Korean Circ J.  2021 Dec;51(12):943-960. 10.4070/kcj.2021.0291.

Biomaterials-based Approaches for Cardiac Regeneration

Affiliations
  • 1Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
  • 2Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA

Abstract

The limited ability of cardiomyocytes to proliferate is a major cause of mortality and morbidity in cardiovascular diseases. There exist therapies for cardiac regeneration that are cell-based as well as that involve bioactive molecules. However, delivery remains one of the major challenges impeding such therapies from having clinical impact. Recent advancements in biomaterials-based approaches for cardiac regeneration have shown promise in clinical trials and animal studies in improving cardiac function, promoting angiogenesis, and reducing adverse immune response. This review will focus on current clinical studies of three contemporary biomaterials-based approaches for cardiac regeneration (extracellular vesicles, injectable hydrogels, and cardiac patches), remaining challenges and shortcomings to be overcome, and future directions for the use of biomaterials to promote cardiac regeneration.

Keyword

Tissue engineering; Extracellular vesicles; Heart failure; Cardiovascular diseases

Figure

  • Figure 1 Biomaterials-based approaches for cardiac regeneration. Traditional therapies including cell-based therapies and bioactive molecules can be directly administered to the heart. The 3 biomaterials-based approaches discussed in this review for administering these traditional therapies as well as novel therapies are: extracellular vesicles, cardiac patches, and injectable hydrogels. While these therapies can be directly administered, extracellular vesicles can also be administered using injectable hydrogels or cardiac patches.

  • Figure 2 Extracellular vesicles for endogenous cardiac regeneration. Extracellular vesicles derived from many origins, including iPSCs, iPSC-CM, HSCs, MSCs, VECs, and hPF, have shown potential for cardiac regeneration therapy. Exosomes may include mRNA, proteins, miRNA, and other bioactive molecules that facilitate a range of effects including angiogenesis, reduction of infarct size, cell survival and proliferation, paracrine factor release, and modulation of immune response. Delivery of extracellular vesicles through hydrogels and cardiac patches can enable the tailored release of extracellular vesicles, while the injection of parent cells leads to variable release in vivo.hPF = human pericardial fluid; HSC = hematopoietic stem cell; iPSC = induced pluripotent stem cell; iPSC-CM = cardiomyocytes derived from induced pluripotent stem cells; miRNA = microRNA ; mRNA = messenger RNA; MSC = mesenchymal stem cell; VEC = vascular endothelial cell.

  • Figure 3 Overview of engineered cardiac tissue patches. Cardiac patches are laboratory-manufactured sheets generally consisted of a scaffolding base and embedded therapeutic molecules that can be applied to the heart to ‘patch’ damaged tissue and help improve cardiac function after injury. Patches come in unique designs and structures such as cell patterning, microneedles, auxetic design, and 4D Physiology Adaptable Patch (A). Vascularization in the patch construct is critical for function of incorporated cells. Several strategies including direct 3D printing, co-culture, bi-layer patches, and BMVs have been employed to achieve higher degree of angiogenesis (B).3D = 3-dimensional; BMV = biomimetic micro-vessel; EC = endothelial cell; ECM = extracellular matrix; hMSC = human mesenchymal stem cell.


Cited by  2 articles

Cardiovascular Regeneration via Stem Cells and Direct Reprogramming: A Review
Choon-Soo Lee, Joonoh Kim, Hyun-Jai Cho, Hyo-Soo Kim
Korean Circ J. 2022;52(5):341-353.    doi: 10.4070/kcj.2022.0005.

The Gut-Heart Axis: Updated Review for The Roles of Microbiome in Cardiovascular Health
Thi Van Anh Bui, Hyesoo Hwangbo, Yimin Lai, Seok Beom Hong, Yeon-Jik Choi, Hun-Jun Park, Kiwon Ban
Korean Circ J. 2023;53(8):499-518.    doi: 10.4070/kcj.2023.0048.


Reference

1. Virani SS, Alonso A, Aparicio HJ, et al. Heart disease and stroke statistics-2021 update: a report from the American Heart Association. Circulation. 2021; 143:e254–e743. PMID: 33501848.
2. He L, Zhou B. Cardiomyocyte proliferation: remove brakes and push accelerators. Cell Res. 2017; 27:959–960. PMID: 28707671.
Article
3. Ludhwani D, Abraham J, Kanmanthareddy A. StatPearls. Heart transplantation rejection [Internet]. Treasure Island (FL): StatPearls Publishing;2021. 03. 12. cited 2021 July 22. Available from: https://www.ncbi.nlm.nih.gov/books/NBK537057/.
4. Bar A, Cohen S. Inducing endogenous cardiac regeneration: can biomaterials connect the dots? Front Bioeng Biotechnol. 2020; 8:126. PMID: 32175315.
Article
5. Nussbaum J, Minami E, Laflamme MA, et al. Transplantation of undifferentiated murine embryonic stem cells in the heart: teratoma formation and immune response. FASEB J. 2007; 21:1345–1357. PMID: 17284483.
Article
6. Uto K, Tsui JH, DeForest CA, Kim DH. Dynamically tunable cell culture platforms for tissue engineering and mechanobiology. Prog Polym Sci. 2017; 65:53–82. PMID: 28522885.
Article
7. Williams MA, Mair DB, Lee W, Lee E, Kim DH. Engineering three-dimensional vascularized cardiac tissues. Tissue Eng Part B Rev. 2021; ten.teb.2020.0343.
Article
8. Bang C, Thum T. Exosomes: new players in cell-cell communication. Int J Biochem Cell Biol. 2012; 44:2060–2064. PMID: 22903023.
Article
9. Kwon S, Shin S, Do M, et al. Engineering approaches for effective therapeutic applications based on extracellular vesicles. J Control Release. 2021; 330:15–30. PMID: 33278480.
Article
10. Yadid M, Lind JU, Ardoña HA, et al. Endothelial extracellular vesicles contain protective proteins and rescue ischemia-reperfusion injury in a human heart-on-chip. Sci Transl Med. 2020; 12:eaax8005. PMID: 33055246.
Article
11. Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013; 200:373–383. PMID: 23420871.
Article
12. Chun C, Smith AS, Kim H, et al. Astrocyte-derived extracellular vesicles enhance the survival and electrophysiological function of human cortical neurons in vitro. Biomaterials. 2021; 271:120700. PMID: 33631652.
Article
13. Liu B, Lee BW, Nakanishi K, et al. Cardiac recovery via extended cell-free delivery of extracellular vesicles secreted by cardiomyocytes derived from induced pluripotent stem cells. Nat Biomed Eng. 2018; 2:293–303. PMID: 30271672.
Article
14. Mackie AR, Klyachko E, Thorne T, et al. Sonic hedgehog-modified human CD34+ cells preserve cardiac function after acute myocardial infarction. Circ Res. 2012; 111:312–321. PMID: 22581926.
Article
15. Beltrami C, Besnier M, Shantikumar S, et al. Human pericardial fluid contains exosomes enriched with cardiovascular-expressed microRNAs and promotes therapeutic angiogenesis. Mol Ther. 2017; 25:679–693. PMID: 28159509.
Article
16. Lai RC, Arslan F, Lee MM, et al. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res (Amst). 2010; 4:214–222.
Article
17. Antes TJ, Middleton RC, Luther KM, et al. Targeting extracellular vesicles to injured tissue using membrane cloaking and surface display. J Nanobiotechnology. 2018; 16:61. PMID: 30165851.
Article
18. Wang X, Chen Y, Zhao Z, et al. Engineered exosomes with ischemic myocardium-targeting peptide for targeted therapy in myocardial infarction. J Am Heart Assoc. 2018; 7:e008737. PMID: 30371236.
Article
19. Gallet R, Dawkins J, Valle J, et al. Exosomes secreted by cardiosphere-derived cells reduce scarring, attenuate adverse remodelling, and improve function in acute and chronic porcine myocardial infarction. Eur Heart J. 2017; 38:201–211. PMID: 28158410.
Article
20. Gao L, Wang L, Wei Y, et al. Exosomes secreted by hiPSC-derived cardiac cells improve recovery from myocardial infarction in swine. Sci Transl Med. 2020; 12:eaay1318. PMID: 32938792.
Article
21. Luo Q, Guo D, Liu G, Chen G, Hang M, Jin M. Exosomes from MiR-126-overexpressing Adscs are therapeutic in relieving acute myocardial ischaemic injury. Cell Physiol Biochem. 2017; 44:2105–2116. PMID: 29241208.
Article
22. Hirai K, Ousaka D, Fukushima Y, et al. Cardiosphere-derived exosomal microRNAs for myocardial repair in pediatric dilated cardiomyopathy. Sci Transl Med. 2020; 12:eabb3336. PMID: 33298561.
Article
23. Hamada T, Dubois JL, Bellamy V, et al. In vitro controlled release of extracellular vesicles for cardiac repair from poly(glycerol sebacate) acrylate-based polymers. Acta Biomater. 2020; 115:92–103. PMID: 32814141.
Article
24. Davis ME, Hsieh PC, Grodzinsky AJ, Lee RT. Custom design of the cardiac microenvironment with biomaterials. Circ Res. 2005; 97:8–15. PMID: 16002755.
Article
25. Huang K, Ozpinar EW, Su T, et al. An off-the-shelf artificial cardiac patch improves cardiac repair after myocardial infarction in rats and pigs. Sci Transl Med. 2020; 12:9683.
Article
26. Qian Z, Sharma D, Jia W, Radke D, Kamp T, Zhao F. Engineering stem cell cardiac patch with microvascular features representative of native myocardium. Theranostics. 2019; 9:2143–2157. PMID: 31149034.
Article
27. Shah M, Kc P, Zhang G. In vivo assessment of decellularized porcine myocardial slice as an acellular cardiac patch. ACS Appl Mater Interfaces. 2019; 11:23893–23900. PMID: 31188555.
Article
28. Chen J, Zhan Y, Wang Y, et al. Chitosan/silk fibroin modified nanofibrous patches with mesenchymal stem cells prevent heart remodeling post-myocardial infarction in rats. Acta Biomater. 2018; 80:154–168. PMID: 30218777.
Article
29. Pok S, Benavides OM, Hallal P, Jacot JG. Use of myocardial matrix in a chitosan-based full-thickness heart patch. Tissue Eng Part A. 2014; 20:1877–1887. PMID: 24433519.
Article
30. Kapnisi M, Mansfield C, Marijon C, et al. Auxetic cardiac patches with tunable mechanical and conductive properties toward treating myocardial infarction. Adv Funct Mater. 2018; 28:1800618. PMID: 29875619.
Article
31. Hosoyama K, Ahumada M, McTiernan CD, et al. Nanoengineered electroconductive collagen-based cardiac patch for infarcted myocardium repair. ACS Appl Mater Interfaces. 2018; 10:44668–44677. PMID: 30508481.
Article
32. Lakshmanan R, Kumaraswamy P, Krishnan UM, Sethuraman S. Engineering a growth factor embedded nanofiber matrix niche to promote vascularization for functional cardiac regeneration. Biomaterials. 2016; 97:176–195. PMID: 27177129.
Article
33. O’Neill HS, O’Sullivan J, Porteous N, et al. A collagen cardiac patch incorporating alginate microparticles permits the controlled release of hepatocyte growth factor and insulin-like growth factor-1 to enhance cardiac stem cell migration and proliferation. J Tissue Eng Regen Med. 2018; 12:e384–e394. PMID: 27943590.
Article
34. Serpooshan V, Zhao M, Metzler SA, et al. The effect of bioengineered acellular collagen patch on cardiac remodeling and ventricular function post myocardial infarction. Biomaterials. 2013; 34:9048–9055. PMID: 23992980.
Article
35. Cui H, Liu C, Esworthy T, et al. 4D physiologically adaptable cardiac patch: a 4-month in vivo study for the treatment of myocardial infarction. Sci Adv. 2020; 6:eabb5067. PMID: 32637623.
Article
36. Gao L, Kupfer ME, Jung JP, 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. PMID: 28069694.
Article
37. Wang QL, Wang HJ, Li ZH, Wang YL, Wu XP, Tan YZ. Mesenchymal stem cell-loaded cardiac patch promotes epicardial activation and repair of the infarcted myocardium. J Cell Mol Med. 2017; 21:1751–1766. PMID: 28244640.
Article
38. Abbasgholizadeh R, Islas JF, Navran S, Potaman VN, Schwartz RJ, Birla RK. A highly conductive 3D cardiac patch fabricated using cardiac myocytes reprogrammed from human adipogenic mesenchymal stem cells. Cardiovasc Eng Technol. 2020; 11:205–218. PMID: 31916039.
Article
39. Ye L, Chang YH, Xiong Q, 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. PMID: 25479750.
Article
40. Mehrotra S, de Melo BA, Hirano M, et al. Nonmulberry silk based ink for fabricating mechanically robust cardiac patches and endothelialized myocardium-on-a-chip application. Adv Funct Mater. 2020; 30:1907436. PMID: 33071707.
Article
41. Lutolf MP, Hubbell JA. Synthetic biomaterials as instructive extracellular microenvironments for morphogenesis in tissue engineering. Nat Biotechnol. 2005; 23:47–55. PMID: 15637621.
Article
42. Chen WL, Kan CD. Using cell-seeded electrospun patch for myocardial injury: in-vitro and in rat model. Annu Int Conf IEEE Eng Med Biol Soc. 2018; 2018:5338–5341. PMID: 30441542.
Article
43. Rai R, Tallawi M, Barbani N, et al. Biomimetic poly(glycerol sebacate) (PGS) membranes for cardiac patch application. Mater Sci Eng C. 2013; 33:3677–3687.
Article
44. Ravichandran R, Venugopal JR, Mukherjee S, Sundarrajan S, Ramakrishna S. Elastomeric core/shell nanofibrous cardiac patch as a biomimetic support for infarcted porcine myocardium. Tissue Eng Part A. 2015; 21:1288–1298. PMID: 25559869.
Article
45. Cristallini C, Vaccari G, Barbani N, et al. Cardioprotection of PLGA/gelatine cardiac patches functionalised with adenosine in a large animal model of ischaemia and reperfusion injury: a feasibility study. J Tissue Eng Regen Med. 2019; 13:1253–1264. PMID: 31050859.
Article
46. Bahrami S, Solouk A, Mirzadeh H, Seifalian AM. Electroconductive polyurethane/graphene nanocomposite for biomedical applications. Compos, Part B Eng. 2019; 168:421–431.
Article
47. Chung HJ, Kim JT, Kim HJ, et al. Epicardial delivery of VEGF and cardiac stem cells guided by 3-dimensional PLLA mat enhancing cardiac regeneration and angiogenesis in acute myocardial infarction. J Control Release. 2015; 205:218–230. PMID: 25681051.
Article
48. Spadaccio C, Nappi F, De Marco F, et al. Implantation of a poly-L-lactide GCSF-functionalized scaffold in a model of chronic myocardial infarction. J Cardiovasc Transl Res. 2017; 10:47–65. PMID: 28116550.
Article
49. Pushp P, Bhaskar R, Kelkar S, Sharma N, Pathak D, Gupta MK. Plasticized poly(vinylalcohol) and poly(vinylpyrrolidone) based patches with tunable mechanical properties for cardiac tissue engineering applications. Biotechnol Bioeng. 2021; 118:2312–2325. PMID: 33675237.
Article
50. Su T, Huang K, Daniele MA, et al. Cardiac stem cell patch integrated with microengineered blood vessels promotes cardiomyocyte proliferation and neovascularization after acute myocardial infarction. ACS Appl Mater Interfaces. 2018; 10:33088–33096. PMID: 30188113.
Article
51. Tang J, Wang J, Huang K, et al. Cardiac cell-integrated microneedle patch for treating myocardial infarction. Sci Adv. 2018; 4:eaat9365. PMID: 30498778.
Article
52. Park BW, Jung SH, Das S, et al. In vivo priming of human mesenchymal stem cells with hepatocyte growth factor-engineered mesenchymal stem cells promotes therapeutic potential for cardiac repair. Sci Adv. 2020; 6:eaay6994. PMID: 32284967.
Article
53. Wang Q, Yang H, Bai A, et al. Functional engineered human cardiac patches prepared from nature's platform improve heart function after acute myocardial infarction. Biomaterials. 2016; 105:52–65. PMID: 27509303.
Article
54. Weinberger F, Breckwoldt K, Pecha S, et al. Cardiac repair in guinea pigs with human engineered heart tissue from induced pluripotent stem cells. Sci Transl Med. 2016; 8:363ra148.
Article
55. Gao L, Gregorich ZR, Zhu W, 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. PMID: 29233823.
Article
56. Schaefer JA, Guzman PA, Riemenschneider SB, Kamp TJ, Tranquillo RT. A cardiac patch from aligned microvessel and cardiomyocyte patches. J Tissue Eng Regen Med. 2018; 12:546–556. PMID: 28875579.
Article
57. Noor N, Shapira A, Edri R, Gal I, Wertheim L, Dvir T. 3D printing of personalized thick and perfusable cardiac patches and hearts. Adv Sci (Weinh). 2019; 6:1900344. PMID: 31179230.
Article
58. Kim DH, Lipke EA, Kim P, et al. Nanoscale cues regulate the structure and function of macroscopic cardiac tissue constructs. Proc Natl Acad Sci U S A. 2010; 107:565–570. PMID: 20018748.
Article
59. Kshitiz AJ, Afzal J, Kim SY, Kim DH. A nanotopography approach for studying the structure-function relationships of cells and tissues. Cell Adhes Migr. 2015; 9:300–307.
Article
60. Mengsteab PY, Uto K, Smith AS, et al. Spatiotemporal control of cardiac anisotropy using dynamic nanotopographic cues. Biomaterials. 2016; 86:1–10. PMID: 26874887.
Article
61. Carson D, Hnilova M, Yang X, et al. Nanotopography-Induced Structural Anisotropy and Sarcomere Development in Human Cardiomyocytes Derived from Induced Pluripotent Stem Cells. ACS Appl Mater Interfaces. 2016; 8:21923–21932. PMID: 26866596.
Article
62. Tsui JH, Janebodin K, Ieronimakis N, et al. Harnessing sphingosine-1 phosphate signaling and nanotopographical cuesto regulate skeletal muscle maturation and vascularization. ACS Nano. 2017; 11:11954–11968. PMID: 29156133.
63. Kim DH, Kshitiz , Smith RR, et al. Nanopatterned cardiac cell patches promote stem cell niche formation and myocardial regeneration. Integr Biol. 2012; 4:1019–1033.
Article
64. Lin YD, Ko MC, Wu ST, et al. A nanopatterned cell-seeded cardiac patch prevents electro-uncoupling and improves the therapeutic efficacy of cardiac repair. Biomater Sci. 2014; 2:567–580. PMID: 26827729.
Article
65. Macadangdang J, Lee HJ, Carson D, et al. Capillary force lithography for cardiac tissue engineering. J Vis Exp. 2014; e50039.
Article
66. Kim P, Yuan A, Nam KH, Jiao A, Kim DH. Fabrication of poly(ethylene glycol): gelatin methacrylate composite nanostructures with tunable stiffness and degradation for vascular tissue engineering. Biofabrication. 2014; 6:024112. PMID: 24717683.
Article
67. Uto K, Aoyagi T, Kim DH, Ebara M. Free-standing nanopatterned poly(ε-caprolactone) thin films as a multifunctional scaffold. IEEE Trans NanoTechnol. 2018; 17:389–392.
Article
68. Penland N, Choi E, Perla M, Park J, Kim DH. Facile fabrication of tissue-engineered constructs using nanopatterned cell sheets and magnetic levitation. Nanotechnology. 2017; 28:075103. PMID: 28028248.
Article
69. Jiao A, Trosper NE, Yang HS, et al. Thermoresponsive nanofabricated substratum for the engineering of three-dimensional tissues with layer-by-layer architectural control. ACS Nano. 2014; 8:4430–4439. PMID: 24628277.
Article
70. Williams NP, Rhodehamel M, Yan C, et al. Engineering anisotropic 3D tubular tissues with flexible thermoresponsive nanofabricated substrates. Biomaterials. 2020; 240:119856. PMID: 32105818.
Article
71. Malki M, Fleischer S, Shapira A, Dvir T. Gold nanorod-based engineered cardiac patch for suture-free engraftment by near IR. Nano Lett. 2018; 18:4069–4073. PMID: 29406721.
Article
72. Smith AS, Yoo H, Yi H, et al. Micro- and nano-patterned conductive graphene-PEG hybrid scaffolds for cardiac tissue engineering. Chem Commun (Camb). 2017; 53:7412–7415. PMID: 28634611.
Article
73. Tsui JH, Ostrovsky-Snider NA, Yama DMP, et al. Conductive silk-polypyrrole composite scaffolds with bioinspired nanotopographic cues for cardiac tissue engineering. J Mater Chem B. 2018; 6:7185–7196. PMID: 31448124.
Article
74. Choi JS, Smith AS, Williams NP, et al. Nanopatterned Nafion microelectrode arrays for in vitro cardiac electrophysiology. Adv Funct Mater. 2020; 30:1910660. PMID: 33244297.
Article
75. Smith AS, Choi E, Gray K, et al. NanoMEA: a tool for high-throughput, electrophysiological phenotyping of patterned excitable cells. Nano Lett. 2020; 20:1561–1570. PMID: 31845810.
76. Choi JS, Lee HJ, Rajaraman S, Kim DH. Recent advances in three-dimensional microelectrode array technologies for in vitro and in vivo cardiac and neuronal interfaces. Biosens Bioelectron. 2021; 171:112687. PMID: 33059168.
Article
77. Peña B, Laughter M, Jett S, et al. Injectable hydrogels for cardiac tissue engineering. Macromol Biosci. 2018; 18:e1800079. PMID: 29733514.
Article
78. Pati F, Jang J, Ha DH, et al. Printing three-dimensional tissue analogues with decellularized extracellular matrix bioink. Nat Commun. 2014; 5:3935. PMID: 24887553.
Article
79. Li Z, Guan J. Hydrogels for cardiac tissue engineering. Polymers (Basel). 2011; 3:740–761.
Article
80. Shin YJ, Shafranek RT, Tsui JH, Walcott J, Nelson A, Kim DH. 3D bioprinting of mechanically tuned bioinks derived from cardiac decellularized extracellular matrix. Acta Biomater. 2021; 119:75–88. PMID: 33166713.
Article
81. Mandrycky C, Wang Z, Kim K, Kim DH 3rd. 3D bioprinting for engineering complex tissues. Biotechnol Adv. 2016; 34:422–434. PMID: 26724184.
Article
82. Das S, Jang J. 3D bioprinting and decellularized ECM-based biomaterials for in vitro CV tissue engineering. J 3D Printing Med. 2018; 2:69–87.
83. Anker SD, Coats AJ, Cristian G, 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. PMID: 26082085.
Article
84. Lee LC, Wall ST, Klepach D, et al. 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. PMID: 23394895.
Article
85. Rao SV, Zeymer U, Douglas PS, et al. Bioabsorbable intracoronary matrix for prevention of ventricular remodeling after myocardial infarction. J Am Coll Cardiol. 2016; 68:715–723. PMID: 27515331.
Article
86. Traverse JH, Henry TD, Dib N, et al. 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. PMID: 31709316.
Article
87. Tous E, Purcell B, Ifkovits JL, Burdick JA. Injectable acellular hydrogels for cardiac repair. J Cardiovasc Transl Res. 2011; 4:528–542. PMID: 21710332.
Article
88. Wang H, Zhou J, Liu Z, Wang C. Injectable cardiac tissue engineering for the treatment of myocardial infarction. J Cell Mol Med. 2010; 14:1044–1055. PMID: 20193036.
Article
89. Contessotto P, Orbanić D, Da Costa M, et al. Elastin-like recombinamers-based hydrogel modulates post-ischemic remodeling in a non-transmural myocardial infarction in sheep. Sci Transl Med. 2021; 13:eaaz5380. PMID: 33597263.
Article
90. Chachques JC, Trainini JC, Lago N, Cortes-Morichetti M, Schussler O, Carpentier A. Myocardial Assistance by Grafting a New Bioartificial Upgraded Myocardium (MAGNUM trial): clinical feasibility study. Ann Thorac Surg. 2008; 85:901–908. PMID: 18291168.
Article
91. Hoeeg C, Dolatshahi-Pirouz A, Follin B. Injectable hydrogels for improving cardiac cell therapy-in vivo evidence and translational challenges. Gels. 2021; 7:7. PMID: 33499287.
Article
92. Tsui JH, Leonard A, Camp ND, et al. Tunable electroconductive decellularized extracellular matrix hydrogels for engineering human cardiac microphysiological systems. Biomaterials. 2021; 272:120764. PMID: 33798964.
Article
93. Liao X, Yang X, Deng H, et al. Injectable hydrogel-based nanocomposites for cardiovascular diseases. Front Bioeng Biotechnol. 2020; 8:251. PMID: 32296694.
Article
94. Deng C, Zhang P, Vulesevic B, et al. A collagen–chitosan hydrogel for endothelial differentiation and angiogenesis. Tissue Eng Part A. 2010; 16:3099–3109. PMID: 20586613.
Article
95. Song Y, Zhang C, Zhang J, et al. An injectable silk sericin hydrogel promotes cardiac functional recovery after ischemic myocardial infarction. Acta Biomater. 2016; 41:210–223. PMID: 27262742.
Article
96. Wang H, Liu Z, Li D, et al. Injectable biodegradable hydrogels for embryonic stem cell transplantation: improved cardiac remodelling and function of myocardial infarction. J Cell Mol Med. 2012; 16:1310–1320. PMID: 21838774.
Article
97. Leor J, Tuvia S, Guetta V, 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. PMID: 19729119.
Article
98. Kishore R, Khan M. More than tiny sacks: stem cell exosomes as cell-free modality for cardiac repair. Circ Res. 2016; 118:330–343. PMID: 26838317.
99. Hasan A, Khattab A, Islam MA, et al. Injectable hydrogels for cardiac tissue repair after myocardial infarction. Adv Sci (Weinh). 2015; 2:1500122. PMID: 27668147.
Article
100. Chaterji S, Ahn EH, Kim DH. CRISPR genome engineering for human pluripotent stem cell research. Theranostics. 2017; 7:4445–4469. PMID: 29158838.
Article
Full Text Links
  • KCJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr