Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1588 word(s) 1588 2021-11-19 04:06:17 |
2 The format is correct. Meta information modification 1588 2021-11-29 05:24:19 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Wang, X.W. Cardiac Tissue Engineering for Treating Myocardial Infarction. Encyclopedia. Available online: https://encyclopedia.pub/entry/16479 (accessed on 03 May 2024).
Wang XW. Cardiac Tissue Engineering for Treating Myocardial Infarction. Encyclopedia. Available at: https://encyclopedia.pub/entry/16479. Accessed May 03, 2024.
Wang, Xiao Wei. "Cardiac Tissue Engineering for Treating Myocardial Infarction" Encyclopedia, https://encyclopedia.pub/entry/16479 (accessed May 03, 2024).
Wang, X.W. (2021, November 29). Cardiac Tissue Engineering for Treating Myocardial Infarction. In Encyclopedia. https://encyclopedia.pub/entry/16479
Wang, Xiao Wei. "Cardiac Tissue Engineering for Treating Myocardial Infarction." Encyclopedia. Web. 29 November, 2021.
Cardiac Tissue Engineering for Treating Myocardial Infarction
Edit

Ischemic heart disease (IHD) causes myocardial infarction (MI), which results in the death and loss of cardiomyocytes (CMs). Apoptosis, necrosis, and autophagy in CMs are the typical hallmarks of cardiac pathology in MI. Recent studies have shown that the combination of cell-based therapy and tissue engineering technology can improve stem cell engraftment and promote the therapeutic effects of the treatment for MI.

myocardial infarction stem cells

1. Types of Progenitor or Stem Cells Used for the Treatment of Myocardial Infarction

Cardiac tissue engineering aims to replace fibrotic scars by creating contractile and functional heart tissues. A wide variety of stem cells, their derivatives, and progenitor cells are currently being tested for these purposes. Stem cells can be divided into two types according to their sources: embryonic stem cells (ESCs) and adult progenitor/stem cells [1]. In terms of their differentiation capabilities, stem cells can be divided into four main types: totipotent stem cells, PSCs, multipotent stem cells, and unipotent stem cells [1]. The major progenitor/stem cells used in the field of cellular cardiomyoplasty are myoblasts [2][3][4], mesenchymal stem cells [5][6][7], ESCs [8][9], and iPSCs [10][11][12]. The advantages and disadvantages of each cell type are summarized in Table 1. Since ESCs and iPSCs can be differentiated into cardiovascular cells, including cardiac progenitor cells (CPCs), CMs, endothelial cells (ECs), and smooth muscle cells (SMCs), these stem cells may be used for cardiac tissue engineering and are being extensively investigated.
Table 1. Advantage and disadvantages of four types of cells used for cellular cardiomyoplasty.
Cell Types Advantages Disadvantages
Skeletal myoblasts
  1. Easy to obtain
  2. Easy to expand to get large number of cells in vitro
  3. Low ethical concerns
  4. No risk of tumorigenicity
  1. Risk of inducing ventricular arrhythmias
  2. Failure to transdifferentiate into functional cardiomyocytes
Mesenchymal stem cells
  1. Easy to obtain
  2. Can be selected by defined cell surface marker
  3. Low ethical concerns
  1. Limited cell quantity
  2. Limited differentiation potential
  3. Cells can only differentiate into cardiomyocyte-like cells
Embryonic stem cells
  1. Pluripotent stemness
  2. Well characterized cell lines
  3. Theoretically, they can be differentiated into all somatic cells found in the human body
  1. Genetically unstable
  2. Risk of tumorigenicity
  3. Allogenic transplantation induces immune rejection
  4. Ethical issues
Induced pluripotent stem cells
  1. Pluripotent stemness
  2. Autologous transplantation avoids immune rejection
  3. No ethical concern
  4. Theoretically, they can differentiate into all somatic cells found in the human body
  5. Disease-specific cell lines for disease modeling
  1. Low induction efficiency
  2. Genetically unstable
  3. Risk of tumorigenicity
  4. Disease-specific cell lines are not suitable for autologous transplantation
Mouse embryonic stem cells (mESCs) were established by two research teams led by Martin Evans and Matthew Kaufman in 1981 [1]. On the other hand, human ESCs (hESCs) were established by James Thomson in 1998 [1]. Mouse iPSCs (miPSCs) were established by Yamanak in 2006 using octamer binding transcription factor 3/4, sex determining region y-box 2, the cellular-myelocytomatosis viral oncogene, and kruppel-like factor 4 transcription factors from mouse fibroblasts [1]. The following year, Yamanak and James Thomson independently and simultaneously reported the establishment of hiPSCs from human dermal fibroblasts [1]. Generally, miPSCs or hiPSCs are almost identical to their counterpart, mESCs or hESCs.
PSCs are classified into two distinct states: naïve and primed pluripotent states. Naïve PSCs include mESCs and miPSCs, while primed PSCs include mouse epiblast stem cells (mEpiSCs), hESCs, and hiPSCs [13]. Although both naïve and primed PSCs are capable of self-renewal and can differentiate into the three germ layers in vitro and in vivo, only naïve PSCs can generate germline-competent chimeras in vivo [14].
Recent studies demonstrate that hiPSCs are a powerful tool for modeling diseases and treating various human diseases, including cardiovascular diseases, degenerative neuronal diseases, and diabetes, due to their unique advantage (Table 1). Like hESCs, hiPSCs have self-renewal capabilities and differentiate into all somatic cells found in the body regardless of their tissue of origin. hiSPCs are therefore being extensively explored to develop functional tissues for the treatment of myocardial injury.

2. Application of Tissue Engineering in PSC Reprogramming, Differentiation, and Treatment of Myocardial Infarction

Biomaterial scaffolds, which mimic the natural extracellular matrix (ECM) environment, have been extensively investigated for the engineering of cardiac tissues for PSC reprogramming, differentiation, and treatment of MI.

2.1. Tissue Engineering for the Regulation of PSC Reprogramming

Some biomaterials have been shown to improve reprogramming efficiency by changing epigenetic barriers. Downing et al. showed that the microgroove substrates increased the reprogramming efficiency of fibroblasts into iPSCs [15]. Notably, micro-grooved surfaces increased histone H3 acetylation and methylation, overcoming epigenetic barriers in reprogramming [15]. A study using tumor-initiating stem-like cells showed that a matrix made of fibrin gel promoted H3K9 demethylation and Sox2 expression [16], both of which are known to be involved in iPSC reprogramming [17][18].
An engineering technique, such as the clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 system is sought as a preferred method for the reprogramming and genetic modification of PSCs [19][20]. CRISPR/Cas9-based gene activator (CRISPRa), which has a high multiplexing capacity and direct targeting of endogenous loci, has also been used for cellular reprogramming [21]. Weltner et al. showed that using CRISPRa reduces the reprogramming efficiency of human dermal fibroblasts [21]. Moreover, Howden et al. developed a protocol that combined the CRISPR/Cas9 system and the enhanced episomal-based reprogramming to simultaneously generate gene-edited and passage-matched unmodified iPSC lines through the electroporation of human fibroblasts [22]. Compared to human fibroblasts, erythroblasts are later found to be a better cell candidate for simultaneous reprogramming and gene-editing using CRISPR/Cas9 [23]. These studies suggest that CRISPR/Cas9 technique may be a better option for iPSC reprogramming and gene editing.

2.2. Tissue Engineering for the Differentiation of PSCs into Cardiovascular Cells

Recent advances in tissue engineering allow for the designing of cellular structures or the incorporation of molecules to control the mechanical force and the release of certain factors that affect the differentiation of PSCs. Poh et al. developed a method to generate germ layers from a single line of mESCs [24]. In this study, an embryoid colony formed from a single mESC in 3D matrix, which was then cultured in 1 kPa collagen-1-coated 2D substrates. This resulted in the self-organization of the ectoderm, mesoderm, and endoderm layers in vitro.
When hiPSC-ECs were cultured in peptide-functionalized poly(ethylene glycol) (PEG) hydrogels, polygonal vascular networks were formed [25]. Vascular networks with lumens were stable for at least 14 days. A microcarrier (MC) suspension agitated platform has been shown to differentiate hESCs and hiPSCs into CMs [26]. Agitation results in homogenous and hydrodynamic shear stress, therefore inducing CM differentiation. The resulting yield is 38.3% and 39.3% higher for Troponin T and myosin heavy chain, respectively, than static culture control. Electrospun polylactide-glycolic acid/collagen (PLGA/Col) scaffolds have been used to differentiate ESCs to CMs [27]. Better interaction and growth of differentiated ESCs were observed on the PLGA/Col scaffolds relative to PLGA-only scaffolds. On the other hand, a cryopreserved amniotic membrane has been shown to direct the differentiation of hiPSC-derived cardiac progenitor cells to CMs in the presence of cytokines [28]. Amniotic membranes increased the expression of cardiac transcription factors and myofibril proteins, accelerated the intracellular calcium transients, and enhanced the mitochondrial complexity formation in CMs. These studies suggest that some biomaterials can be designed to provide PSCs with essential mechanical and chemical cues to direct their differentiation.

2.3. Tissue Engineering Using PSC-Derived Cardiovascular Cells for the Treatment of MI

It has been established that poor cell engraftment rate is one of the primary factors limiting the effectiveness of cell transfer therapy for cardiac repair. The low engraftment rate can be improved by combining the technique with cardiac tissue engineering, in which cells are transplanted within a supporting matrix [29]. Biomaterials can provide mechanical support for the stem cells and supply nutrients and oxygen to encapsulated cells [30][31][28][32].
Cardiac tissue engineering combines cells and biological scaffolds to make heart grafts to replace the damaged heart tissue and restore or improve the heart structure and function. In addition to cellular cardiomyoplasty, heart valve reconstruction, and vessel graft manufacturing are also the main target areas of heart tissue engineering.
Common tissue engineering materials are made of either natural or synthetic biomaterials. Natural biomaterials include Matrigel [32][33][34], hyaluronic acid [35][36], gelatin [37][38][39], chitosan [40][41], alginate [42][43][44], collagen [32][45][46][47][48][49], fibrin [10][11][12][45][50][51][52][53], elastin [54], amniotic membrane [28], and spider silk proteins [55]. Synthetic biomaterials, on the other hand, include poly(ethylene glycol) (PEG) [56][57], polylactide-glycolic acid (PLGA) [58][59], polyacrylamide (PAA) [60], poly(N-isopropylacrylamide) (PNIPAAm) [61], polycaprolactone (PCL) [62], and polyurethane (PU) [63]. More recently, natural/synthetic hybrid hydrogels are being developed as an alternative biomaterial that combines natural and synthetic materials through covalent grafting or crosslinking [64][65].

2.4. 3D Printing in Cardiac Tissue Engineering Using PSC Derived Cardiovascular Cells

In recent years, the rapid development of 3D printing technology enabled the construction of hydrogels and myocardial patches into highly precise and repeatable nanoscale 3D structures using multiple cell types. Biomaterials that are used for the 3D printing of myocardial tissue include alginate [66][67], fibrin [67], collagen [68][69], gelatin [70][71][72], hyaluronic acid [71], hydroxypropyl chitin [73], thixotropic magnesium phosphate [74], gellan gum [72], and decellularized ECM scaffolds [75][76][77]. Maiullari et al. manufactured a vascularized heart tissue using human umbilical vein endothelial cell (HUVEC) and miPSC-CMs using alginate and PEG/fibrin hydrogel extruded through a microfluidic printing head [67]. This study successfully generated a functional patch made of miPSC-CMs that aligned along the fiber printing direction. Furthermore, it was also observed that a pre-formed vasculature in 3D heart tissue has the potential to anastomose with the host’s vessels rapidly to supply blood to the implanted sample [67].
Lee et al. described a 3D printing technique to build complex collagen scaffolds for engineering vessels, contractile cardiac ventricle models, trileaflet heart valves, and even neonatal-scale human hearts [68]. Collagen gelation was controlled by modulating the pH and the printing resolution (up to 10 μm). With this technique, cells were successfully embedded in the collagen scaffolds. This is the first study to demonstrate successful 3D printing of a neonatal-sized heart with validated functions.

References

  1. Zakrzewski, W.; Dobrzynski, M.; Szymonowicz, M.; Rybak, Z. Stem cells: Past, present, and future. Stem Cell Res. Ther. 2019, 10, 68.
  2. Ye, L.; Haider, H.; Tan, R.; Toh, W.; Law, P.K.; Tan, W.; Su, L.; Zhang, W.; Ge, R.; Zhang, Y.; et al. Transplantation of nanoparticle transfected skeletal myoblasts overexpressing vascular endothelial growth factor-165 for cardiac repair. Circulation 2007, 116, I113–I120.
  3. Ye, L.; Haider, H.; Tan, R.; Su, L.; Law, P.K.; Zhang, W.; Sim, E.K. Angiomyogenesis using liposome based vascular endothelial growth factor-165 transfection with skeletal myoblast for cardiac repair. Biomaterials 2008, 29, 2125–2137.
  4. Ye, L.; Haider, H.; Jiang, S.; Tan, R.S.; Ge, R.; Law, P.K.; Sim, E.K. Improved angiogenic response in pig heart following ischaemic injury using human skeletal myoblast simultaneously expressing VEGF165 and angiopoietin-1. Eur. J. Heart Fail. 2007, 9, 15–22.
  5. Sid-Otmane, C.; Perrault, L.P.; Ly, H.Q. Mesenchymal stem cell mediates cardiac repair through autocrine, paracrine and endocrine axes. J. Transl. Med. 2020, 18, 336.
  6. Ahmed, L.A.; Al-Massri, K.F. Directions for Enhancement of the Therapeutic Efficacy of Mesenchymal Stem Cells in Different Neurodegenerative and Cardiovascular Diseases: Current Status and Future Perspectives. Curr. Stem Cell Res. Ther. 2021, 16, 858–876.
  7. Ye, L.; Zhang, P.; Duval, S.; Su, L.; Xiong, Q.; Zhang, J. Thymosin beta4 increases the potency of transplanted mesenchymal stem cells for myocardial repair. Circulation 2013, 128, S32–S41.
  8. Xiong, Q.; Ye, L.; Zhang, P.; Lepley, M.; Swingen, C.; Zhang, L.; Kaufman, D.S.; Zhang, J. Bioenergetic and functional consequences of cellular therapy: Activation of endogenous cardiovascular progenitor cells. Circ. Res. 2012, 111, 455–468.
  9. Yap, L.; Wang, J.W.; Moreno-Moral, A.; Chong, L.Y.; Sun, Y.; Harmston, N.; Wang, X.; Chong, S.Y.; Vanezis, K.; Ohman, M.K.; et al. In Vivo Generation of Post-infarct Human Cardiac Muscle by Laminin-Promoted Cardiovascular Progenitors. Cell Rep. 2020, 31, 107714.
  10. 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.
  11. Tao, Z.; Loo, S.; Su, L.; Tan, S.; Tee, G.; Gan, S.U.; Zhang, J.; Chen, X.; Ye, L. Angiopoietin-1 enhanced myocyte mitosis, engraftment, and the reparability of hiPSC-CMs for treatment of myocardial infarction. Cardiovasc. Res. 2021, 117, 1578–1591.
  12. Tan, S.H.; Loo, S.J.; Gao, Y.; Tao, Z.H.; Su, L.P.; Wang, C.X.; Zhang, S.L.; Mu, Y.H.; Cui, Y.H.; Abdurrachim, D.; et al. Thymosin beta4 increases cardiac cell proliferation, cell engraftment, and the reparative potency of human induced-pluripotent stem cell-derived cardiomyocytes in a porcine model of acute myocardial infarction. Theranostics 2021, 11, 7879–7895.
  13. Weinberger, L.; Ayyash, M.; Novershtern, N.; Hanna, J.H. Dynamic stem cell states: Naive to primed pluripotency in rodents and humans. Nat. Rev. Mol. Cell Biol. 2016, 17, 155–169.
  14. Nichols, J.; Smith, A. Naive and primed pluripotent states. Cell Stem Cell 2009, 4, 487–492.
  15. Downing, T.L.; Soto, J.; Morez, C.; Houssin, T.; Fritz, A.; Yuan, F.; Chu, J.; Patel, S.; Schaffer, D.V.; Li, S. Biophysical regulation of epigenetic state and cell reprogramming. Nat. Mater. 2013, 12, 1154–1162.
  16. Tan, Y.; Tajik, A.; Chen, J.; Jia, Q.; Chowdhury, F.; Wang, L.; Chen, J.; Zhang, S.; Hong, Y.; Yi, H.; et al. Matrix softness regulates plasticity of tumour-repopulating cells via H3K9 demethylation and Sox2 expression. Nat. Commun. 2014, 5, 4619.
  17. Chen, J.; Liu, H.; Liu, J.; Qi, J.; Wei, B.; Yang, J.; Liang, H.; Chen, Y.; Chen, J.; Wu, Y.; et al. H3K9 methylation is a barrier during somatic cell reprogramming into iPSCs. Nat. Genet. 2013, 45, 34–42.
  18. Jaenisch, R.; Young, R. Stem cells, the molecular circuitry of pluripotency and nuclear reprogramming. Cell 2008, 132, 567–582.
  19. Kwart, D.; Paquet, D.; Teo, S.; Tessier-Lavigne, M. Precise and efficient scarless genome editing in stem cells using CORRECT. Nat. Protoc. 2017, 12, 329–354.
  20. Hockemeyer, D.; Jaenisch, R. Induced Pluripotent Stem Cells Meet Genome Editing. Cell Stem Cell 2016, 18, 573–586.
  21. Weltner, J.; Balboa, D.; Katayama, S.; Bespalov, M.; Krjutskov, K.; Jouhilahti, E.M.; Trokovic, R.; Kere, J.; Otonkoski, T. Human pluripotent reprogramming with CRISPR activators. Nat. Commun. 2018, 9, 2643.
  22. Howden, S.E.; Thomson, J.A.; Little, M.H. Simultaneous reprogramming and gene editing of human fibroblasts. Nat. Protoc. 2018, 13, 875–898.
  23. Melo, U.S.; de Souza Leite, F.; Costa, S.; Rosenberg, C.; Zatz, M. A fast method to reprogram and CRISPR/Cas9 gene editing from erythroblasts. Stem Cell Res. 2018, 31, 52–54.
  24. Poh, Y.C.; Chen, J.; Hong, Y.; Yi, H.; Zhang, S.; Chen, J.; Wu, D.C.; Wang, L.; Jia, Q.; Singh, R.; et al. Generation of organized germ layers from a single mouse embryonic stem cell. Nat. Commun. 2014, 5, 4000.
  25. Zanotelli, M.R.; Ardalani, H.; Zhang, J.; Hou, Z.; Nguyen, E.H.; Swanson, S.; Nguyen, B.K.; Bolin, J.; Elwell, A.; Bischel, L.L.; et al. Stable engineered vascular networks from human induced pluripotent stem cell-derived endothelial cells cultured in synthetic hydrogels. Acta Biomater. 2016, 35, 32–41.
  26. Ting, S.; Chen, A.; Reuveny, S.; Oh, S. An intermittent rocking platform for integrated expansion and differentiation of human pluripotent stem cells to cardiomyocytes in suspended microcarrier cultures. Stem Cell Res. 2014, 13, 202–213.
  27. Prabhakaran, M.P.; Mobarakeh, L.G.; Kai, D.; Karbalaie, K.; Nasr-Esfahani, M.H.; Ramakrishna, S. Differentiation of embryonic stem cells to cardiomyocytes on electrospun nanofibrous substrates. J. Biomed. Mater. Res. B Appl. Biomater. 2014, 102, 447–454.
  28. Parveen, S.; Singh, S.P.; Panicker, M.M.; Gupta, P.K. Amniotic membrane as novel scaffold for human iPSC-derived cardiomyogenesis. In Vitro Cell Dev. Biol. Anim. 2019, 55, 272–284.
  29. 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.
  30. 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.
  31. Nguyen, A.H.; Marsh, P.; Schmiess-Heine, L.; Burke, P.J.; Lee, A.; Lee, J.; Cao, H. Cardiac tissue engineering: State-of-the-art methods and outlook. J. Biol. Eng. 2019, 13, 57.
  32. 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.
  33. 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.
  34. Bakunts, K.; Gillum, N.; Karabekian, Z.; Sarvazyan, N. Formation of cardiac fibers in Matrigel matrix. Biotechniques 2008, 44, 341–348.
  35. 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.
  36. Borzacchiello, A.; Russo, L.; Malle, B.M.; Schwach-Abdellaoui, K.; Ambrosio, L. Hyaluronic Acid Based Hydrogels for Regenerative Medicine Applications. Biomed. Res. Int. 2015, 2015, 871218.
  37. 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.
  38. McCain, M.L.; Agarwal, A.; Nesmith, H.W.; Nesmith, A.P.; Parker, K.K. Micromolded gelatin hydrogels for extended culture of engineered cardiac tissues. Biomaterials 2014, 35, 5462–5471.
  39. Zhang, F.; Zhang, N.; Meng, H.X.; Liu, H.X.; Lu, Y.Q.; Liu, C.M.; Zhang, Z.M.; Qu, K.Y.; Huang, N.P. Easy Applied Gelatin-Based Hydrogel System for Long-Term Functional Cardiomyocyte Culture and Myocardium Formation. ACS Biomater. Sci. Eng. 2019, 5, 3022–3031.
  40. Ahmadi, F.; Oveisi, Z.; Samani, S.M.; Amoozgar, Z. Chitosan based hydrogels: Characteristics and pharmaceutical applications. Res. Pharm. Sci. 2015, 10, 1–16.
  41. Shariatinia, Z.; Jalali, A.M. Chitosan-based hydrogels: Preparation, properties and applications. Int. J. Biol. Macromol. 2018, 115, 194–220.
  42. 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.
  43. Santana, B.P.; Nedel, F.; Piva, E.; de Carvalho, R.V.; Demarco, F.F.; Carreno, N.L. Preparation, modification, and characterization of alginate hydrogel with nano-/microfibers: A new perspective for tissue engineering. Biomed. Res. Int. 2013, 2013, 307602.
  44. Liberski, A.; Latif, N.; Raynaud, C.; Bollensdorff, C.; Yacoub, M. Alginate for cardiac regeneration: From seaweed to clinical trials. Glob. Cardiol. Sci. Pract. 2016, 2016, e201604.
  45. Kaiser, N.J.; Kant, R.J.; Minor, A.J.; Coulombe, K.L.K. Optimizing Blended Collagen-Fibrin Hydrogels for Cardiac Tissue Engineering with Human iPSC-derived Cardiomyocytes. ACS Biomater. Sci. Eng. 2019, 5, 887–899.
  46. 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.
  47. 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.
  48. 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.
  49. Nakane, T.; Masumoto, H.; Tinney, J.P.; Yuan, F.; Kowalski, W.J.; Ye, F.; LeBlanc, A.J.; Sakata, R.; Yamashita, J.K.; Keller, B.B. Impact of Cell Composition and Geometry on Human Induced Pluripotent Stem Cells-Derived Engineered Cardiac Tissue. Sci. Rep. 2017, 7, 45641.
  50. 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.
  51. 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.
  52. Querdel, E.; Reinsch, M.; Castro, L.; Kose, D.; Bahr, 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.
  53. Pecha, S.; Yorgan, K.; Rohl, 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.
  54. Contessotto, P.; Orbanic, D.; Da Costa, M.; Jin, C.; Owens, P.; Chantepie, S.; Chinello, C.; Newell, J.; Magni, F.; Papy-Garcia, D.; et al. Elastin-like recombinamers-based hydrogel modulates post-ischemic remodeling in a non-transmural myocardial infarction in sheep. Sci. Transl. Med. 2021, 13.
  55. Esser, T.U.; Trossmann, V.T.; Lentz, S.; Engel, F.B.; Scheibel, T. Designing of spider silk proteins for human induced pluripotent stem cell-based cardiac tissue engineering. Mater. Today Biol. 2021, 11, 100114.
  56. 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.
  57. 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.
  58. 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.
  59. 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.
  60. Daliri, K.; Pfannkuche, K.; Garipcan, B. Effects of physicochemical properties of polyacrylamide (PAA) and (polydimethylsiloxane) PDMS on cardiac cell behavior. Soft Matter 2021, 17, 1156–1172.
  61. 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.
  62. Wanjare, M.; Hou, L.; Nakayama, K.H.; Kim, J.J.; Mezak, N.P.; Abilez, O.J.; Tzatzalos, E.; Wu, J.C.; Huang, N.F. Anisotropic microfibrous scaffolds enhance the organization and function of cardiomyocytes derived from induced pluripotent stem cells. Biomater. Sci. 2017, 5, 1567–1578.
  63. Chiono, V.; Mozetic, P.; Boffito, M.; Sartori, S.; Gioffredi, E.; Silvestri, A.; Rainer, A.; Giannitelli, S.M.; Trombetta, M.; Nurzynska, D.; et al. Polyurethane-based scaffolds for myocardial tissue engineering. Interface Focus 2014, 4, 20130045.
  64. Vasile, C.; Pamfil, D.; Stoleru, E.; Baican, M. New Developments in Medical Applications of Hybrid Hydrogels Containing Natural Polymers. Molecules 2020, 25, 1539.
  65. 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.
  66. Gaetani, R.; Doevendans, P.A.; Metz, C.H.; Alblas, J.; Messina, E.; Giacomello, A.; Sluijter, J.P. Cardiac tissue engineering using tissue printing technology and human cardiac progenitor cells. Biomaterials 2012, 33, 1782–1790.
  67. Maiullari, F.; Costantini, M.; Milan, M.; Pace, V.; Chirivi, 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.
  68. 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.
  69. Tytgat, L.; Dobos, A.; Markovic, M.; Van Damme, L.; Van Hoorick, J.; Bray, F.; Thienpont, H.; Ottevaere, H.; Dubruel, P.; Ovsianikov, A.; et al. High-Resolution 3D Bioprinting of Photo-Cross-linkable Recombinant Collagen to Serve Tissue Engineering Applications. Biomacromolecules 2020, 21, 3997–4007.
  70. AnilKumar, S.; Allen, S.C.; Tasnim, N.; Akter, T.; Park, S.; Kumar, A.; Chattopadhyay, M.; Ito, Y.; Suggs, L.J.; Joddar, B. The applicability of furfuryl-gelatin as a novel bioink for tissue engineering applications. J. Biomed. Mater. Res. B Appl. Biomater. 2019, 107, 314–323.
  71. Gaetani, R.; Feyen, D.A.; Verhage, V.; Slaats, R.; Messina, E.; Christman, K.L.; Giacomello, A.; Doevendans, P.A.; Sluijter, J.P. 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.
  72. Koivisto, J.T.; Gering, C.; Karvinen, J.; Maria Cherian, R.; Belay, B.; Hyttinen, J.; Aalto-Setala, K.; Kellomaki, M.; Parraga, J. Mechanically Biomimetic Gelatin-Gellan Gum Hydrogels for 3D Culture of Beating Human Cardiomyocytes. ACS Appl. Mater. Interfaces 2019, 11, 20589–20602.
  73. Li, Y.; Jiang, X.; Li, L.; Chen, Z.N.; Gao, G.; Yao, R.; Sun, W. 3D printing human induced pluripotent stem cells with novel hydroxypropyl chitin bioink: Scalable expansion and uniform aggregation. Biofabrication 2018, 10, 044101.
  74. Chen, Y.; Wang, Y.; Yang, Q.; Liao, Y.; Zhu, B.; Zhao, G.; Shen, R.; Lu, X.; Qu, S. A novel thixotropic magnesium phosphate-based bioink with excellent printability for application in 3D printing. J. Mater. Chem. B 2018, 6, 4502–4513.
  75. 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.
  76. Kc, P.; Hong, Y.; Zhang, G. Cardiac tissue-derived extracellular matrix scaffolds for myocardial repair: Advantages and challenges. Regen. Biomater. 2019, 6, 185–199.
  77. Basara, G.; Ozcebe, S.G.; Ellis, B.W.; Zorlutuna, P. Tunable Human Myocardium Derived Decellularized Extracellular Matrix for 3D Bioprinting and Cardiac Tissue Engineering. Gels 2021, 7, 70.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 433
Entry Collection: Biopharmaceuticals Technology
Revisions: 2 times (View History)
Update Date: 29 Nov 2021
1000/1000