Review Article

Potential Applications of Induced Pluripotent Stem Cells for Cardiovascular Diseases

Author(s): Xiaotong Wang, Zhenbo Han, Ying Yu, Zihang Xu, Benzhi Cai and Ye Yuan*

Volume 20, Issue 7, 2019

Page: [763 - 774] Pages: 12

DOI: 10.2174/1389450120666181211164147

Price: $65

Abstract

Owning the high incidence and disability rate in the past decades, to be expected, cardiovascular diseases (CVDs) have become one of the leading death causes worldwide. Currently, induced pluripotent stem cells (iPSCs), with the potential to form fresh myocardium and improve the functions of damaged hearts, have been studied widely in experimental CVD therapy. Moreover, iPSC-derived cardiomyocytes (CMs), as novel disease models, play a significant role in drug screening, drug safety assessment, along with the exploration of pathological mechanisms of diseases. Furthermore, a lot of studies have been carried out to clarify the biological basis of iPSCs and its derived cells in the treatment of CVDs. Their molecular mechanisms were associated with release of paracrine factors, regulation of miRNAs, mechanical support of new tissues, activation of specific pathways and specific enzymes, etc. In addition, a few small chemical molecules and suitable biological scaffolds play positive roles in enhancing the efficiency of iPSC transplantation. This article reviews the development and limitations of iPSCs in CVD therapy, and summarizes the latest research achievements regarding the application of iPSCs in CVDs.

Keywords: Induced pluripotent stem cells, cell therapy, cardiovascular disease, myocardium, pathological mechanisms, biological scaffolds.

Graphical Abstract
[1]
Ilic D, Devito L, Miere C, Codognotto S. Human embryonic and induced pluripotent stem cells in clinical trials. British medical bulletin 2015; 116: 19-27.
[2]
Yu J, Vodyanik MA, Smuga-Otto K, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science (New York, NY) 2007; 318(5858): 1917-20.
[3]
Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007; 131(5): 861-72.
[4]
Yang M, Liu Y, Hou W, et al. Mitomycin C-treated human-induced pluripotent stem cells as a safe delivery system of gold nanorods for targeted photothermal therapy of gastric cancer. Nanoscale 2017; 9(1): 334-40.
[5]
Saito H, Okita K, Chang AE, Ito F. Adoptive transfer of cd8+ t cells generated from induced pluripotent stem cells triggers regressions of large tumors along with immunological memory. Cancer Res 2016; 76(12): 3473-83.
[6]
Yao X, Salingova B, Dani C. Brown-like adipocyte progenitors derived from human ips cells: a new tool for anti-obesity drug discovery and cell-based therapy? Handbook of experimental pharmacology 2018.
[7]
Matsa E, Burridge PW, Yu KH, et al. Transcriptome profiling of patient-specific human ipsc-cardiomyocytes predicts individual drug safety and efficacy responses In Vitro. Cell Stem Cell 2016; 19(3): 311-25.
[8]
Yamanaka S, Takahashi K. Induction of pluripotent stem cells from mouse fibroblast cultures. Tanpakushitsu kakusan koso Protein Nucleic Acid Enzyme 2006; 51(15): 2346-51.
[9]
Hanna J, Wernig M, Markoulaki S, et al. Treatment of sickle cell anemia mouse model with iPS cells generated from autologous skin. Science (New York, NY) 2007; 318(5858): 1920-3.
[10]
Yang W, Mills JA, Sullivan S, et al. iPSC reprogramming from human peripheral blood using sendai virus mediated gene transfer. stembook. cambridge (ma): Harvard stem cell institute copyright: (c) 2012 Wenli Yang, Jason A. Mills, Spencer Sullivan, Ying Liu, Deborah L. French, and Paul Gadue.; 2008.
[11]
Cai J, DeLaForest A, Fisher J, et al. Protocol for directed differentiation of human pluripotent stem cells toward a hepatocyte fate. StemBook. Cambridge (MA): Harvard Stem Cell Institute Copyright: (c) 2012 Uri Ben-Davi and Nissim Benvenisty 2008.
[12]
Sommer AG, Rozelle SS, Sullivan S, et al. Generation of human induced pluripotent stem cells from peripheral blood using the STEMCCA lentiviral vector. J Vis Exp 2012; (68): 4327.
[13]
Heng BC, Richards M. Induced pluripotent stem cells (ipsc)--can direct delivery of transcription factors into the cytosol overcome the perils of permanent genetic modification? Minimally invasive therapy & allied technologies. MITAT 2008; 17(5): 326-7.
[14]
Tam PP. human stem cells can differentiate in post-implantation mouse embryos. Cell Stem Cell 2016; 18(1): 3-4.
[15]
Sundberg M, Bogetofte H, Lawson T, et al. Improved cell therapy protocols for Parkinson’s disease based on differentiation efficiency and safety of hESC-, hiPSC-, and non-human primate iPSC-derived dopaminergic neurons. Stem Cells (Dayton, Ohio) 2013; 31(8): 1548-62.
[16]
Hitomi H, Kasahara T, Katagiri N, et al. Human pluripotent stem cell-derived erythropoietin-producing cells ameliorate renal anemia in mice. Sci Transl Med 2017; 9(409)
[17]
Sakai-Takemura F, Narita A, Masuda S, et al. Premyogenic progenitors derived from human pluripotent stem cells expand in floating culture and differentiate into transplantable myogenic progenitors. Scientific Reports 2018; 8(1): 6555.
[18]
Kooreman NG, Kim Y, de Almeida PE, et al. autologous ipsc-based vaccines elicit anti-tumor responses in vivo. Cell Stem Cell 2018; 22(4): 501-13.e7.
[19]
Turner M, Leslie S, Martin NG, et al. Toward the development of a global induced pluripotent stem cell library. Cell Stem Cell 2013; 13(4): 382-4.
[20]
Stacey GN, Crook JM, Hei D, Ludwig T. Banking human induced pluripotent stem cells: lessons learned from embryonic stem cells? Cell Stem Cell 2013; 13(4): 385-8.
[21]
Mauritz C, Schwanke K, Reppel M, et al. Generation of functional murine cardiac myocytes from induced pluripotent stem cells. Circulation 2008; 118(5): 507-17.
[22]
Yan B, Abdelli LS, Singla DK. Transplanted induced pluripotent stem cells improve cardiac function and induce neovascularization in the infarcted hearts of db/db mice. Mol Pharmaceutics 2011; 8(5): 1602-10.
[23]
Taura D, Sone M, Homma K, et al. Induction and isolation of vascular cells from human induced pluripotent stem cells--brief report. Arterioscler Thromb Vasc Biol 2009; 29(7): 1100-3.
[24]
Narazaki G, Uosaki H, Teranishi M, et al. Directed and systematic differentiation of cardiovascular cells from mouse induced pluripotent stem cells. Circulation 2008; 118(5): 498-506.
[25]
Zhang F, Song G, Li X, et al. Transplantation of iPSc ameliorates neural remodeling and reduces ventricular arrhythmias in a post-infarcted swine model. J Cell Biochem 2014; 115(3): 531-9.
[26]
Mauritz C, Martens A, Rojas SV, et al. Induced pluripotent stem cell (iPSC)-derived Flk-1 progenitor cells engraft, differentiate, and improve heart function in a mouse model of acute myocardial infarction. Eur Heart J 2011; 32(21): 2634-41.
[27]
Miao Q, Shim W, Tee N, et al. iPSC-derived human mesenchymal stem cells improve myocardial strain of infarcted myocardium. J Cell Mol Med 2014; 18(8): 1644-54.
[28]
Gu M, Nguyen PK, Lee AS, et al. Microfluidic single-cell analysis shows that porcine induced pluripotent stem cell-derived endothelial cells improve myocardial function by paracrine activation. Circ Res 2012; 111(7): 882-93.
[29]
Wang Y, Huang W, Liang J, et al. Suicide gene-mediated sequencing ablation revealed the potential therapeutic mechanism of induced pluripotent stem cell-derived cardiovascular cell patch post-myocardial infarction. Antioxid Redox Signal 2014; 21(16): 2177-91.
[30]
Huang W, Dai B, Wen Z, et al. Molecular strategy to reduce in vivo collagen barrier promotes entry of NCX1 positive inducible pluripotent stem cells (iPSC(NCX(1)(+))) into ischemic (or injured) myocardium. PLoS One 2013; 8(8): e70023.
[31]
Dai B, Huang W, Xu M, et al. Reduced collagen deposition in infarcted myocardium facilitates induced pluripotent stem cell engraftment and angiomyogenesis for improvement of left ventricular function. J Am Coll Cardiol 2011; 58(20): 2118-27.
[32]
Higuchi T, Miyagawa S, Pearson JT, et al. Functional and electrical integration of induced pluripotent stem cell-derived cardiomyocytes in a myocardial infarction rat heart. Cell Transplant 2015; 24(12): 2479-89.
[33]
Chang D, Wen Z, Wang Y, et al. Ultrastructural features of ischemic tissue following application of a bio-membrane based progenitor cardiomyocyte patch for myocardial infarction repair. PLoS One 2014; 9(10): e107296.
[34]
Shiba Y, Gomibuchi T, Seto T, et al. Allogeneic transplantation of iPS cell-derived cardiomyocytes regenerates primate hearts. Nat 2016; 538(7625): 388-91.
[35]
Wang X, Chun YW, Zhong L, et al. A temperature-sensitive, self-adhesive hydrogel to deliver iPSC-derived cardiomyocytes for heart repair. Int J Cardiol 2015; 190: 177-80.
[36]
Francis MP, Breathwaite E, Bulysheva AA, et al. Human placenta hydrogel reduces scarring in a rat model of cardiac ischemia and enhances cardiomyocyte and stem cell cultures. Acta Biomater 2017; 52: 92-104.
[37]
Chow A, Stuckey DJ, Kidher E, et al. Human induced pluripotent stem cell-derived cardiomyocyte encapsulating bioactive hydrogels improve rat heart function post myocardial infarction. Stem Cell Reports 2017; 9(5): 1415-22.
[38]
Rojas SV, Martens A, Zweigerdt R, et al. Transplantation effectiveness of induced pluripotent stem cells is improved by a fibrinogen biomatrix in an experimental model of ischemic heart failure. Tissue Eng Part A 2015; 21(13-14): 1991-2000.
[39]
Liu T, Zhang R, Guo T, et al. Cardiotrophin-1 promotes cardiomyocyte differentiation from mouse induced pluripotent stem cells via JAK2/STAT3/Pim-1 signaling pathway. J Geriatr Cardiol 2015; 12(6): 591-9.
[40]
Kirby RJ, Divlianska DB, Whig K, et al. Discovery of novel small-molecule inducers of heme oxygenase-1 that protect human ipsc-derived cardiomyocytes from oxidative stress. J Pharmacol Exp Ther 2018; 364(1): 87-96.
[41]
Rojas SV, Kensah G, Rotaermel A, et al. Transplantation of purified iPSC-derived cardiomyocytes in myocardial infarction. PLoS One 2017; 12(5): e0173222.
[42]
Adamiak M, Cheng G, Bobis-Wozowicz S, et al. Induced pluripotent stem cell (ipsc)-derived extracellular vesicles are safer and more effective for cardiac repair than ipscs. Circulation Res 2018; 122(2): 296-309.
[43]
Tachibana A, Santoso MR, Mahmoudi M, et al. Paracrine effects of the pluripotent stem cell-derived cardiac myocytes salvage the injured myocardium. Circulation Res 2017; 121(6): e22-36.
[44]
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(6): 750-61.
[45]
Bar-Nur O, Russ HA, Efrat S, Benvenisty N. Epigenetic memory and preferential lineage-specific differentiation in induced pluripotent stem cells derived from human pancreatic islet beta cells. Cell Stem Cell 2011; 9(1): 17-23.
[46]
Kim K, Zhao R, Doi A, et al. Donor cell type can influence the epigenome and differentiation potential of human induced pluripotent stem cells. Nat Biotechnol 2011; 29(12): 1117-9.
[47]
Zhang L, Guo J, Zhang P, et al. Derivation and high engraftment of patient-specific cardiomyocyte sheet using induced pluripotent stem cells generated from adult cardiac fibroblast. Circulation Heart Fail 2015; 8(1): 156-66.
[48]
Ong SG, Huber BC, Lee WH, et al. Microfluidic single-cell analysis of transplanted human induced pluripotent stem cell-derived cardiomyocytes after acute myocardial infarction. Circulation 2015; 132(8): 762-71.
[49]
Ja KP, Miao Q, Zhen Tee NG, et al. iPSC-derived human cardiac progenitor cells improve ventricular remodelling via angiogenesis and interstitial networking of infarcted myocardium. J Cell Mol Med 2016; 20(2): 323-32.
[50]
Zhao X, Chen H, Xiao D, et al. Comparison of non-human primate versus human induced pluripotent stem cell-derived cardiomyocytes for treatment of myocardial infarction. Stem Cell Reports 2018; 10(2): 422-35.
[51]
Yu T, Miyagawa S, Miki K, et al. In vivo differentiation of induced pluripotent stem cell-derived cardiomyocytes. Circulation J 2013; 77(5): 1297-306.
[52]
Aggarwal P, Turner A, Matter A, et al. Cardiomyocytes in a cardiac hypertrophy model. PLoS One 2014; 9(9): e108051.
[53]
Zhang Y, Liang X, Liao S, et al. Potent paracrine effects of human induced pluripotent stem cell-derived mesenchymal stem cells attenuate doxorubicin-induced cardiomyopathy. Scientific Reports 2015; 5: 11235.
[54]
Bernardo ME, Fibbe WE. Mesenchymal stromal cells: Sensors and switchers of inflammation. Cell Stem Cell 2013; 13(4): 392-402.
[55]
Martens A, Rojas SV, Baraki H, et al. Substantial early loss of induced pluripotent stem cells following transplantation in myocardial infarction. Artif Organs 2014; 38(11): 978-84.
[56]
Schmidt C, Wiedmann F. Kallenberger, et al. Stretch-activated two-pore-domain (K2P) potassium channels in the heart: Focus on atrial fibrillation and heart failure. Prog Biophys Mol Biol 2017; 130(Pt B): 233-43.
[57]
Chai S, Wan X, Nassal DM, et al. Contribution of two-pore K(+) channels to cardiac ventricular action potential revealed using human iPSC-derived cardiomyocytes. Am J Physiol Heart Circ Physiol 2017; 312(6): H1144-h53.
[58]
Hou L, Coller J, Natu V, Hastie TJ, Huang NF. Combinatorial extracellular matrix microenvironments promote survival and phenotype of human induced pluripotent stem cell-derived endothelial cells in hypoxia. Acta Biomater 2016; 44: 188-99.
[59]
Masumoto H, Nakane T, Tinney JP, et al. The myocardial regenerative potential of three-dimensional engineered cardiac tissues composed of multiple human iPS cell-derived cardiovascular cell lineages. Scientific Reports 2016; 6: 29933.
[60]
Iseoka H, Miyagawa S, Fukushima S, et al. Pivotal role of non-cardiomyocytes in electromechanical and therapeutic potential of induced pluripotent stem cell-derived engineered cardiac tissue. Tissue Eng Part A 2018; 24(3-4): 287-300.
[61]
Rojas SV, Meier M, Zweigerdt R, et al. Multimodal imaging for in vivo evaluation of induced pluripotent stem cells in a murine model of heart failure. Artif Organs 2017; 41(2): 192-9.
[62]
Judge LM, Perez-Bermejo JA, Truong A, et al. A BAG3 chaperone complex maintains cardiomyocyte function during proteotoxic stress. JCI Insight 2017; 2(14)
[63]
Wu H, Lee J, Vincent LG, et al. Epigenetic regulation of phosphodiesterases 2a and 3a underlies compromised beta-adrenergic signaling in an ipsc model of dilated cardiomyopathy. Cell Stem Cell 2015; 17(1): 89-100.
[64]
Sayer G, Bhat G. The renin-angiotensin-aldosterone system and heart failure. Cardiol Clin 2014; 32(1): 21-32.
[65]
Jiang X, Sucharov J, Stauffer BL. Exosomes from pediatric dilated cardiomyopathy patients modulate a pathological response in cardiomyocytes. Am J Physiol Heart Circ Physiol 2017; 312(4): H818-h26.
[66]
Sucharov CC, Mariner PD, Nunley KR, et al. A beta1-adrenergic receptor CaM kinase II-dependent pathway mediates cardiac myocyte fetal gene induction. Am J Physiol Heart Circ Physiol 2006; 291(3): H1299-308.
[67]
Bang C, Batkai S, Dangwal S, et al. Cardiac fibroblast-derived microRNA passenger strand-enriched exosomes mediate cardiomyocyte hypertrophy. J Clin Invest 2014; 124(5): 2136-46.
[68]
Hashem SI, Perry CN, Bauer M, et al. Brief report: Oxidative stress mediates cardiomyocyte apoptosis in a human model of danon disease and heart failure. Stem Cells (Dayton, Ohio) 2015; 33(7): 2343-50.
[69]
Abarbanell AM, Wang Y, Herrmann JL, et al. Toll-like receptor 2 mediates mesenchymal stem cell-associated myocardial recovery and VEGF production following acute ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol 2010; 298(5): H1529-36.
[70]
Arslan F, Lai RC, Smeets MB, 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(3): 301-12.
[71]
Zhang H, Xiang M, Meng D, Sun N, Chen S. Inhibition of Myocardial Ischemia/Reperfusion Injury by Exosomes Secreted from Mesenchymal Stem Cells. Stem Cells Int 2016; 2016: 4328362.
[72]
Pennella S, Reggiani Bonetti L, Migaldi M, et al. Does stem cell therapy induce myocardial neoangiogenesis? Histological evaluation in an ischemia/reperfusion animal model. J Cardiovasc Med 2017; 18(4): 277-82.
[73]
Wang Y, Zhang L, Li Y, et al. Exosomes/microvesicles from induced pluripotent stem cells deliver cardioprotective miRNAs and prevent cardiomyocyte apoptosis in the ischemic myocardium. Int J Cardiol 2015; 192: 61-9.
[74]
Chen Z, Li Y, Yu H, et al. Isolation of extracellular vesicles from stem cells. Methods Mol Biol 2017; 1660: 389-94.
[75]
Zhu W, Gao L, Zhang J. Pluripotent stem cell derived cardiac cells for myocardial repair. J Vis Exp 2017; (120).
[76]
Wei W, Liu Y, Zhang Q, et al. Danshen-enhanced cardioprotective effect of cardioplegia on ischemia reperfusion injury in a human-induced pluripotent stem cell-derived cardiomyocytes model. Artif Organs 2017; 41(5): 452-60.
[77]
Devalla HD, Gelinas R, Aburawi EH, Beqqali A. TECRL, a new life-threatening inherited arrhythmia gene associated with overlapping clinical features of both LQTS and CPVT. EMBO Mol Med 2016; 8(12): 1390-408.
[78]
Chaudhari U, Nemade H, Wagh V, et al. Identification of genomic biomarkers for anthracycline-induced cardiotoxicity in human iPSC-derived cardiomyocytes: an in vitro repeated exposure toxicity approach for safety assessment. Arch Toxicol 2016; 90(11): 2763-77.
[79]
Necela BM, Axenfeld BC, Serie DJ, et al. The antineoplastic drug, trastuzumab, dysregulates metabolism in iPSC-derived cardiomyocytes. Clin Transl Med 2017; 6(1): 5.
[80]
Lee TI, Young RA. Transcriptional regulation and its misregulation in disease. Cell 2013; 152(6): 1237-51.
[81]
Anand P, Brown JD, Lin CY, et al. BET bromodomains mediate transcriptional pause release in heart failure. Cell 2013; 154(3): 569-82.
[82]
Spiltoir JI, Stratton MS, Cavasin MA, et al. BET acetyl-lysine binding proteins control pathological cardiac hypertrophy. J Mol Cell Cardiol 2013; 63: 175-9.
[83]
Duan Q, McMahon S. BET bromodomain inhibition suppresses innate inflammatory and profibrotic transcriptional networks in heart failure. Sci Transl Med 2017; 9(390)
[84]
Hershberger RE, Hedges DJ, Morales A. Dilated cardiomyopathy: the complexity of a diverse genetic architecture. Nat Rev Cardiol 2013; 10(9): 531-47.
[85]
Schmitt JP, Kamisago M, Asahi M, et al. Dilated cardiomyopathy and heart failure caused by a mutation in phospholamban. Sci 2003; 299(5611): 1410-3.
[86]
Haghighi K, Kolokathis F, Pater L, et al. Human phospholamban null results in lethal dilated cardiomyopathy revealing a critical difference between mouse and human. J Clin Invest 2003; 111(6): 869-76.
[87]
Karakikes I, Stillitano F, Nonnenmacher M, Tzimas C, Sanoudou D. Correction of human phospholamban R14del mutation associated with cardiomyopathy using targeted nucleases and combination therapy. Nat Commun 2015; 6: 6955.
[88]
Broughton KM, Li J, Sarmah E, et al. A myosin activator improves actin assembly and sarcomere function of human-induced pluripotent stem cell-derived cardiomyocytes with a troponin T point mutation. Am J Physiol Heart Circ Physiol 2016; 311(1): H107-17.
[89]
Jonsson MK, Vos MA, Mirams GR, et al. Application of human stem cell-derived cardiomyocytes in safety pharmacology requires caution beyond hERG. J Mol Cell Cardiol 2012; 52(5): 998-1008.
[90]
Doss MX, Di Diego JM, Goodrow RJ, et al. Maximum diastolic potential of human induced pluripotent stem cell-derived cardiomyocytes depends critically on I(Kr). PLoS One 2012; 7(7): e40288.
[91]
Bett GC, Kaplan AD, Lis A, et al. Electronic “expression” of the inward rectifier in cardiocytes derived from human-induced pluripotent stem cells. Heart Rhythm 2013; 10(12): 1903-10.
[92]
Huebsch N, Loskill P, Deveshwar N, et al. Miniaturized ips-cell-derived cardiac muscles for physiologically relevant drug response analyses. Sci Rep 2016; 6: 24726.
[93]
Okita K, Ichisaka T, Yamanaka S. Generation of germline-competent induced pluripotent stem cells. Nature 2007; 448(7151): 313-7.
[94]
Stadtfeld M, Nagaya M, Utikal J, Weir G, Hochedlinger K. Induced pluripotent stem cells generated without viral integration. Sci 2008; 322(5903): 945-9.
[95]
Okita K, Nakagawa M, Hyenjong H, Ichisaka T, Yamanaka S. Generation of mouse induced pluripotent stem cells without viral vectors. Science 2008; 322(5903): 949-53.
[96]
Kaji K, Norrby K, Paca A, et al. Virus-free induction of pluripotency and subsequent excision of reprogramming factors. Nature 2009; 458(7239): 771-5.
[97]
Woltjen K, Michael IP, Mohseni P, et al. piggyBac transposition reprograms fibroblasts to induced pluripotent stem cells. Nature 2009; 458(7239): 766-70.
[98]
Soldner F, Hockemeyer D, Beard C, et al. Parkinson’s disease patient-derived induced pluripotent stem cells free of viral reprogramming factors. Cell 2009; 136(5): 964-77.
[99]
Mackey LC, Annab LA, Yang J, et al. Epigenetic enzymes, age, and ancestry regulate the efficiency of human ipsc reprogramming. Stem Cells 2018; 36(11): 1697-708.
[100]
Tu J, Tian G, Cheung HH, Wei W, Lee TL. Gas5 is an essential lncRNA regulator for self-renewal and pluripotency of mouse embryonic stem cells and induced pluripotent stem cells. Stem Cell Res Ther 2018; 9(1): 71.
[101]
Medhekar SK, Shende VS, Chincholkar AB. Recent stem cell advances: cord blood and induced pluripotent stem cell for cardiac regeneration- a review. Int J Stem Cells 2016; 9(1): 21-30.
[102]
Kensah G, Roa Lara A, Dahlmann J, et al. Murine and human pluripotent stem cell-derived cardiac bodies form contractile myocardial tissue in vitro. Eur Heart J 2013; 34(15): 1134-46.
[103]
Xu XQ, Graichen R, Soo SY, et al. Chemically defined medium supporting cardiomyocyte differentiation of human embryonic stem cells. Differentiation 2008; 76(9): 958-70.
[104]
Tohyama S, Fujita J, Hishiki T, et al. Glutamine oxidation is indispensable for survival of human pluripotent stem cells. Cell Metab 2016; 23(4): 663-74.
[105]
El Harane N, Kervadec A, Bellamy V, et al. Acellular therapeutic approach for heart failure: In vitro production of extracellular vesicles from human cardiovascular progenitors. Eur Heart J 2018; 39(20): 1835-47.
[106]
Kawamura T, Miyagawa S, Fukushima S, et al. Cardiomyocytes derived from mhc-homozygous induced pluripotent stem cells exhibit reduced allogeneic immunogenicity in mhc-matched non-human primates. Stem Cell Reports 2016; 6(3): 312-20.
[107]
Sugita S, Iwasaki Y, Makabe K, et al. Successful transplantation of retinal pigment epithelial cells from mhc homozygote ipscs in mhc-matched models. Stem Cell Reports 2016; 7(4): 635-48.
[108]
Sugita S, Iwasaki Y, Makabe K, et al. Lack of t cell response to ipsc-derived retinal pigment epithelial cells from hla homozygous donors. Stem Cell Reports 2016; 7(4): 619-34.
[109]
Lo Sardo V, Ferguson W. Influence of donor age on induced pluripotent stem cells. Nat Biotechnol 2017; 35(1): 69-74.
[110]
Kang E, Wang X, Tippner-Hedges R, et al. Age-related accumulation of somatic mitochondrial dna mutations in adult-derived human ipscs. Cell Stem Cell 2016; 18(5): 625-36.
[111]
Chen W, Liu N, Zhang H, et al. Sirt6 promotes dna end joining in ipscs derived from old mice. Cell Reports 2017; 18(12): 2880-92.
[112]
Merkle FT, Ghosh S, Kamitaki N, et al. Human pluripotent stem cells recurrently acquire and expand dominant negative P53 mutations. Nature 2017; 545(7653): 229-33.
[113]
Cyranoski D. ‘Reprogrammed’ stem cells approved to mend human hearts for the first time. Nature 2018; 557(7707): 619-20.

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy