino, R.S.; Perez-Terzic, C.; Behfar, A.; Terzic, A. Mitochondrial oxidative metabolism is required for the cardiac differentiation of stem cells. Nat. Clin. Pract. Cardiovasc. Med. 2007, 4, S60–S67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Khanna, A.; Zamani, M.; Huang, N.F. Extracellular matrix-based biomaterials for cardiovascular tissue engineering. J. Cardiovasc. Dev. Dis. 2021, 8, 137. [Google Scholar] [CrossRef]
- Moroni, L.; Burdick, J.A.; Highley, C.; Lee, S.J.; Morimoto, Y.; Takeuchi, S.; Yoo, J.J. Biofabrication strategies for 3D in vitro models and regenerative medicine. Nat. Rev. Mater. 2018, 3, 21–37. [Google Scholar] [CrossRef]
- Tous, E.; Purcell, B.; Ifkovits, J.L.; Burdick, J.A. Injectable acellular hydrogels for cardiac repair. J. Cardiovasc. Transl. Res. 2011, 4, 528–542. [Google Scholar] [CrossRef]
- Hasan, A.; Khattab, A.; Islam, M.A.; Hweij, K.A.; Zeitouny, J.; Waters, R.; Sayegh, M.; Hossain, M.M.; Paul, A. Injectable hydrogels for cardiac tissue repair after myocardial infarction. Adv. Sci. 2015, 2, 1500122. [Google Scholar] [CrossRef]
- Ye, Z.; Zhou, Y.; Cai, H.; Tan, W. Myocardial regeneration: Roles of stem cells and hydrogels. Adv. Drug Deliv. Rev. 2011, 63, 688–697. [Google Scholar] [CrossRef] [PubMed]
- Johnson, T.D.; Christman, K.L. Injectable hydrogel therapies and their delivery strategies for treating myocardial infarction. Expert Opin. Drug Deliv. 2013, 10, 59–72. [Google Scholar] [CrossRef] [PubMed]
- Slaughter, B.V.; Khurshid, S.S.; Fisher, O.Z.; Khademhosseini, A.; Peppas, N.A. Hydrogels in regenerative medicine. Adv. Mater. 2009, 21, 3307–3329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Malda, J.; Visser, J.; Melchels, F.P.; Jüngst, T.; Hennink, W.E.; Dhert, W.J.; Groll, J.; Hutmacher, D.W. 25th anniversary article: Engineering hydrogels for biofabrication. Adv. Mater. 2013, 25, 5011–5028. [Google Scholar] [CrossRef]
- Lee, S.; Serpooshan, V.; Tong, X.; Venkatraman, S.; Lee, M.; Lee, J.; Chirikian, O.; Wu, J.C.; Wu, S.M.; Yang, F. Contractile force generation by 3D hiPSC-derived cardiac tissues is enhanced by rapid establishment of cellular interconnection in matrix with muscle-mimicking stiffness. Biomaterials 2017, 131, 111–120. [Google Scholar] [CrossRef]
- Huebsch, N.; Loskill, P.; Deveshwar, N.; Spencer, C.I.; Judge, L.M.; Mandegar, M.A.; Fox, C.B.; Mohamed, T.M.; Ma, Z.; Mathur, A.; et al. Miniaturized iPS-cell-derived cardiac muscles for physiologically relevant drug response analyses. Sci. Rep. 2016, 6, 24726. [Google Scholar] [CrossRef] [Green Version]
- Mannhardt, I.; Breckwoldt, K.; Letuffe-Brenière, D.; Schaaf, S.; Schulz, H.; Neuber, C.; Benzin, A.; Werner, T.; Eder, A.; Schulze, T.; et al. Human engineered heart tissue: Analysis of contractile force. Stem Cell Rep. 2016, 7, 29–42. [Google Scholar] [CrossRef] [Green Version]
- Xu, X.; Jha, A.K.; Harrington, D.A.; Farach-Carson, M.C.; Jia, X. Hyaluronic acid-based hydrogels: From a natural polysaccharide to complex networks. Soft Matter. 2012, 8, 3280–3294. [Google Scholar] [CrossRef] [Green Version]
- Gao, L.; Kupfer, M.E.; Jung, J.P.; Yang, L.; Zhang, P.; Da Sie, Y.; Tran, Q.; Ajeti, V.; Freeman, B.T.; Fast, V.G.; 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. 2017, 120, 1318–1325. [Google Scholar] [CrossRef] [Green Version]
- Neves, M.I.; Moroni, L.; Barrias, C.C. Modulating alginate hydrogels for improved biological performance as cellular 3D microenvironments. Front. Bioeng. Biotechnol. 2020, 8, 665. [Google Scholar] [CrossRef]
- Riegler, J.; Tiburcy, M.; Ebert, A.; Tzatzalos, E.; Raaz, U.; Abilez, O.J.; Shen, Q.; Kooreman, N.G.; Neofytou, E.; Chen, V.C.; et al. Human engineered heart muscles engraft and survive long term in a rodent myocardial infarction model. Circ. Res. 2015, 117, 720–730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ye, L.; Chang, Y.H.; Xiong, Q.; Zhang, P.; Zhang, L.; Somasundaram, P.; Lepley, M.; Swingen, C.; Su, L.; Wendel, J.S.; et al. Cardiac repair in a porcine model of acute myocardial infarction with human induced pluripotent stem cell-derived cardiovascular cells. Cell Stem Cell 2014, 15, 750–761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Riemenschneider, S.B.; Mattia, D.J.; Wendel, J.S.; Schaefer, J.A.; Ye, L.; Guzman, P.A.; Tranquillo, R.T. Inosculation and perfusion of pre-vascularized tissue patches containing aligned human microvessels after myocardial infarction. Biomaterials 2016, 97, 51–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, Q.; Tian, S.; Zhao, C.; Chen, X.; Lei, I.; Wang, Z.; Ma, P.X. Porous nanofibrous poly(L-lactic acid) scaffolds supporting cardiovascular progenitor cells for cardiac tissue engineering. Acta. Biomater. 2015, 26, 105–114. [Google Scholar] [CrossRef] [Green Version]
- Chow, A.; Stuckey, D.J.; Kidher, E.; Rocco, M.; Jabbour, R.J.; Mansfield, C.A.; Darzi, A.; Harding, S.E.; Stevens, M.M.; Athanasiou, T. Human induced pluripotent stem cell-derived cardiomyocyte encapsulating bioactive hydrogels improve rat heart function post myocardial infarction. Stem Cell Rep. 2017, 9, 1415–1422. [Google Scholar] [CrossRef] [Green Version]
- Song, S.Y.; Kim, H.; Yoo, J.; Kwon, S.P.; Park, B.W.; Kim, J.J.; Ban, K.; Char, K.; Park, H.J.; Kim, B.S. Prevascularized, multiple-layered cell sheets of direct cardiac reprogrammed cells for cardiac repair. Biomater. Sci. 2020, 8, 4508–4520. [Google Scholar] [CrossRef]
- Ren, S.; Jiang, X.; Li, Z.; Wen, Y.; Chen, D.; Li, X.; Zhang, X.; Zhuo, R.; Chu, H. Physical properties of poly(N-isopropylacrylamide) hydrogel promote its effects on cardiac protection after myocardial infarction. J. Int. Med. Res. 2012, 40, 2167–2182. [Google Scholar] [CrossRef]
- Vasile, C.; Pamfil, D.; Stoleru, E.; Baican, M. New developments in medical applications of hybrid hydrogels containing natural polymers. Molecules 2020, 25, 1539. [Google Scholar] [CrossRef] [Green Version]
- Navaei, A.; Truong, D.; Heffernan, J.; Cutts, J.; Brafman, D.; Sirianni, R.W.; Vernon, B.; Nikkhah, M. PNIPAAm-based biohybrid injectable hydrogel for cardiac tissue engineering. Acta Biomater. 2016, 32, 10–23. [Google Scholar] [CrossRef]
- Jiang, X.J.; Wang, T.; Li, X.Y.; Wu, D.Q.; Zheng, Z.B.; Zhang, J.F.; Chen, J.L.; Peng, B.; Jiang, H.; Huang, C.; et al. Injection of a novel synthetic hydrogel preserves left ventricle function after myocardial infarction. J. Biomed. Mater. Res. A 2009, 90, 472–477. [Google Scholar] [CrossRef]
- Lee, L.C.; Wall, S.T.; Klepach, D.; Ge, L.; Zhang, Z.; Lee, R.J.; Hinson, A.; Gorman, J.H., 3rd; Gorman, R.C.; Guccione, J.M. Algisyl-LVR™ with coronary artery bypass grafting reduces left ventricular wall stress and improves function in the failing human heart. Int. J. Cardiol. 2013, 168, 2022–2028. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, L.C.; Zhihong, Z.; Hinson, A.; Guccione, J.M. Reduction in left ventricular wall stress and improvement in function in failing hearts using Algisyl-LVR. J. Vis. Exp. 2013, 74, 50096. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shaikh, F.M.; Callanan, A.; Kavanagh, E.G.; Burke, P.E.; Grace, P.A.; McGloughlin, T.M. Fibrin: A natural biodegradable scaffold in vascular tissue engineering. Cells Tissues Organs 2008, 188, 333–346. [Google Scholar] [CrossRef] [PubMed]
- Christman, K.L.; Vardanian, A.J.; Fang, Q.; Sievers, R.E.; Fok, H.H.; Lee, R.J. Injectable fibrin scaffold improves cell transplant survival, reduces infarct expansion, and induces neovasculature formation in ischemic myocardium. J. Am. Coll. Cardiol. 2004, 44, 654–660. [Google Scholar] [CrossRef] [Green Version]
- Wendel, J.S.; Ye, L.; Tao, R.; Zhang, J.; Zhang, J.; Kamp, T.J.; Tranquillo, R.T. Functional effects of a tissue-engineered cardiac patch from human induced pluripotent stem cell-derived cardiomyocytes in a rat infarct model. Stem Cells Transl. Med. 2015, 4, 1324–1332. [Google Scholar] [CrossRef]
- Christman, K.L.; Fok, H.H.; Sievers, R.E.; Fang, Q.; Lee, R.J. Fibrin glue alone and skeletal myoblasts in a fibrin scaffold preserve cardiac function after myocardial infarction. Tissue Eng. 2004, 10, 403–409. [Google Scholar] [CrossRef]
- Gao, L.; Gregorich, Z.R.; Zhu, W.; Mattapally, S.; Oduk, Y.; Lou, X.; Kannappan, R.; Borovjagin, A.V.; Walcott, G.P.; Pollard, A.E.; et al. Large cardiac muscle patches engineered from human induced-pluripotent stem cell-derived cardiac cells improve recovery from myocardial infarction in Swine. Circulation 2018, 137, 1712–1730. [Google Scholar] [CrossRef]
- Edalat, S.G.; Jang, Y.; Kim, J.; Park, Y. Collagen type I containing hybrid hydrogel enhances cardiomyocyte maturation in a 3D cardiac model. Polymers 2019, 11, 687. [Google Scholar] [CrossRef] [Green Version]
- Qin, X.; Riegler, J.; Tiburcy, M.; Zhao, X.; Chour, T.; Ndoye, B.; Nguyen, M.; Adams, J.; Ameen, M.; Denney, T.S., Jr.; et al. Magnetic resonance imaging of cardiac strain pattern following transplantation of human tissue engineered heart muscles. Circ. Cardiovasc. Imaging 2016, 9, e004731. [Google Scholar] [CrossRef] [Green Version]
- Rowley, J.A.; Madlambayan, G.; Mooney, D.J. Alginate hydrogels as synthetic extracellular matrix materials. Biomaterials 1999, 20, 45–53. [Google Scholar] [CrossRef]
- Burdick, J.A.; Mauck, R.L.; Gerecht, S. To serve and protect: Hydrogels to improve stem cell-based therapies. Cell Stem Cell 2016, 18, 13–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Landa, N.; Miller, L.; Feinberg, M.S.; Holbova, R.; Shachar, M.; Freeman, I.; Cohen, S.; Leor, J. Effect of injectable alginate implant on cardiac remodeling and function after recent and old infarcts in rat. Circulation 2008, 117, 1388–1396. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leor, J.; Tuvia, S.; Guetta, V.; Manczur, F.; Castel, D.; Willenz, U.; Petneházy, O.; Landa, N.; Feinberg, M.S.; Konen, E.; et al. Intracoronary injection of in situ forming alginate hydrogel reverses left ventricular remodeling after myocardial infarction in Swine. J. Am. Coll. Cardiol. 2009, 54, 1014–1023. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, Y.; Rodrigues, J.; Tomás, H. Injectable and biodegradable hydrogels: Gelation, biodegradation and biomedical applications. Chem. Soc. Rev. 2012, 41, 2193–2221. [Google Scholar] [CrossRef] [PubMed]
- Deng, C.; Zhang, P.; Vulesevic, B.; Kuraitis, D.; Li, F.; Yang, A.F.; Griffith, M.; Ruel, M.; Suuronen, E.J. A collagen–chitosan hydrogel for endothelial differentiation and angiogenesis. Tissue Eng. Part A 2010, 16, 3099–3109. [Google Scholar] [CrossRef]
- Henning, R.J.; Khan, A.; Jimenez, E. Chitosan hydrogels significantly limit left ventricular infarction and remodeling and preserve myocardial contractility. J. Surg. Res. 2016, 201, 490–497. [Google Scholar] [CrossRef]
- Wang, T.; Wu, D.Q.; Jiang, X.J.; Zhang, X.Z.; Li, X.Y.; Zhang, J.F.; Zheng, Z.B.; Zhuo, R.; Jiang, H.; Huang, C. Novel thermosensitive hydrogel injection inhibits post-infarct ventricle remodelling. Eur. J. Heart Fail. 2009, 11, 14–19. [Google Scholar] [CrossRef] [Green Version]
- Dobner, S.; Bezuidenhout, D.; Govender, P.; Zilla, P.; Davies, N. A synthetic non-degradable polyethylene glycol hydrogel retards adverse post-infarct left ventricular remodeling. J. Card. Fail. 2009, 15, 629–636. [Google Scholar] [CrossRef]
- Chen, Y.; Wang, J.; Shen, B.; Chan, C.W.; Wang, C.; Zhao, Y.; Chan, H.N.; Tian, Q.; Chen, Y.; Yao, C.; et al. Engineering a freestanding biomimetic cardiac patch using biodegradable poly(lactic-co-glycolic acid) (PLGA) and human embryonic stem cell-derived ventricular cardiomyocytes (hESC-VCMs). Macromol. Biosci. 2015, 15, 426–436. [Google Scholar] [CrossRef]
- Li, J.; Minami, I.; Shiozaki, M.; Yu, L.; Yajima, S.; Miyagawa, S.; Shiba, Y.; Morone, N.; Fukushima, S.; Yoshioka, M.; et al. Human pluripotent stem cell-derived cardiac tissue-like constructs for repairing the infarcted myocardium. Stem Cell Rep. 2017, 9, 1546–1559. [Google Scholar] [CrossRef] [Green Version]
- Xu, X.; Liu, Y.; Fu, W.; Yao, M.; Ding, Z.; Xuan, J.; Li, D.; Wang, S.; Xia, Y.; Cao, M. Poly(N-isopropylacrylamide)-Based thermoresponsive composite hydrogels for biomedical applications. Polymers 2020, 12, 580. [Google Scholar] [CrossRef] [Green Version]
- Fujimoto, K.L.; Ma, Z.; Nelson, D.M.; Hashizume, R.; Guan, J.; Tobita, K.; Wagner, W.R. Synthesis, characterization and therapeutic efficacy of a biodegradable, thermoresponsive hydrogel designed for application in chronic infarcted myocardium. Biomaterials 2009, 30, 4357–4368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rebouças, J.S.; Santos-Magalhães, N.S.; Formiga, F.R. Cardiac regeneration using growth factors: Advances and challenges. Arq. Bras. Cardiol. 2016, 107, 271–275. [Google Scholar] [CrossRef]
- Shachar, M.; Tsur-Gang, O.; Dvir, T.; Leor, J.; Cohen, S. The effect of immobilized RGD peptide in alginate scaffolds on cardiac tissue engineering. Acta. Biomater. 2011, 7, 152–162. [Google Scholar] [CrossRef]
- Sondermeijer, H.P.; Witkowski, P.; Seki, T.; van der Laarse, A.; Itescu, S.; Hardy, M.A. RGDfK-peptide modified alginate scaffold for cell transplantation and cardiac neovascularization. Tissue Eng. Part A 2018, 24, 740–751. [Google Scholar] [CrossRef] [PubMed]
- Boateng, S.Y.; Lateef, S.S.; Mosley, W.; Hartman, T.J.; Hanley, L.; Russell, B. RGD and YIGSR synthetic peptides facilitate cellular adhesion identical to that of laminin and fibronectin but alter the physiology of neonatal cardiac myocytes. Am. J. Physiol. Cell Physiol. 2005, 288, C30–C38. [Google Scholar] [CrossRef] [PubMed]
- Schussler, O.; Coirault, C.; Louis-Tisserand, M.; Al-Chare, W.; Oliviero, P.; Menard, C.; Michelot, R.; Bochet, P.; Salomon, D.R.; Chachques, J.C.; et al. Use of arginine-glycine-aspartic acid adhesion peptides coupled with a new collagen scaffold to engineer a myocardium-like tissue graft. Nat. Clin. Pract. Cardiovasc. Med. 2009, 6, 240–249. [Google Scholar] [CrossRef]
- Kuraitis, D.; Hosoyama, K.; Blackburn, N.J.R.; Deng, C.; Zhong, Z.; Suuronen, E.J. Functionalization of soft materials for cardiac repair and regeneration. Crit. Rev. Biotechnol. 2019, 39, 451–468. [Google Scholar] [CrossRef]
- Lutolf, M.P.; Gilbert, P.M.; Blau, H.M. Designing materials to direct stem-cell fate. Nature 2009, 462, 433–441. [Google Scholar] [CrossRef] [Green Version]
- Chen, W.C.; Lee, B.G.; Park, D.W.; Kim, K.; Chu, H.; Kim, K.; Huard, J.; Wang, Y. Controlled dual delivery of fibroblast growth factor-2 and Interleukin-10 by heparin-based coacervate synergistically enhances ischemic heart repair. Biomaterials 2015, 72, 138–151. [Google Scholar] [CrossRef] [Green Version]
- Turner, N.A.; Porter, K.E. Function and fate of myofibroblasts after myocardial infarction. Fibrogenesis Tissue Repair 2013, 6, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Julier, Z.; Park, A.J.; Briquez, P.S.; Martino, M.M. Promoting tissue regeneration by modulating the immune system. Acta. Biomater. 2017, 53, 13–28. [Google Scholar] [CrossRef] [PubMed]
- Suarez, S.L.; Rane, A.A.; Muñoz, A.; Wright, A.T.; Zhang, S.X.; Braden, R.L.; Almutairi, A.; McCulloch, A.D.; Christman, K.L. Intramyocardial injection of hydrogel with high interstitial spread does not impact action potential propagation. Acta. Biomater. 2015, 26, 13–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhu, Y.; Matsumura, Y.; Wagner, W.R. Ventricular wall biomaterial injection therapy after myocardial infarction: Advances in material design, mechanistic insight and early clinical experiences. Biomaterials 2017, 129, 37–53. [Google Scholar] [CrossRef] [PubMed]
- Das, D.; Noh, I. Overviews of biomimetic medical materials. Adv. Exp. Med. Biol. 2018, 1064, 3–24. [Google Scholar]
- Perea-Gil, I.; Prat-Vidal, C.; Bayes-Genis, A. In vivo experience with natural scaffolds for myocardial infarction: The times they are a-changin. Stem Cell Res. Ther. 2015, 6, 248. [Google Scholar] [CrossRef] [Green Version]
- Ott, H.C.; Matthiesen, T.S.; Goh, S.K.; Black, L.D.; Kren, S.M.; Netoff, T.I.; Taylor, D.A. Perfusion-decellularized matrix: Using nature’s platform to engineer a bioartificial heart. Nat. Med. 2008, 14, 213–221. [Google Scholar] [CrossRef]
- Lee, P.F.; Chau, E.; Cabello, R.; Yeh, A.T.; Sampaio, L.C.; Gobin, A.S.; Taylor, D.A. Inverted orientation improves decellularization of whole porcine hearts. Acta. Biomater. 2017, 49, 181–191. [Google Scholar] [CrossRef]
- Gilbert, T.W.; Sellaro, T.L.; Badylak, S.F. Decellularization of tissues and organs. Biomaterials 2006, 27, 3675–3683. [Google Scholar] [CrossRef]
- Crapo, P.M.; Gilbert, T.W.; Badylak, S.F. An overview of tissue and whole organ decellularization processes. Biomaterials 2011, 32, 3233–3243. [Google Scholar] [CrossRef] [Green Version]
- Singelyn, J.M.; Sundaramurthy, P.; Johnson, T.D.; Schup-Magoffin, P.J.; Hu, D.P.; Faulk, D.M.; Wang, J.; Mayle, K.M.; Bartels, K.; Salvatore, M.; et al. Catheter-deliverable hydrogel derived from decellularized ventricular extracellular matrix increases endogenous cardiomyocytes and preserves cardiac function post-myocardial infarction. J. Am. Coll. Cardiol. 2012, 59, 751–763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Seif-Naraghi, S.B.; Singelyn, J.M.; Salvatore, M.A.; Osborn, K.G.; Wang, J.J.; Sampat, U.; Kwan, O.L.; Strachan, G.M.; Wong, J.; Schup-Magoffin, P.J.; et al. Safety and efficacy of an injectable extracellular matrix hydrogel for treating myocardial infarction. Sci. Transl. Med. 2013, 5, 173ra25. [Google Scholar] [CrossRef] [Green Version]
- D’Amore, A.; Yoshizumi, T.; Luketich, S.K.; Wolf, M.T.; Gu, X.; Cammarata, M.; Hoff, R.; Badylak, S.F.; Wagner, W.R. Bi-layered polyurethane—Extracellular matrix cardiac patch improves ischemic ventricular wall remodeling in a rat model. Biomaterials 2016, 107, 1–14. [Google Scholar] [CrossRef] [PubMed]
- Huang, K.; Ozpinar, E.W.; Su, T.; Tang, J.; Shen, D.; Qiao, L.; Hu, S.; Li, Z.; Liang, H.; Mathews, K.; et al. An off-the-shelf artificial cardiac patch improves cardiac repair after myocardial infarction in rats and pigs. Sci. Transl. Med. 2020, 12, eaat9683. [Google Scholar] [CrossRef] [PubMed]
- Guyette, J.P.; Charest, J.M.; Mills, R.W.; Jank, B.J.; Moser, P.T.; Gilpin, S.E.; Gershlak, J.R.; Okamoto, T.; Gonzalez, G.; Milan, D.J.; et al. Bioengineering human myocardium on native extracellular matrix. Circ. Res. 2016, 118, 56–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shimizu, T.; Yamato, M.; Isoi, Y.; Akutsu, T.; Setomaru, T.; Abe, K.; Kikuchi, A.; Umezu, M.; Okano, T. Fabrication of pulsatile cardiac tissue grafts using a novel 3-dimensional cell sheet manipulation technique and temperature-responsive cell culture surfaces. Circ. Res. 2002, 90, e40. [Google Scholar] [CrossRef] [Green Version]
- Kawamura, M.; Miyagawa, S.; Miki, K.; Saito, A.; Fukushima, S.; Higuchi, T.; Kawamura, T.; Kuratani, T.; Daimon, T.; Shimizu, T.; 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, S29–S37. [Google Scholar] [CrossRef] [Green Version]
- Goldfracht, I.; Efraim, Y.; Shinnawi, R.; Kovalev, E.; Huber, I.; Gepstein, A.; Arbel, G.; Shaheen, N.; Tiburcy, M.; Zimmermann, W.H.; et al. Engineered heart tissue models from hiPSC-derived cardiomyocytes and cardiac ECM for disease modeling and drug testing applications. Acta Biomater. 2019, 92, 145–159. [Google Scholar] [CrossRef]
- Weinberger, F.; Breckwoldt, K.; Pecha, S.; Kelly, A.; Geertz, B.; Starbatty, J.; Yorgan, T.; Cheng, K.H.; Lessmann, K.; Stolen, T.; et al. Cardiac repair in guinea pigs with human engineered heart tissue from induced pluripotent stem cells. Sci. Transl. Med. 2016, 8, 363ra148. [Google Scholar] [CrossRef]
- Bian, W.; Badie, N.; Himel, H.D., 4th; Bursac, N. Robust T-tubulation and maturation of cardiomyocytes using tissue-engineered epicardial mimetics. Biomaterials 2014, 35, 3819–3828. [Google Scholar] [CrossRef] [Green Version]
- Shadrin, I.Y.; Allen, B.W.; Qian, Y.; Jackman, C.P.; Carlson, A.L.; Juhas, M.E.; Bursac, N. Cardiopatch platform enables maturation and scale-up of human pluripotent stem cell-derived engineered heart tissues. Nat. Commun. 2017, 8, 1825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ma, Z.; Wang, J.; Loskill, P.; Huebsch, N.; Koo, S.; Svedlund, F.L.; Marks, N.C.; Hua, E.W.; Grigoropoulos, C.P.; Conklin, B.R.; et al. Self-organizing human cardiac microchambers mediated by geometric confinement. Nat. Commun. 2015, 6, 7413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Drakhlis, L.; Biswanath, S.; Farr, C.M.; Lupanow, V.; Teske, J.; Ritzenhoff, K.; Franke, A.; Manstein, F.; Bolesani, E.; Kempf, H.; et al. Human heart-forming organoids recapitulate early heart and foregut development. Nat. Biotechnol. 2021, 39, 737–746. [Google Scholar] [CrossRef] [PubMed]
- Hofbauer, P.; Jahnel, S.M.; Papai, N.; Giesshammer, M.; Deyett, A.; Schmidt, C.; Penc, M.; Tavernini, K.; Grdseloff, N.; Meledeth, C.; et al. Cardioids reveal self-organizing principles of human cardiogenesis. Cell 2021, 184, 3299–3317. [Google Scholar] [CrossRef]
- MacQueen, L.A.; Sheehy, S.P.; Chantre, C.O.; Zimmerman, J.F.; Pasqualini, F.S.; Liu, X.; Goss, J.A.; Campbell, P.H.; Gonzalez, G.M.; Park, S.J.; et al. A tissue-engineered scale model of the heart ventricle. Nat. Biomed. Eng. 2018, 2, 930–941. [Google Scholar] [CrossRef]
- Gouveia, P.J.; Rosa, S.; Ricotti, L.; Abecasis, B.; Almeida, H.V.; Monteiro, L.; Nunes, J.; Carvalho, F.S.; Serra, M.; Luchkin, S.; et al. Flexible nanofilms coated with aligned piezoelectric microfibers preserve the contractility of cardiomyocytes. Biomaterials 2017, 139, 213–228. [Google Scholar] [CrossRef]
- Feiner, R.; Engel, L.; Fleischer, S.; Malki, M.; Gal, I.; Shapira, A.; Shacham-Diamand, Y.; Dvir, T. Engineered hybrid cardiac patches with multifunctional electronics for online monitoring and regulation of tissue function. Nat. Mater. 2016, 15, 679–685. [Google Scholar] [CrossRef] [Green Version]
- Feinberg, A.W.; Feigel, A.; Shevkoplyas, S.S.; Sheehy, S.; Whitesides, G.M.; Parker, K.K. Muscular thin films for building actuators and powering devices. Science 2007, 317, 1366–1370. [Google Scholar] [CrossRef] [Green Version]
- Zhang, B.; Montgomery, M.; Chamberlain, M.D.; Ogawa, S.; Korolj, A.; Pahnke, A.; Wells, L.A.; Massé, S.; Kim, J.; Reis, L.; et al. Biodegradable scaffold with built-in vasculature for organ-on-a-chip engineering and direct surgical anastomosis. Nat. Mater. 2016, 15, 669–678. [Google Scholar] [CrossRef] [Green Version]
- Bian, W.; Jackman, C.P.; Bursac, N. Controlling the structural and functional anisotropy of engineered cardiac tissues. Biofabrication 2014, 6, 024109. [Google Scholar] [CrossRef]
- Vunjak-Novakovic, G.; Ronaldson-Bouchard, K.; Radisic, M. Organs-on-a-chip models for biological research. Cell 2021, 184, 4597–4611. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Y.; Rafatian, N.; Feric, N.T.; Cox, B.J.; Aschar-Sobbi, R.; Wang, E.Y.; Aggarwal, P.; Zhang, B.; Conant, G.; Ronaldson-Bouchard, K.; et al. A platform for generation of chamber-specific cardiac tissues and disease modeling. Cell 2019, 176, 913–927. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lind, J.U.; Busbee, T.A.; Valentine, A.D.; Pasqualini, F.S.; Yuan, H.; Yadid, M.; Park, S.J.; Kotikian, A.; Nesmith, A.P.; Campbell, P.H.; et al. Instrumented cardiac microphysiological devices via multimaterial three-dimensional printing. Nat. Mater. 2017, 16, 303–308. [Google Scholar] [CrossRef] [PubMed]
- Lee, A.; Hudson, A.R.; Shiwarski, D.J.; Tashman, J.W.; Hinton, T.J.; Yerneni, S.; Bliley, J.M.; Campbell, P.G.; Feinberg, A.W. 3D bioprinting of collagen to rebuild components of the human heart. Science 2019, 365, 482–487. [Google Scholar] [CrossRef]
- Hinton, T.J.; Jallerat, Q.; Palchesko, R.N.; Park, J.H.; Grodzicki, M.S.; Shue, H.J.; Ramadan, M.H.; Hudson, A.R.; Feinberg, A.W. Three-dimensional printing of complex biological structures by freeform reversible embedding of suspended hydrogels. Sci. Adv. 2015, 1, e1500758. [Google Scholar] [CrossRef] [Green Version]
- Grigoryan, B.; Paulsen, S.J.; Corbett, D.C.; Sazer, D.W.; Fortin, C.L.; Zaita, A.J.; Greenfield, P.T.; Calafat, N.J.; Gounley, J.P.; Ta, A.H.; et al. Multivascular networks and functional intravascular topologies within biocompatible hydrogels. Science 2019, 364, 458–464. [Google Scholar] [CrossRef]
- Zhang, Y.S.; Arneri, A.; Bersini, S.; Shin, S.R.; Zhu, K.; Goli-Malekabadi, Z.; Aleman, J.; Colosi, C.; Busignani, F.; Dell’Erba, V.; et al. Bioprinting 3D microfibrous scaffolds for engineering endothelialized myocardium and heart-on-a-chip. Biomaterials 2016, 110, 45–59. [Google Scholar] [CrossRef] [Green Version]
- Skylar-Scott, M.A.; Uzel, S.G.M.; Nam, L.L.; Ahrens, J.H.; Truby, R.L.; Damaraju, S.; Lewis, J.A. Biomanufacturing of organ-specific tissues with high cellular density and embedded vascular channels. Sci. Adv. 2019, 5, eaaw2459. [Google Scholar] [CrossRef] [Green Version]
- Eschenhagen, T.; Fink, C.; Remmers, U.; Scholz, H.; Wattchow, J.; Weil, J.; Zimmermann, W.; Dohmen, H.H.; Schäfer, H.; Bishopric, N.; et al. Three-dimensional reconstitution of embryonic cardiomyocytes in a collagen matrix: A new heart muscle model system. FASEB J. 1997, 11, 683–694. [Google Scholar] [CrossRef]
- Boudou, T.; Legant, W.R.; Mu, A.; Borochin, M.A.; Thavandiran, N.; Radisic, M.; Zandstra, P.W.; Epstein, J.A.; Margulies, K.B.; Chen, C.S. A microfabricated platform to measure and manipulate the mechanics of engineered cardiac microtissues. Tissue Eng. Part A 2012, 18, 910–919. [Google Scholar] [CrossRef]
- Takasato, M.; Er, P.X.; Chiu, H.S.; Maier, B.; Baillie, G.J.; Ferguson, C.; Parton, R.G.; Wolvetang, E.J.; Roost, M.S.; Chuva de Sousa Lopes, S.M.; et al. Kidney organoids from human iPS cells contain multiple lineages and model human nephrogenesis. Nature 2015, 526, 564–568. [Google Scholar] [CrossRef] [PubMed]
- Serra, D.; Mayr, U.; Boni, A.; Lukonin, I.; Rempfler, M.; Challet Meylan, L.; Stadler, M.B.; Strnad, P.; Papasaikas, P.; Vischi, D.; et al. Self-organization and symmetry breaking in intestinal organoid development. Nature 2019, 569, 66–72. [Google Scholar] [CrossRef] [PubMed]
- Mansour, A.A.; Gonçalves, J.T.; Bloyd, C.W.; Li, H.; Fernandes, S.; Quang, D.; Johnston, S.; Parylak, S.L.; Jin, X.; Gage, F.H. An in vivo model of functional and vascularized human brain organoids. Nat. Biotechnol. 2018, 36, 432–441. [Google Scholar] [CrossRef] [PubMed]
- Lee, R.T.; Walsh, K. The future of cardiovascular regenerative medicine. Circulation 2016, 133, 2618–2625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Richards, D.J.; Coyle, R.C.; Tan, Y.; Jia, J.; Wong, K.; Toomer, K.; Menick, D.R.; Mei, Y. Inspiration from heart development: Biomimetic development of functional human cardiac organoids. Biomaterials 2017, 142, 112–123. [Google Scholar] [CrossRef] [PubMed]
- Richards, D.J.; Li, Y.; Kerr, C.M.; Yao, J.; Beeson, G.C.; Coyle, R.C.; Chen, X.; Jia, J.; Damon, B.; Wilson, R.; et al. Human cardiac organoids for the modelling of myocardial infarction and drug cardiotoxicity. Nat. Biomed. Eng. 2020, 4, 446–462. [Google Scholar] [CrossRef]
- Ogle, B.M.; Bursac, N.; Domian, I.; Huang, N.F.; Menasché, P.; Murry, C.E.; Pruitt, B.; Radisic, M.; Wu, J.C.; Wu, S.M.; et al. Distilling complexity to advance cardiac tissue engineering. Sci. Transl. Med. 2016, 8, 342ps13. [Google Scholar] [CrossRef] [Green Version]
- Borovjagin, A.V.; Ogle, B.M.; Berry, J.L.; Zhang, J. From microscale devices to 3D printing: Advances in fabrication of 3D cardiovascular tissues. Circ. Res. 2017, 120, 150–165. [Google Scholar] [CrossRef]
- Murphy, S.V.; Atala, A. 3D bioprinting of tissues and organs. Nat. Biotechnol. 2014, 32, 773–785. [Google Scholar] [CrossRef] [PubMed]
- Wang, Z.; Lee, S.J.; Cheng, H.J.; Yoo, J.J.; Atala, A. 3D bioprinted functional and contractile cardiac tissue constructs. Acta. Biomater. 2018, 70, 48–56. [Google Scholar] [CrossRef]
- Qasim, M.; Haq, F.; Kang, M.H.; Kim, J.H. 3D printing approaches for cardiac tissue engineering and role of immune modulation in tissue regeneration. Int. J. Nanomed. 2019, 14, 1311–1333. [Google Scholar] [CrossRef] [Green Version]
- Maiullari, F.; Costantini, M.; Milan, M.; Pace, V.; Chirivì, M.; Maiullari, S.; Rainer, A.; Baci, D.; Marei, H.E.; Seliktar, D.; et al. A multi-cellular 3D bioprinting approach for vascularized heart tissue engineering based on HUVECs and iPSC-derived cardiomyocytes. Sci. Rep. 2018, 8, 13532. [Google Scholar] [CrossRef] [PubMed]
- Pati, F.; Jang, J.; Ha, D.H.; Won Kim, S.; Rhie, J.W.; Shim, J.H.; Kim, D.H.; Cho, D.W. Printing three-dimensional tissue analogues with decellularized extracellular matrix bioink. Nat. Commun. 2014, 5, 3935. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Badylak, S.F.; Taylor, D.; Uygun, K. Whole-organ tissue engineering: Decellularization and recellularization of three-dimensional matrix scaffolds. Annu. Rev. Biomed. Eng. 2011, 13, 27–53. [Google Scholar] [CrossRef] [PubMed]
- Kupfer, M.E.; Lin, W.H.; Ravikumar, V.; Qiu, K.; Wang, L.; Gao, L.; Bhuiyan, D.B.; Lenz, M.; Ai, J.; Mahutga, R.R.; et al. In situ expansion, differentiation, and electromechanical coupling of human cardiac muscle in a 3D bioprinted, chambered organoid. Circ. Res. 2020, 127, 207–224. [Google Scholar] [CrossRef]
- Vegas, A.J.; Veiseh, O.; Doloff, J.C.; Ma, M.; Tam, H.H.; Bratlie, K.; Li, J.; Bader, A.R.; Langan, E.; Olejnik, K.; et al. Combinatorial hydrogel library enables identification of materials that mitigate the foreign body response in primates. Nat. Biotechnol. 2016, 34, 345–352. [Google Scholar] [CrossRef] [Green Version]
- Dondossola, E.; Holzapfel, B.M.; Alexander, S.; Filippini, S.; Hutmacher, D.W.; Friedl, P. Examination of the foreign body response to biomaterials by nonlinear intravital microscopy. Nat. Biomed. Eng. 2016, 1, 0007. [Google Scholar] [CrossRef] [Green Version]
- Doloff, J.C.; Veiseh, O.; Vegas, A.J.; Tam, H.H.; Farah, S.; Ma, M.; Li, J.; Bader, A.; Chiu, A.; Sadraei, A.; et al. Colony stimulating factor-1 receptor is a central component of the foreign body response to biomaterial implants in rodents and non-human primates. Nat. Mater. 2017, 16, 671–680. [Google Scholar] [CrossRef]
- 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 2016, 113, 3179–3184. [Google Scholar] [CrossRef] [Green Version]
- Zhu, W.; Qu, X.; Zhu, J.; Ma, X.; Patel, S.; Liu, J.; Wang, P.; Lai, C.S.; Gou, M.; Xu, Y.; et al. Direct 3D bioprinting of prevascularized tissue constructs with complex microarchitecture. Biomaterials 2017, 124, 106–115. [Google Scholar] [CrossRef] [Green Version]
- Bernal, P.N.; Delrot, P.; Loterie, D.; Li, Y.; Malda, J.; Moser, C.; Levato, R. Volumetric bioprinting of complex living-tissue constructs within seconds. Adv. Mater. 2019, 31, e1904209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Z.; Wang, L.; Li, T.; Liu, S.; Guo, B.; Huang, W.; Wu, Y. 3D bioprinting in cardiac tissue engineering. Theranostics 2021, 11, 7948–7969. [Google Scholar] [CrossRef] [PubMed]
- Kozaniti, F.K.; Metsiou, D.N.; Manara, A.E.; Athanassiou, G.; Deligianni, D.D. Recent advancements in 3D printing and bioprinting methods for cardiovascular tissue engineering. Bioengineering 2021, 8, 133. [Google Scholar] [CrossRef] [PubMed]
- Tulloch, N.L.; Muskheli, V.; Razumova, M.V.; Korte, F.S.; Regnier, M.; Hauch, K.D.; Pabon, L.; Reinecke, H.; Murry, C.E. Growth of engineered human myocardium with mechanical loading and vascular coculture. Circ. Res. 2011, 109, 47–59. [Google Scholar] [CrossRef] [PubMed]
- Feric, N.T.; Pallotta, I.; Singh, R.; Bogdanowicz, D.R.; Gustilo, M.; Chaudhary, K.; Willette, R.N.; Chendrimada, T.; Xu, X.; Graziano, M.P.; et al. Engineered cardiac tissues generated in the Biowire™ II: A platform for human-based drug discovery. Toxicol. Sci. 2019, 172, 89–97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Braam, S.R.; Passier, R.; Mummery, C.L. Cardiomyocytes from human pluripotent stem cells in regenerative medicine and drug discovery. Trends Pharmacol. Sci. 2009, 30, 536–545. [Google Scholar] [CrossRef] [PubMed]
- Magdy, T.; Schuldt, A.J.T.; Wu, J.C.; Bernstein, D.; Burridge, P.W. Human induced pluripotent stem cell (hiPSC)-derived cells to assess drug cardiotoxicity: Opportunities and problems. Annu. Rev. Pharmacol. Toxicol. 2018, 58, 83–103. [Google Scholar] [CrossRef]
- Marsano, A.; Conficconi, C.; Lemme, M.; Occhetta, P.; Gaudiello, E.; Votta, E.; Cerino, G.; Redaelli, A.; Rasponi, M. Beating heart on a chip: A novel microfluidic platform to generate functional 3D cardiac microtissues. Lab Chip 2016, 16, 599–610. [Google Scholar] [CrossRef]
- Kensah, G.; Roa Lara, A.; Dahlmann, J.; Zweigerdt, R.; Schwanke, K.; Hegermann, J.; Skvorc, D.; Gawol, A.; Azizian, A.; Wagner, S.; et al. Murine and human pluripotent stem cell-derived cardiac bodies form contractile myocardial tissue in vitro. Eur. Heart J. 2013, 34, 1134–1146. [Google Scholar] [CrossRef] [Green Version]
- Bristow, M.R.; Ginsburg, R.; Strosberg, A.; Montgomery, W.; Minobe, W. Pharmacology and inotropic potential of forskolin in the human heart. J. Clin. Investig. 1984, 74, 212–223. [Google Scholar] [CrossRef]
- Molenaar, P.; Christ, T.; Hussain, R.I.; Engel, A.; Berk, E.; Gillette, K.T.; Chen, L.; Galindo-Tovar, A.; Krobert, K.A.; Ravens, U.; et al. PDE3, but not PDE4, reduces β1- and β2;-adrenoceptor-mediated inotropic and lusitropic effects in failing ventricle from metoprolol-treated patients. Br. J. Pharmacol. 2013, 169, 528–538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zuppinger, C. 3D cardiac cell culture: A critical review of current technologies and applications. Front. Cardiovasc. Med. 2019, 6, 87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lind, J.U.; Yadid, M.; Perkins, I.; O’Connor, B.B.; Eweje, F.; Chantre, C.O.; Hemphill, M.A.; Yuan, H.; Campbell, P.H.; Vlassak, J.J.; et al. Cardiac microphysiological devices with flexible thin-film sensors for higher-throughput drug screening. Lab Chip 2017, 17, 3692–3703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, B.; Radisic, M. Organ-on-a-chip devices advance to market. Lab Chip 2017, 17, 2395–2420. [Google Scholar] [CrossRef] [PubMed]
- Smith, J.G.W.; Owen, T.; Bhagwan, J.R.; Mosqueira, D.; Scott, E.; Mannhardt, I.; Patel, A.; Barriales-Villa, R.; Monserrat, L.; Hansen, A.; et al. Isogenic pairs of hiPSC-CMs with hypertrophic cardiomyopathy/LVNC-associated ACTC1 E99K mutation unveil differential functional deficits. Stem Cell Rep. 2018, 11, 1226–1243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- de la Roche, J.; Angsutararux, P.; Kempf, H.; Janan, M.; Bolesani, E.; Thiemann, S.; Wojciechowski, D.; Coffee, M.; Franke, A.; Schwanke, K.; et al. Comparing human iPSC-cardiomyocytes versus HEK293T cells unveils disease-causing effects of Brugada mutation A735V of NaV1.5 sodium channels. Sci. Rep. 2019, 9, 11173. [Google Scholar] [CrossRef] [Green Version]
- Bellin, M.; Casini, S.; Davis, R.P.; D’Aniello, C.; Haas, J.; Ward-van Oostwaard, D.; Tertoolen, L.G.; Jung, C.B.; Elliott, D.A.; Welling, A.; et al. Isogenic human pluripotent stem cell pairs reveal the role of a KCNH2 mutation in long-QT syndrome. EMBO J. 2013, 32, 3161–3175. [Google Scholar] [CrossRef]
- Wang, G.; McCain, M.L.; Yang, L.; He, A.; Pasqualini, F.S.; Agarwal, A.; Yuan, H.; Jiang, D.; Zhang, D.; Zangi, L.; et al. Modeling the mitochondrial cardiomyopathy of Barth syndrome with induced pluripotent stem cell and heart-on-chip technologies. Nat. Med. 2014, 20, 616–623. [Google Scholar] [CrossRef]
- Mosqueira, D.; Mannhardt, I.; Bhagwan, J.R.; Lis-Slimak, K.; Katili, P.; Scott, E.; Hassan, M.; Prondzynski, M.; Harmer, S.C.; Tinker, A.; 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, 3879–3892. [Google Scholar] [CrossRef]
- Hinson, J.T.; Chopra, A.; Nafissi, N.; Polacheck, W.J.; Benson, C.C.; Swist, S.; Gorham, J.; Yang, L.; Schafer, S.; Sheng, C.C.; et al. HEART DISEASE. Titin mutations in iPS cells define sarcomere insufficiency as a cause of dilated cardiomyopathy. Science 2015, 349, 982–986. [Google Scholar] [CrossRef] [Green Version]
- Prondzynski, M.; Lemoine, M.D.; Zech, A.T.; Horváth, A.; Di Mauro, V.; Koivumäki, J.T.; Kresin, N.; Busch, J.; Krause, T.; Krämer, E.; et al. Disease modeling of a mutation in α-actinin 2 guides clinical therapy in hypertrophic cardiomyopathy. EMBO Mol. Med. 2019, 11, e11115. [Google Scholar] [CrossRef] [PubMed]
- Gangadhar, T.C.; Von der Lohe, E.; Sawada, S.G.; Helft, P.R. Takotsubo cardiomyopathy in a patient with esophageal cancer: A case report. J. Med. Case Rep. 2008, 2, 379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rosen, M.R.; Myerburg, R.J.; Francis, D.P.; Cole, G.D.; Marbán, E. Translating stem cell research to cardiac disease therapies: Pitfalls and prospects for improvement. J. Am. Coll. Cardiol. 2014, 64, 922–937. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gerbin, K.A.; Murry, C.E. The winding road to regenerating the human heart. Cardiovasc. Pathol. 2015, 24, 133–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Eschenhagen, T.; Bolli, R.; Braun, T.; Field, L.J.; Fleischmann, B.K.; Frisén, J.; Giacca, M.; Hare, J.M.; Houser, S.; Lee, R.T.; et al. Cardiomyocyte regeneration: A consensus statement. Circulation 2017, 136, 680–686. [Google Scholar] [CrossRef] [PubMed]
- Terrovitis, J.; Lautamäki, R.; Bonios, M.; Fox, J.; Engles, J.M.; Yu, J.; Leppo, M.K.; Pomper, M.G.; Wahl, R.L.; Seidel, J.; et al. Noninvasive quantification and optimization of acute cell retention by in vivo positron emission tomography after intramyocardial cardiac-derived stem cell delivery. J. Am. Coll. Cardiol. 2009, 54, 1619–1626. [Google Scholar] [CrossRef] [Green Version]
- Chong, J.J.; Yang, X.; Don, C.W.; Minami, E.; Liu, Y.W.; Weyers, J.J.; Mahoney, W.M.; Van Biber, B.; Cook, S.M.; Palpant, N.J.; et al. Human embryonic-stem-cell-derived cardiomyocytes regenerate non-human primate hearts. Nature 2014, 510, 273–277. [Google Scholar] [CrossRef]
- Shiba, Y.; Gomibuchi, T.; Seto, T.; Wada, Y.; Ichimura, H.; Tanaka, Y.; Ogasawara, T.; Okada, K.; Shiba, N.; Sakamoto, K.; et al. Allogeneic transplantation of iPS cell-derived cardiomyocytes regenerates primate hearts. Nature 2016, 538, 388–391. [Google Scholar] [CrossRef]
- Galdos, F.X.; Guo, Y.; Paige, S.L.; VanDusen, N.J.; Wu, S.M.; Pu, W.T. Cardiac Regeneration: Lessons From Development. Circ. Res. 2017, 120, 941–959. [Google Scholar] [CrossRef] [Green Version]
- Madonna, R.; Van Laake, L.W.; Botker, H.E.; Davidson, S.M.; De Caterina, R.; Engel, F.B.; Eschenhagen, T.; Fernandez-Aviles, F.; Hausenloy, D.J.; Hulot, J.S.; et al. ESC working group on cellular biology of the heart: Position paper for cardiovascular research: Tissue engineering strategies combined with cell therapies for cardiac repair in ischaemic heart disease and heart failure. Cardiovasc. Res. 2019, 115, 488–500. [Google Scholar] [CrossRef] [Green Version]
- Silver, S.E.; Barrs, R.W.; Mei, Y. Transplantation of human pluripotent stem cell-derived cardiomyocytes for cardiac regenerative therapy. Front. Cardiovasc. Med. 2021, 8, 707890. [Google Scholar] [CrossRef] [PubMed]
- Mirotsou, M.; Jayawardena, T.M.; Schmeckpeper, J.; Gnecchi, M.; Dzau, V.J. Paracrine mechanisms of stem cell reparative and regenerative actions in the heart. J. Mol. Cell Cardiol. 2011, 50, 280–289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pecha, S.; Yorgan, K.; Röhl, M.; Geertz, B.; Hansen, A.; Weinberger, F.; Sehner, S.; Ehmke, H.; Reichenspurner, H.; Eschenhagen, T.; et al. Human iPS cell-derived engineered heart tissue does not affect ventricular arrhythmias in a guinea pig cryo-injury model. Sci. Rep. 2019, 9, 9831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lin, X.; Liu, Y.; Bai, A.; Cai, H.; Bai, Y.; Jiang, W.; Yang, H.; Wang, X.; Yang, L.; Sun, N.; et al. A viscoelastic adhesive epicardial patch for treating myocardial infarction. Nat. Biomed. Eng. 2019, 3, 632–643. [Google Scholar] [CrossRef]
- Zimmermann, W.H.; Melnychenko, I.; Wasmeier, G.; Didié, M.; Naito, H.; Nixdorff, U.; Hess, A.; Budinsky, L.; Brune, K.; Michaelis, B.; et al. Engineered heart tissue grafts improve systolic and diastolic function in infarcted rat hearts. Nat. Med. 2006, 12, 452–458. [Google Scholar] [CrossRef]
- Querdel, E.; Reinsch, M.; Castro, L.; Köse, D.; Bähr, A.; Reich, S.; Geertz, B.; Ulmer, B.; Schulze, M.; Lemoine, M.D.; et al. Human engineered heart tissue patches remuscularize the injured heart in a dose-dependent manner. Circulation 2021, 143, 1991–2006. [Google Scholar] [CrossRef]
- Kawamura, T.; Miyagawa, S.; Fukushima, S.; Maeda, A.; Kashiyama, N.; Kawamura, A.; Miki, K.; Okita, K.; Yoshida, Y.; Shiina, T.; et al. Cardiomyocytes derived from MHC-Homozygous induced pluripotent stem cells exhibit reduced allogeneic immunogenicity in MHC-matched non-human primates. Stem Cell Rep. 2016, 6, 312–320. [Google Scholar] [CrossRef] [Green Version]
- Madonna, R. Human-induced pluripotent stem cells: In quest of clinical applications. Mol. Biotechnol. 2012, 52, 193–203. [Google Scholar] [CrossRef]
- Almeida, S.O.; Skelton, R.J.; Adigopula, S.; Ardehali, R. Arrhythmia in stem cell transplantation. Card. Electrophysiol. Clin. 2015, 7, 357–370. [Google Scholar] [CrossRef] [Green Version]
- 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. 2016, 1, 953–962. [Google Scholar] [CrossRef] [Green Version]
- Frey, N.; Linke, A.; Süselbeck, T.; Müller-Ehmsen, J.; Vermeersch, P.; Schoors, D.; Rosenberg, M.; Bea, F.; Tuvia, S.; Leor, J. Intracoronary delivery of injectable bioabsorbable scaffold (IK-5001) to treat left ventricular remodeling after ST-elevation myocardial infarction: A first-in-man study. Circ. Cardiovasc. Interv. 2014, 7, 806–812. [Google Scholar] [CrossRef] [PubMed]
- Rao, S.V.; Zeymer, U.; Douglas, P.S.; Al-Khalidi, H.; Liu, J.; Gibson, C.M.; Harrison, R.W.; Joseph, D.S.; Heyrman, R.; Krucoff, M.W. A randomized, double-blind, placebo-controlled trial to evaluate the safety and effectiveness of intracoronary application of a novel bioabsorbable cardiac matrix for the prevention of ventricular remodeling after large ST-segment elevation myocardial infarction: Rationale and design of the PRESERVATION I trial. Am. Heart J. 2015, 170, 929–937. [Google Scholar] [PubMed]
- Rao, S.V.; Zeymer, U.; Douglas, P.S.; Al-Khalidi, H.; White, J.A.; Liu, J.; Levy, H.; Guetta, V.; Gibson, C.M.; Tanguay, J.F.; et al. Bioabsorbable intracoronary matrix for prevention of ventricular remodeling after myocardial infarction. J. Am. Coll. Cardiol. 2016, 68, 715–723. [Google Scholar] [CrossRef] [PubMed]
- Anker, S.D.; Coats, A.J.; Cristian, G.; Dragomir, D.; Pusineri, E.; Piredda, M.; Bettari, L.; Dowling, R.; Volterrani, M.; Kirwan, B.A.; et al. A prospective comparison of alginate-hydrogel with standard medical therapy to determine impact on functional capacity and clinical outcomes in patients with advanced heart failure (AUGMENT-HF trial). Eur. Heart J. 2015, 36, 2297–2309. [Google Scholar] [CrossRef] [Green Version]
- Mann, D.L.; Lee, R.J.; Coats, A.J.; Neagoe, G.; Dragomir, D.; Pusineri, E.; Piredda, M.; Bettari, L.; Kirwan, B.A.; Dowling, R.; et al. One-year follow-up results from AUGMENT-HF: A multicentre randomized controlled clinical trial of the efficacy of left ventricular augmentation with Algisyl in the treatment of heart failure. Eur. J. Heart Fail. 2016, 18, 314–325. [Google Scholar] [CrossRef]
- Lee, R.J.; Hinson, A.; Bauernschmitt, R.; Matschke, K.; Fang, Q.; Mann, D.L.; Dowling, R.; Schiller, N.; Sabbah, H.N. The feasibility and safety of Algisyl-LVR™ as a method of left ventricular augmentation in patients with dilated cardiomyopathy: Initial first in man clinical results. Int. J. Cardiol. 2015, 199, 18–24. [Google Scholar] [CrossRef]
- Yanagawa, B.; Rao, V.; Yau, T.M.; Cusimano, R.J. Potential myocardial regeneration with CorMatrix ECM: A case report. J. Thorac. Cardiovasc. Surg. 2014, 147, e41–e43. [Google Scholar] [CrossRef] [Green Version]
- Mewhort, H.E.; Turnbull, J.D.; Meijndert, H.C.; Ngu, J.M.; Fedak, P.W. Epicardial infarct repair with basic fibroblast growth factor-enhanced CorMatrix-ECM biomaterial attenuates postischemic cardiac remodeling. J. Thorac. Cardiovasc. Surg. 2014, 147, 1650–1659. [Google Scholar] [CrossRef] [Green Version]
- Mewhort, H.E.; Turnbull, J.D.; Satriano, A.; Chow, K.; Flewitt, J.A.; Andrei, A.C.; Guzzardi, D.G.; Svystonyuk, D.A.; White, J.A.; Fedak, P.W. Epicardial infarct repair with bioinductive extracellular matrix promotes vasculogenesis and myocardial recovery. J. Heart Lung Transplant. 2016, 35, 661–670. [Google Scholar] [CrossRef] [Green Version]
- Traverse, J.H.; Henry, T.D.; Dib, N.; Patel, A.N.; Pepine, C.; Schaer, G.L.; DeQuach, J.A.; Kinsey, A.M.; Chamberlin, P.; Christman, K.L. First-in-man study of a cardiac extracellular matrix hydrogel in early and late myocardial infarction patients. JACC Basic Transl. Sci. 2019, 4, 659–669. [Google Scholar] [CrossRef]
- Menasché, P.; Vanneaux, V.; Hagège, A.; Bel, A.; Cholley, B.; Cacciapuoti, I.; Parouchev, A.; Benhamouda, N.; Tachdjian, G.; Tosca, L.; et al. Human embryonic stem cell-derived cardiac progenitors for severe heart failure treatment: First clinical case report. Eur. Heart J. 2015, 36, 2011–2017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Menasché, P.; Vanneaux, V.; Hagège, A.; Bel, A.; Cholley, B.; Parouchev, A.; Cacciapuoti, I.; Al-Daccak, R.; Benhamouda, N.; Blons, H.; et al. Transplantation of human embryonic stem cell-derived cardiovascular progenitors for severe ischemic left ventricular dysfunction. J. Am. Coll. Cardiol. 2018, 71, 429–438. [Google Scholar] [CrossRef] [PubMed]
- Kashiyama, N.; Miyagawa, S.; Fukushima, S.; Kawamura, T.; Kawamura, A.; Yoshida, S.; Eiraku, S.; Harada, A.; Matsunaga, K.; Watabe, T.; et al. MHC-mismatched allotransplantation of induced pluripotent stem cell-derived cardiomyocyte sheets to improve cardiac function in a primate ischemic cardiomyopathy model. Transplantation 2019, 103, 1582–1590. [Google Scholar] [CrossRef] [PubMed]
- Gyöngyösi, M.; Dib, N. Diagnostic and prognostic value of 3D NOGA mapping in ischemic heart disease. Nat. Rev. Cardiol. 2011, 8, 393–404. [Google Scholar] [CrossRef]
- Schmauss, D.; Haeberle, S.; Hagl, C.; Sodian, R. Three-dimensional printing in cardiac surgery and interventional cardiology: A single-centre experience. Eur. J. Cardiothorac. Surg. 2015, 47, 1044–1052. [Google Scholar] [CrossRef] [Green Version]
- Vukicevic, M.; Mosadegh, B.; Min, J.K.; Little, S.H. Cardiac 3D printing and its future directions. JACC Cardiovasc. Imaging 2017, 10, 171–184. [Google Scholar] [CrossRef]
- Hendrickson, T.; Mancino, C.; Whitney, L.; Tsao, C.; Rahimi, M.; Taraballi, F. Mimicking cardiac tissue complexity through physical cues: A review on cardiac tissue engineering approaches. Nanomedicine 2021, 33, 102367. [Google Scholar] [CrossRef]
- Chien, K.R.; Zangi, L.; Lui, K.O. Synthetic chemically modified mRNA (modRNA): Toward a new technology platform for cardiovascular biology and medicine. Cold Spring Harb. Perspect. Med. 2014, 5, a014035. [Google Scholar] [CrossRef] [Green Version]
- Carlsson, L.; Clarke, J.C.; Yen, C.; Gregoire, F.; Albery, T.; Billger, M.; Egnell, A.C.; Gan, L.M.; Jennbacken, K.; Johansson, E.; et al. Biocompatible, purified VEGF-A mRNA improves cardiac function after intracardiac injection 1 week post-myocardial infarction in Swine. Mol. Ther. Methods Clin. Dev. 2018, 9, 330–346. [Google Scholar] [CrossRef] [Green Version]
- Gan, L.M.; Lagerström-Fermér, M.; Carlsson, L.G.; Arfvidsson, C.; Egnell, A.C.; Rudvik, A.; Kjaer, M.; Collén, A.; Thompson, J.D.; Joyal, J.; et al. Intradermal delivery of modified mRNA encoding VEGF-A in patients with type 2 diabetes. Nat. Commun. 2019, 10, 871. [Google Scholar] [CrossRef]
- Sluijter, J.P.; Verhage, V.; Deddens, J.C.; van den Akker, F.; Doevendans, P.A. Microvesicles and exosomes for intracardiac communication. Cardiovasc. Res. 2014, 102, 302–311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Adamiak, M.; Sahoo, S. Exosomes in myocardial repair: Advances and challenges in the development of next-generation therapeutics. Mol. Ther. 2018, 26, 1635–1643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Arslan, F.; Lai, R.C.; Smeets, M.B.; Akeroyd, L.; Choo, A.; Aguor, E.N.; Timmers, L.; van Rijen, H.V.; Doevendans, P.A.; Pasterkamp, G.; et al. Mesenchymal stem cell-derived exosomes increase ATP levels, decrease oxidative stress and activate PI3K/Akt pathway to enhance myocardial viability and prevent adverse remodeling after myocardial ischemia/reperfusion injury. Stem Cell Res. 2013, 10, 301–312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Major Classification | Type of Strategies | Subtype | References |
---|---|---|---|
Physiological stimulation | Mechanical stress loading | [40,41,42,43] | |
Electrical stimulation/pacing | [42,44,45] | ||
Advanced culture | 3D cultures | EHTs with a scaffold protein | [46,47,48,49] |
Cardiac spheroids without scaffold proteins | [47,50,51] | ||
Intercellular crosstalk | Co-culture/administration with ECs | [52,53] | |
Co-culture/administration with FBs | [48] | ||
Co-culture/administration with epicardial cells | [54] | ||
Co-culture/administration with SMCs/ECs and FBs | [55,56,57,58] | ||
Biochemical intervention | Metabolic intervention | High fatty acid/low glucose diet | [59,60] |
Hormone intervention | Tri-iodothyronine (T3) | [61,62] | |
Glucocorticoid | [63] | ||
Paracrine signals | Growth factors | [23,64] | |
Genetic regulation | Overexpression of maturation-inducing genes | Ion channels (e.g., KCNJ2), etc. | [23,32] |
Induction of cell cycle arrest | [23,32] | ||
In vivo transplantation | Transplantation into a normal or injured heart | [32,65] |
In Vivo Preclinical Studies | |||||||
Tissue Engineering Strategy | Biomaterial | Cell Source | Animal | Disease Model | Route of Administration | References | |
Cardiac patch | Fibrin | hiPSC-CM, pericytes | Rat | MI | Epicardium | [95] | |
Cardiac patch | Fibrin (IGF-loaded) | hiPSC-CM, -EC, and -SMC | Pig | MI | Epicardium | [97] | |
Muscle strip/EHM | Fibrin | hiPSC-CM, -EC | Guinea pig | Cryoinjury | Epicardium | [139,213] | |
EHM | Collagen | hESC-CM | Rat | MI/IR | Epicardium | [99] | |
EHM | Collagen | hESC-CM | Rat | Chronic MI/IR | Epicardium | [81] | |
In situ gelling system | Alginate | Acellular | Rat/Pig | MI | Intramyocardial/ Intracoronary IJ | [102,103] | |
In situ gelling system | Chitosan | Acellular | Rat | MI | Intramyocardial IJ | [106] | |
Nanofibers/EHM | PLGA | hiPSC-CM | Rat | MI | Epicardium | [110] | |
In situ gelling system | PNIPAAm-based | Acellular | Rat | Chronic MI | Intramyocardial IJ | [112] | |
In situ gelling system | Decellularized cardiac ECM hydrogel | Acellular | Pig | MI | Trans-endocardial IJ | [132] | |
3D-printing-based cardiac patch | ECM scaffold | hiPSC-CM, -EC, and -SMC | Mouse | MI | Epicardium | [79] | |
Cell sheet | Scaffold-free | hiPSC-CM | Pig | MI | Epicardium | [137] | |
Human Clinical Trials | |||||||
Trial Name (Trial Identifier) | Patient Population | Phase | Tissue Engineering Strategy | Biomaterial | Cell Source | Route of Administration | References |
PRESERVATION I (NCT01226563) | Acute MI; Congestive HF; ST elevation MI | Phase 1 completed | In situ gelling system | Alginate-based hydrogel | acellular | Intracoronary IJ | [221,222,223] |
AUGMENT-HF (NCT01311791) | Severe chronic HF (EF ≤ 35%) [57% ischemic and 43% nonischemic] | Phase 2 completed | In situ gelling system | Alginate-based hydrogel | acellular | Intramyocardial IJ | [224,225,226] |
Epicardial infarct repair using CorMatrixTM-ECM (NCT02887768) | Acute~subacute MI (within 6 weeks); CABG scheduled | Phase 1 completed | Cardiac patch | Porcine small intestine-derived ECM epicardial patch | acellular | Epicardium | [227,228,229] |
VentriGel in post-MI patients (NCT02305602) | Recent ~ late MI (60 days to 3 yrs); Ischemic HF (EF ≥ 25%; ≤45%) | Phase 1 completed | In situ gelling system | Porcine myocardium-derived ECM hydrogel | acellular | Trans-endocardial IJ | [230] |
ESCORT (NCT02057900) | Severe chronic ischemic HF (EF ≤3 5%) | Phase 1 completed | Cardiac patch | Fibrin patch | hESC-derived CD15+Isl1+ progenitors | Epicardium | [231,232] |
BioVAT-HF (NCT04396899) | Severe HF (EF ≤ 35%) | Phase 1 & 2 recruiting | EHM | Collagen type I hydrogel | hiPSC-CM, -stromal cell | Epicardium | |
Human (allogenic) iPSC-CM sheet for ischemic cardiomyopathy (NCT04696328) | Severe chronic ischemic HF (EF ≤ 35%) | Phase 1 recruiting | Cell sheet | Scaffold-free cell sheet | hiPSC-CM | Epicardium | [217,233] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Häneke, T.; Sahara, M. Progress in Bioengineering Strategies for Heart Regenerative Medicine. Int. J. Mol. Sci. 2022, 23, 3482. https://doi.org/10.3390/ijms23073482
Häneke T, Sahara M. Progress in Bioengineering Strategies for Heart Regenerative Medicine. International Journal of Molecular Sciences. 2022; 23(7):3482. https://doi.org/10.3390/ijms23073482
Chicago/Turabian StyleHäneke, Timm, and Makoto Sahara. 2022. "Progress in Bioengineering Strategies for Heart Regenerative Medicine" International Journal of Molecular Sciences 23, no. 7: 3482. https://doi.org/10.3390/ijms23073482
APA StyleHäneke, T., & Sahara, M. (2022). Progress in Bioengineering Strategies for Heart Regenerative Medicine. International Journal of Molecular Sciences, 23(7), 3482. https://doi.org/10.3390/ijms23073482