Induced pluripotent stem cells (iPSCs) can be generated by reprogramming of adult/somatic cells. The somatic cell reprogramming technology offers a promising strategy for patient-specific cardiac regenerative medicine, disease modeling, and drug discovery. iPSCs are an ideal potential option for an autologous cell source, as compared to other stem/progenitor cells, because they can be propagated indefinitely and are able to generate a large number of functional cardiovascular cells. However, there are concerns about the specificity, efficiency, immunogenicity, and safety of iPSCs which are major challenges in current translational studies. In order to bring iPSC technology closer to clinical use, fundamental changes in this technique are required to ensure that therapeutic progenies are functional and nontumorigenic. It is therefore critical to understand and investigate the biology, genetic, and epigenetic mechanisms of iPSCs generation and differentiation. In this spotlight paper the discovery, history, and relative mechanisms of iPSC generation are summarized. The current technological improvements and potential applications are highlighted along with the important challenges and perspectives. Finally, emerging technologies are presented in which improvements to iPSC generation and differentiation approaches might warrant further investigation, such as integration-free approaches, direct reprogramming, and the development of iPSC banking. 1. Introduction Myocardial infarction (MI) is an important manifestation of coronary artery disease (CAD) and major cause of death and disability worldwide. MI occurs when prolonged ischemia irreversibly destroys distal blood vessels and myocardium, causing apoptosis or cell death, eventually triggering cardiac remodeling or sudden death [1, 2]. Recurrent MI leads to chronic postinfarct heart failure in patients with a longer life span. Currently, traditional therapeutic approaches focus on limitation of the initial injury and secondary maladaptive complications in order to prevent the death of existing myocardium. Despite significant advances in medical treatments over the past decades, chronic heart failure remains as a leading cause of death [3, 4]. Indeed, heart transplantation is the only available viable therapeutic option for end-stage heart failure, but this option is limited by the paucity of matched donor tissue specimens and by the requirement for life-long treatment with immunosuppresive agents. Regenerative therapies offer great promise for patients with heart disease by using angiomyogenesis to create
References
[1]
K. Thygesen, J. S. Alpert, A. S. Jaffe, M. L. Simoons, B. R. Chaitman, and H. D. White, “Third universal definition of myocardial infarction,” Journal of the American College of Cardiology, vol. 60, no. 16, pp. 1581–1598, 2012.
[2]
B. Swynghedauw, “Molecular mechanisms of myocardial remodeling,” Physiological Reviews, vol. 79, no. 1, pp. 215–262, 1999.
[3]
R. A. Pedersen, V. Mascetti, and S. Mendjan, “Synthetic organs for regenerative medicine,” Cell Stem Cell, vol. 10, no. 6, pp. 646–647, 2012.
[4]
G. C. Gurtner, M. J. Callaghan, and M. T. Longaker, “Progress and potential for regenerative medicine,” Annual Review of Medicine, vol. 58, pp. 299–312, 2007.
[5]
J. A. Thomson, “Embryonic stem cell lines derived from human blastocysts,” Science, vol. 282, no. 5391, pp. 1145–1147, 1998.
[6]
M. J. Evans and M. H. Kaufman, “Establishment in culture of pluripotential cells from mouse embryos,” Nature, vol. 292, no. 5819, pp. 154–156, 1981.
[7]
M. Evans, “Discovering pluripotency: 30 years of mouse embryonic stem cells,” Nature Reviews Molecular Cell Biology, vol. 12, no. 10, pp. 680–686, 2011.
[8]
H. S. Bernstein, “Cardiac repair and restoration using human embryonic stem cells,” Regenerative Medicine, vol. 7, no. 5, pp. 697–712, 2012.
[9]
C. W. Siu, J. C. Moore, and R. A. Li, “Human embryonic stem cell-derived cardiomyocytes for heart therapies,” Cardiovascular and Hematological Disorders Drug Targets, vol. 7, no. 2, pp. 145–152, 2007.
[10]
S. Levenberg, J. S. Golub, M. Amit, J. Itskovitz-Eldor, and R. Langer, “Endothelial cells derived from human embryonic stem cells,” Proceedings of the National Academy of Sciences of the United States of America, vol. 99, no. 7, pp. 4391–4396, 2002.
[11]
M. Boiani and H. R. Sch?ler, “Regulatory networks in embryo-derived pluripotent stem cells,” Nature Reviews Molecular Cell Biology, vol. 6, no. 11, pp. 872–884, 2005.
[12]
K.-J. Won, Z. Xu, X. Zhang et al., “Global identification of transcriptional regulators of pluripotency and differentiation in embryonic stem cells,” Nucleic Acids Research, vol. 40, no. 17, pp. 8199–8209, 2012.
[13]
L. A. Boyer, I. L. Tong, M. F. Cole et al., “Core transcriptional regulatory circuitry in human embryonic stem cells,” Cell, vol. 122, no. 6, pp. 947–956, 2005.
[14]
I. Aksoy and L. W. Stanton, “Pluripotency-regulating networks provide basis for reprogramming,” Current Molecular Medicine, vol. 13, no. 5, pp. 695–706, 2013.
[15]
T. S. Tanaka, T. Kunath, W. L. Kimber et al., “Gene expression profiling of embryo-derived stem cells reveals candidate genes associated with pluripotency and lineage specificity,” Genome Research, vol. 12, no. 12, pp. 1921–1928, 2002.
[16]
H.-H. Ng and M. A. Surani, “The transcriptional and signalling networks of pluripotency,” Nature Cell Biology, vol. 13, no. 5, pp. 490–496, 2011.
[17]
X. Chen, H. Xu, P. Yuan et al., “Integration of external signaling pathways with the core transcriptional network in embryonic stem cells,” Cell, vol. 133, no. 6, pp. 1106–1117, 2008.
[18]
T. J. King and R. Briggs, “Changes in the nuclei of differentiating gastrula cells, as demonstrated by nuclear transplantation,” Proceedings of the National Academy of Sciences of the United States of America, vol. 41, no. 5, pp. 321–325, 1955.
[19]
R. Briggs and T. J. King, “Transplantation of living nuclei from blastula cells into enucleated frogs' eggs,” Proceedings of the National Academy of Sciences of the United States of America, vol. 38, no. 5, pp. 455–463, 1952.
[20]
S. Noggle, H.-L. Fung, A. Gore et al., “Human oocytes reprogram somatic cells to a pluripotent state,” Nature, vol. 478, no. 7367, pp. 70–75, 2011.
[21]
I. Hyun, “Moving human SCNT research forward ethically,” Cell Stem Cell, vol. 9, no. 4, pp. 295–297, 2011.
[22]
M. Stadtfeld and K. Hochedlinger, “Induced pluripotency: history, mechanisms, and applications,” Genes and Development, vol. 24, no. 20, pp. 2239–2263, 2010.
[23]
H. Sumer, J. Liu, P. Tat, C. Heffernan, K. L. Jones, and P. J. Verma, “Somatic cell nuclear transfer: pros and cons,” Journal of Stem Cells, vol. 4, no. 2, pp. 85–93, 2009.
[24]
K. Takahashi, K. Tanabe, M. Ohnuki et al., “Induction of pluripotent stem cells from adult human fibroblasts by defined factors,” Cell, vol. 131, no. 5, pp. 861–872, 2007.
[25]
K. Takahashi and S. Yamanaka, “Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors,” Cell, vol. 126, no. 4, pp. 663–676, 2006.
[26]
J. Yu, M. A. Vodyanik, K. Smuga-Otto et al., “Induced pluripotent stem cell lines derived from human somatic cells,” Science, vol. 318, no. 5858, pp. 1917–1920, 2007.
[27]
N. Maherali and K. Hochedlinger, “Guidelines and techniques for the generation of induced pluripotent stem cells,” Cell Stem Cell, vol. 3, no. 6, pp. 595–605, 2008.
[28]
A. Saunders, F. Faiola, and J. Wang, “Concise review: pursuing self-renewal and pluripotency with the stem cell factor Nanog,” Stem Cells, vol. 31, no. 7, pp. 1227–1236, 2013.
[29]
S. Eminli, A. Foudi, M. Stadtfeld et al., “Differentiation stage determines potential of hematopoietic cells for reprogramming into induced pluripotent stem cells,” Nature Genetics, vol. 41, no. 9, pp. 968–976, 2009.
[30]
N. Malik and M. S. Rao, “A review of the methods for human iPSC derivation,” Methods in Molecular Biology, vol. 997, pp. 23–33, 2013.
[31]
Y. Xu, X. Wei, M. Wang et al., “Proliferation rate of somatic cells affects reprogramming efficiency,” The Journal of Biological Chemistry, vol. 288, no. 14, pp. 9767–9778, 2013.
[32]
T. A. Baudino, C. McKay, H. Pendeville-Samain et al., “c-Myc is essential for vasculogenesis and angiogenesis during development and tumor progression,” Genes and Development, vol. 16, no. 19, pp. 2530–2543, 2002.
[33]
C. M. D'Cruz, E. J. Gunther, R. B. Boxer et al., “c-MYC induces mammary tumorigenesis by means of a preferred pathway involving spontaneous Kras2 mutations,” Nature Medicine, vol. 7, no. 2, pp. 235–239, 2001.
[34]
R. Blelloch, M. Venere, J. Yen, and M. Ramalho-Santos, “Generation of induced pluripotent stem cells in the absence of drug selection,” Cell Stem Cell, vol. 1, no. 3, pp. 245–247, 2007.
[35]
M. Nakagawa, M. Koyanagi, K. Tanabe et al., “Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts,” Nature Biotechnology, vol. 26, no. 1, pp. 101–106, 2008.
[36]
O. Habib, G. Habib, H. W. Choi et al., “An improved method for the derivation of high quality iPSCs in the absence of c-Myc,” Experimental Cell Research, vol. 319, no. 20, pp. 3190–3200, 2013.
[37]
S.-H. Chiou, B.-H. Jiang, Y.-L. Yu et al., “Poly(ADP-ribose) polymerase 1 regulates nuclear reprogramming and promotes iPSC generation without c-Myc,” Journal of Experimental Medicine, vol. 210, no. 1, pp. 85–98, 2013.
[38]
A. Martinez-Fernandez, T. J. Nelson, Y. Ikeda, and A. Terzic, “c-MYC independent nuclear reprogramming favors cardiogenic potential of induced pluripotent stem cells,” Journal of Cardiovascular Translational Research, vol. 3, no. 1, pp. 13–23, 2010.
[39]
A. Giorgetti, N. Montserrat, I. Rodriguez-Piza, C. Azqueta, A. Veiga, and J. C. Izpisúa Belmonte, “Generation of induced pluripotent stem cells from human cord blood cells with only two factors: Oct4 and Sox2,” Nature protocols, vol. 5, no. 4, pp. 811–820, 2010.
[40]
N. Montserrat, E. Nivet, I. Sancho-Martinez et al., “Reprogramming of human fibroblasts to pluripotency with lineage specifiers,” Cell Stem Cell, vol. 13, no. 3, pp. 341–350, 2013.
[41]
J. Shu, C. Wu, Y. Wu et al., “Induction of pluripotency in mouse somatic cells with lineage specifiers,” Cell, vol. 153, no. 5, pp. 963–975, 2013.
[42]
Y. Gao, J. Chen, K. Li et al., “Replacement of Oct4 by Tet1 during iPSC induction reveals an important role of DNA methylation and hydroxymethylation in reprogramming,” Cell Stem Cell, vol. 12, no. 4, pp. 453–469, 2013.
[43]
M. Wernig, C. J. Lengner, J. Hanna et al., “A drug-inducible transgenic system for direct reprogramming of multiple somatic cell types,” Nature Biotechnology, vol. 26, no. 8, pp. 916–924, 2008.
[44]
C. A. Sommer, M. Stadtfeld, G. J. Murphy, K. Hochedlinger, D. N. Kotton, and G. Mostoslavsky, “Induced pluripotent stem cell generation using a single lentiviral stem cell cassette,” Stem Cells, vol. 27, no. 3, pp. 543–549, 2009.
[45]
C.-W. Chang, Y.-S. Lai, K. M. Pawlik et al., “Polycistronic lentiviral vector for “hit and run” reprogramming of adult skin fibroblasts to induced pluripotent stem cells,” Stem Cells, vol. 27, no. 5, pp. 1042–1049, 2009.
[46]
R. Sprengel and M. T. Hasan, “Tetracycline-controlled genetic switches,” Handbook of experimental pharmacology, vol. 178, pp. 49–72, 2007.
[47]
T. Yamaguchi, S. Hamanaka, A. Kamiya et al., “Development of an all-in-one inducible lentiviral vector for gene specific analysis of reprogramming,” PLoS ONE, vol. 7, no. 7, Article ID e41007, 2012.
[48]
A. Nagy, “Secondary cell reprogramming systems: as years go by,” Current Opinion in Genetics and Development, vol. 23, no. 5, pp. 534–539, 2013.
[49]
N. Maherali, T. Ahfeldt, A. Rigamonti, J. Utikal, C. Cowan, and K. Hochedlinger, “A high-efficiency system for the generation and study of human induced pluripotent stem cells,” Cell Stem Cell, vol. 3, no. 3, pp. 340–345, 2008.
[50]
D. Hockemeyer, F. Soldner, E. G. Cook, Q. Gao, M. Mitalipova, and R. Jaenisch, “A drug-inducible system for direct reprogramming of human somatic cells to pluripotency,” Cell Stem Cell, vol. 3, no. 3, pp. 346–353, 2008.
[51]
T. Matsui, D. Leung, H. Miyashita et al., “Proviral silencing in embryonic stem cells requires the histone methyltransferase ESET,” Nature, vol. 464, no. 7290, pp. 927–931, 2010.
[52]
M. Stadtfeld, N. Maherali, D. T. Breault, and K. Hochedlinger, “Defining molecular cornerstones during fibroblast to iPS cell reprogramming in mouse,” Cell Stem Cell, vol. 2, no. 3, pp. 230–240, 2008.
[53]
R. Sridharan, J. Tchieu, M. J. Mason et al., “Role of the murine reprogramming factors in the induction of pluripotency,” Cell, vol. 136, no. 2, pp. 364–377, 2009.
[54]
J. Ellis, “Silencing and variegation of gammaretrovirus and lentivirus vectors,” Human Gene Therapy, vol. 16, no. 11, pp. 1241–1246, 2005.
[55]
T. Brambrink, R. Foreman, G. G. Welstead et al., “Sequential expression of pluripotency markers during direct reprogramming of mouse somatic cells,” Cell Stem Cell, vol. 2, no. 2, pp. 151–159, 2008.
[56]
T. P. Zwaka, “Reduce, reuse, reprogram,” Nature Methods, vol. 7, no. 1, pp. 39–40, 2010.
[57]
S. Markoulaki, J. Hanna, C. Beard et al., “Transgenic mice with defined combinations of drug-inducible reprogramming factors,” Nature Biotechnology, vol. 27, no. 2, pp. 169–171, 2009.
[58]
B. W. Carey, S. Markoulaki, C. Beard, J. Hanna, and R. Jaenisch, “Single-gene transgenic mouse strains for reprogramming adult somatic cells,” Nature Methods, vol. 7, no. 1, pp. 56–59, 2010.
[59]
M. Stadtfeld, N. Maherali, M. Borkent, and K. Hochedlinger, “A reprogrammable mouse strain from gene-targeted embryonic stem cells,” Nature Methods, vol. 7, no. 1, pp. 53–55, 2010.
[60]
M. Abad, L. Mosteiro, C. Pantoja et al., et al., “Reprogramming in vivo produces teratomas and iPS cells with totipotency features,” Nature, vol. 502, no. 7471, pp. 340–345, 2013.
[61]
A. Golipour, L. David, Y. Liu et al., “A late transition in somatic cell reprogramming requires regulators distinct from the pluripotency network,” Cell Stem Cell, vol. 11, no. 6, pp. 769–782, 2012.
[62]
A. S. Lee, C. Tang, M. S. Rao, I. L. Weissman, and J. C. Wu, “Tumorigenicity as a clinical hurdle for pluripotent stem cell therapies,” Nature Medicine, vol. 19, no. 8, pp. 998–1004, 2013.
[63]
S. G. Hong, C. E. Dunbar, and T. Winkler, “Assessing the risks of genotoxicity in the therapeutic development of induced pluripotent stem cells,” Molecular Therapy, vol. 21, no. 2, pp. 272–281, 2013.
[64]
M. Stadtfeld, M. Nagaya, J. Utikal, G. Weir, and K. Hochedlinger, “Induced pluripotent stem cells generated without viral integration,” Science, vol. 322, no. 5903, pp. 945–949, 2008.
[65]
K. Okita, M. Nakagawa, H. Hyenjong, T. Ichisaka, and S. Yamanaka, “Generation of mouse induced pluripotent stem cells without viral vectors,” Science, vol. 322, no. 5903, pp. 949–953, 2008.
[66]
N. Fusaki, H. Ban, A. Nishiyama, K. Saeki, and M. Hasegawa, “Efficient induction of transgene-free human pluripotent stem cells using a vector based on Sendai virus, an RNA virus that does not integrate into the host genome,” Proceedings of the Japan Academy B: Physical and Biological Sciences, vol. 85, no. 8, pp. 348–362, 2009.
[67]
T. Seki, S. Yuasa, and K. Fukuda, “Generation of induced pluripotent stem cells from a small amount of human peripheral blood using a combination of activated T cells and Sendai virus,” Nature Protocols, vol. 7, no. 4, pp. 718–728, 2012.
[68]
Y. Junying, H. Kejin, S.-O. Kim et al., “Human induced pluripotent stem cells free of vector and transgene sequences,” Science, vol. 324, no. 5928, pp. 797–801, 2009.
[69]
K. Hu and I. Slukvin, “Generation of transgene-free iPSC lines from human normal and neoplastic blood cells using episomal vectors,” Methods in Molecular Biology, vol. 997, pp. 163–176, 2013.
[70]
F. Jia, K. D. Wilson, N. Sun et al., “A nonviral minicircle vector for deriving human iPS cells,” Nature Methods, vol. 7, no. 3, pp. 197–199, 2010.
[71]
B. W. Carey, S. Markoulaki, J. Hanna et al., “Reprogramming of murine and human somatic cells using a single polycistronic vector,” Proceedings of the National Academy of Sciences of the United States of America, vol. 106, no. 1, pp. 157–162, 2009.
[72]
X. Qu, T. Liu, K. Song, X. Li, and D. Ge, “Induced pluripotent stem cells generated from human adipose-derived stem cells using a non-viral polycistronic plasmid in feeder-free conditions,” PLoS ONE, vol. 7, no. 10, Article ID e48161, 2012.
[73]
K. Woltjen, R. H?m?l?inen, M. Kibschull, M. Mileikovsky, and A. Nagy, “Transgene-free production of pluripotent stem cells using piggyBac transposons,” Methods in Molecular Biology, vol. 767, pp. 87–103, 2011.
[74]
Z. Ivics, M. A. Li, L. Mátés et al., “Transposon-mediated genome manipulation in vertebrates,” Nature Methods, vol. 6, no. 6, pp. 415–422, 2009.
[75]
M. Di Matteo, E. Belay, M. K. Chuah, and T. Vandendriessche, “Recent developments in transposon-mediated gene therapy,” Expert Opinion on Biological Therapy, vol. 12, no. 7, pp. 841–858, 2012.
[76]
K. Yusa, R. Rad, J. Takeda, and A. Bradley, “Generation of transgene-free induced pluripotent mouse stem cells by the piggyBac transposon,” Nature Methods, vol. 6, no. 5, pp. 363–369, 2009.
[77]
K. Woltjen, I. P. Michael, P. Mohseni et al., “PiggyBac transposition reprograms fibroblasts to induced pluripotent stem cells,” Nature, vol. 458, no. 7239, pp. 766–770, 2009.
[78]
I. Grabundzija, J. Wang, A. Sebe et al., “Sleeping Beauty transposon-based system for cellular reprogramming and targeted gene insertion in induced pluripotent stem cells,” Nucleic Acids Research, vol. 41, no. 3, pp. 1829–1847, 2013.
[79]
R. P. Davis, C. Nemes, E. Varga et al., “Generation of induced pluripotent stem cells from human foetal fibroblasts using the Sleeping Beauty transposon gene delivery system,” Differentiation, vol. 86, no. 1-2, pp. 30–37, 2013.
[80]
S. Muenthaisong, O. Ujhelly, Z. Polgar et al., “Generation of mouse induced pluripotent stem cells from different genetic backgrounds using Sleeping beauty transposon mediated gene transfer,” Experimental Cell Research, vol. 318, no. 19, pp. 2482–2489, 2012.
[81]
E. P. Papapetrou and M. Sadelain, “Generation of transgene-free human induced pluripotent stem cells with an excisable single polycistronic vector,” Nature Protocols, vol. 6, no. 9, pp. 1251–1273, 2011.
[82]
K. Kaji, K. Norrby, A. Paca, M. Mileikovsky, P. Mohseni, and K. Woltjen, “Virus-free induction of pluripotency and subsequent excision of reprogramming factors,” Nature, vol. 458, no. 7239, pp. 771–775, 2009.
[83]
S. Chakraborty, N. Christoforou, A. Fattahi, R. W. Herzog, and K. W. Leong, “A robust strategy for negative selection of Cre-loxP recombination-based excision of transgenes in induced pluripotent stem cells,” PLoS ONE, vol. 8, no. 5, Article ID e64342, 2013.
[84]
T. Ma, M. Xie, T. Laurent, and S. Ding, “Progress in the reprogramming of somatic cells,” Circulation Research, vol. 112, no. 3, pp. 562–574, 2013.
[85]
L. Warren, P. D. Manos, T. Ahfeldt et al., “Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA,” Cell Stem Cell, vol. 7, no. 5, pp. 618–630, 2010.
[86]
P. K. Mandal and D. J. Rossi, “Reprogramming human fibroblasts to pluripotency using modified mRNA,” Nature Protocols, vol. 8, no. 3, pp. 568–582, 2013.
[87]
Y. Buganim, D. A. Faddah, and R. Jaenisch, “Mechanisms and models of somatic cell reprogramming,” Nature Reviews Genetics, vol. 14, no. 6, pp. 427–439, 2013.
[88]
B. Papp and K. Plath, “Epigenetics of reprogramming to induced pluripotency,” Cell, vol. 152, no. 6, pp. 1324–1343, 2013.
[89]
K. Plath and W. E. Lowry, “Progress in understanding reprogramming to the induced pluripotent state,” Nature Reviews Genetics, vol. 12, no. 4, pp. 253–265, 2011.
[90]
H. Zhou, S. Wu, J. Y. Joo et al., “Generation of induced pluripotent stem cells using recombinant proteins,” Cell Stem Cell, vol. 4, no. 5, pp. 381–384, 2009.
[91]
D. Kim, C.-H. Kim, J.-I. Moon et al., “Generation of human induced pluripotent stem cells by direct delivery of reprogramming proteins,” Cell Stem Cell, vol. 4, no. 6, pp. 472–476, 2009.
[92]
A. El-Sayed, S. Futaki, and H. Harashima, “Delivery of macromolecules using arginine-rich cell-penetrating peptides: ways to overcome endosomal entrapment,” AAPS Journal, vol. 11, no. 1, pp. 13–22, 2009.
[93]
A. Ziegler, P. Nervi, M. Dürrenberger, and J. Seelig, “The cationic cell-penetrating peptide CPPTAT derived from the HIV-1 protein TAT is rapidly transported into living fibroblasts: optical, biophysical, and metabolic evidence,” Biochemistry, vol. 44, no. 1, pp. 138–148, 2005.
[94]
H.-J. Cho, C.-S. Lee, Y.-W. Kwon et al., “Induction of pluripotent stem cells from adult somatic cells by protein-based reprogramming without genetic manipulation,” Blood, vol. 116, no. 3, pp. 386–395, 2010.
[95]
M. Thier, B. Münst, S. Mielke, and F. Edenhofer, “Cellular reprogramming employing recombinant Sox2 protein,” Stem Cells International, vol. 2012, Article ID 549846, 10 pages, 2012.
[96]
M. Bosnali and F. Edenhofer, “Generation of transducible versions of transcription factors Oct4 and Sox2,” Biological Chemistry, vol. 389, no. 7, pp. 851–861, 2008.
[97]
D. Huangfu, K. Osafune, R. Maehr et al., “Induction of pluripotent stem cells from primary human fibroblasts with only Oct4 and Sox2,” Nature Biotechnology, vol. 26, no. 11, pp. 1269–1275, 2008.
[98]
D. Huangfu, R. Maehr, W. Guo et al., “Induction of pluripotent stem cells by defined factors is greatly improved by small-molecule compounds,” Nature Biotechnology, vol. 26, no. 7, pp. 795–797, 2008.
[99]
Y. Yoshida, K. Takahashi, K. Okita, T. Ichisaka, and S. Yamanaka, “Hypoxia enhances the generation of induced pluripotent stem cells,” Cell Stem Cell, vol. 5, no. 3, pp. 237–241, 2009.
[100]
A. Grace, M. McMillan, S. Schmoelzl, and G. Hinch, “187 increased efficiency of deriving bovine stem cell-like colonies using valproic Acid and small-molecule cocktails,” Reproduction, Fertility and Development, vol. 26, no. 1, p. 208, 2013.
[101]
H. F. Teng, Y.-L. Kuo, M. R. Loo et al., “Valproic acid enhances Oct4 promoter activity in myogenic cells,” Journal of Cellular Biochemistry, vol. 110, no. 4, pp. 995–1004, 2010.
[102]
G. N. Pandian, K.-I. Shinohara, A. Ohtsuki et al., “Synthetic small molecules for epigenetic activation of pluripotency genes in mouse embryonic fibroblasts,” ChemBioChem, vol. 12, no. 18, pp. 2822–2828, 2011.
[103]
Z. Zhang and W.-S. Wu, “Sodium butyrate promotes generation of human induced pluripotent stem cells through induction of the miR302/367 cluster,” Stem Cells and Development, vol. 22, no. 16, pp. 2268–2277, 2013.
[104]
P. Mali, B.-K. Chou, J. Yen et al., “Butyrate greatly enhances derivation of human induced pluripotent stem cells by promoting epigenetic remodeling and the expression of pluripotency-associated genes,” Stem Cells, vol. 28, no. 4, pp. 713–720, 2010.
[105]
T. S. Mikkelsen, J. Hanna, X. Zhang et al., “Dissecting direct reprogramming through integrative genomic analysis,” Nature, vol. 454, no. 7200, pp. 49–55, 2008.
[106]
Y. Shi, J. T. Do, C. Desponts, H. S. Hahm, H. R. Sch?ler, and S. Ding, “A combined chemical and genetic approach for the generation of induced pluripotent stem cells,” Cell Stem Cell, vol. 3, no. 1, p. 119, 2008.
[107]
Y. Shi, C. Desponts, J. T. Do, H. S. Hahm, H. R. Sch?ler, and S. Ding, “Induction of pluripotent stem cells from mouse embryonic fibroblasts by Oct4 and Klf4 with small-molecule compounds,” Cell Stem Cell, vol. 3, no. 5, pp. 568–574, 2008.
[108]
Z. Pasha, H. K. Haider, and M. Ashraf, “Efficient non-viral reprogramming of myoblasts to stemness with a single small molecule to generate cardiac progenitor cells,” PLoS ONE, vol. 6, no. 8, Article ID e23667, 2011.
[109]
J. R. Plews, J. Li, M. Jones et al., “Activation of pluripotency genes in human fibroblast cells by a novel mRNA based approach,” PLoS ONE, vol. 5, no. 12, Article ID e14397, 2010.
[110]
H. Niwa, “Wnt: what's Needed to maintain pluripotency?” Nature Cell Biology, vol. 13, no. 9, pp. 1024–1026, 2011.
[111]
W. Li, H. Zhou, R. Abujarour et al., “Generation of human-induced pluripotent stem cells in the absence of exogenous Sox2,” Stem Cells, vol. 27, no. 12, pp. 2992–3000, 2009.
[112]
S. Petkov and H. Niemann, “189 small molecule inhibitors pd0325901 and chir99021 cause reduced expression of pluripotency genes in putative porcine induced pluripotent stem cells,” Reproduction, Fertility and Development, vol. 26, no. 1, p. 209, 2013.
[113]
T. Lin, R. Ambasudhan, X. Yuan et al., “A chemical platform for improved induction of human iPSCs,” Nature Methods, vol. 6, no. 11, pp. 805–808, 2009.
[114]
T. Chen, D. Yuan, B. Wei et al., “E-cadherin-mediated cell-cell contact is critical for induced pluripotent stem cell generation,” Stem Cells, vol. 28, no. 8, pp. 1315–1325, 2010.
[115]
X. Yuan, H. Wan, X. Zhao, S. Zhu, Q. Zhou, and S. Ding, “Brief report: combined chemical treatment enables Oct4-induced reprogramming from mouse embryonic fibroblasts,” Stem Cells, vol. 29, no. 3, pp. 549–553, 2011.
[116]
Y. Wang and J. Adjaye, “A cyclic AMP analog, 8-Br-cAMP, enhances the induction of pluripotency in human fibroblast cells,” Stem Cell Reviews and Reports, vol. 7, no. 2, pp. 331–341, 2011.
[117]
J. Staerk, C. A. Lyssiotis, L. A. Medeiro et al., “Pan-src family kinase inhibitors replace Sox2 during the direct reprogramming of somatic cells,” Angewandte Chemie, vol. 50, no. 25, pp. 5734–5736, 2011.
[118]
A. Banito, S. T. Rashid, J. C. Acosta et al., “Senescence impairs successful reprogramming to pluripotent stem cells,” Genes and Development, vol. 23, no. 18, pp. 2134–2139, 2009.
[119]
P. Phanthong, H. Raveh-Amit, T. Li, Y. Kitiyanant, and A. Dinnyes, “Is aging a barrier to reprogramming? Lessons from induced pluripotent stem cells,” Biogerontology, vol. 14, no. 6, pp. 591–602, 2013.
[120]
R. M. Marion, K. Strati, H. Li et al., “Telomeres acquire embryonic stem cell characteristics in induced pluripotent stem cells,” Cell Stem Cell, vol. 4, no. 2, pp. 141–154, 2009.
[121]
T. Chen, L. Shen, J. Yu et al., “Rapamycin and other longevity-promoting compounds enhance the generation of mouse induced pluripotent stem cells,” Aging Cell, vol. 10, no. 5, pp. 908–911, 2011.
[122]
M. A. Esteban, T. Wang, B. Qin et al., “Vitamin C enhances the generation of mouse and human induced pluripotent stem cells,” Cell Stem Cell, vol. 6, no. 1, pp. 71–79, 2010.
[123]
X. Xu, S. Duan, F. Yi, A. Ocampo, G. H. Liu, and B. J. Izpisua, “Mitochondrial regulation in pluripotent stem cells,” Cell Metabolism, vol. 18, no. 3, pp. 325–332, 2013.
[124]
C. D. Folmes, T. J. Nelson, and A. Terzic, “Energy metabolism in nuclear reprogramming,” Biomarkers in Medicine, vol. 5, no. 6, pp. 715–729, 2011.
[125]
S. Zhu, W. Wei, and S. Ding, “Chemical strategies for stem cell biology and regenerative medicine,” Annual Review of Biomedical Engineering, vol. 13, pp. 73–90, 2011.
[126]
J.-B. Su, D.-Q. Pei, and B.-M. Qin, “Roles of small molecules in somatic cell reprogramming,” Acta Pharmacologica Sinica, vol. 34, no. 6, pp. 719–724, 2013.
[127]
P. Hou, Y. Li, X. Zhang et al., “Pluripotent stem cells induced from mouse somatic cells by small-molecule compounds,” Science, vol. 341, no. 6146, pp. 651–654, 2013.
[128]
B. K. Chou and L. Cheng, “And then there were none: no need for pluripotency factors to induce reprogramming,” Cell Stem Cell, vol. 13, no. 3, pp. 261–262, 2013.
[129]
M.-R. Suh, Y. Lee, J. Y. Kim et al., “Human embryonic stem cells express a unique set of microRNAs,” Developmental Biology, vol. 270, no. 2, pp. 488–498, 2004.
[130]
H. B. Houbaviy, M. F. Murray, and P. A. Sharp, “Embryonic stem cell-specific microRNAs,” Developmental Cell, vol. 5, no. 2, pp. 351–358, 2003.
[131]
R. L. Judson, J. E. Babiarz, M. Venere, and R. Blelloch, “Embryonic stem cell-specific microRNAs promote induced pluripotency,” Nature Biotechnology, vol. 27, no. 5, pp. 459–461, 2009.
[132]
C. Melton, R. L. Judson, and R. Blelloch, “Opposing microRNA families regulate self-renewal in mouse embryonic stem cells,” Nature, vol. 463, no. 7281, pp. 621–626, 2010.
[133]
M. Lagos-Quintana, R. Rauhut, W. Lendeckel, and T. Tuschl, “Identification of novel genes coding for small expressed RNAs,” Science, vol. 294, no. 5543, pp. 853–858, 2001.
[134]
E. Huntzinger and E. Izaurralde, “Gene silencing by microRNAs: contributions of translational repression and mRNA decay,” Nature Reviews Genetics, vol. 12, no. 2, pp. 99–110, 2011.
[135]
J. M. Friedman and P. A. Jones, “MicroRNAs: critical mediators of differentiation, development and disease,” Swiss Medical Weekly, vol. 139, no. 33-34, pp. 466–472, 2009.
[136]
T. Thum, D. Catalucci, and J. Bauersachs, “MicroRNAs: novel regulators in cardiac development and disease,” Cardiovascular Research, vol. 79, no. 4, pp. 562–570, 2008.
[137]
S.-L. Lin, D. C. Chang, S. Chang-Lin et al., “Mir-302 reprograms human skin cancer cells into a pluripotent ES-cell-like state,” RNA, vol. 14, no. 10, pp. 2115–2124, 2008.
[138]
R. L. Judson, J. E. Babiarz, M. Venere, and R. Blelloch, “Embryonic stem cell-specific microRNAs promote induced pluripotency,” Nature Biotechnology, vol. 27, no. 5, pp. 459–461, 2009.
[139]
R. Sridharan and K. Plath, “Small RNAs loom large during reprogramming,” Cell Stem Cell, vol. 8, no. 6, pp. 599–601, 2011.
[140]
F. Anokye-Danso, C. M. Trivedi, D. Juhr et al., “Highly efficient miRNA-mediated reprogramming of mouse and human somatic cells to pluripotency,” Cell Stem Cell, vol. 8, no. 4, pp. 376–388, 2011.
[141]
G. Wang, X. Guo, W. Hong et al., “Critical regulation of miR-200/ZEB2 pathway in Oct4/Sox2-induced mesenchymal-to-epithelial transition and induced pluripotent stem cell generation,” Proceedings of the National Academy of Sciences of the United States of America, vol. 110, no. 8, pp. 2858–2863, 2013.
[142]
X. Guo, Q. Liu, G. Wang et al., “MicroRNA-29b is a novel mediator of Sox2 function in the regulation of somatic cell reprogramming,” Cell Research, vol. 23, no. 1, pp. 142–156, 2013.
[143]
D. Subramanyam, S. Lamouille, R. L. Judson et al., “Multiple targets of miR-302 and miR-372 promote reprogramming of human fibroblasts to induced pluripotent stem cells,” Nature Biotechnology, vol. 29, no. 5, pp. 443–448, 2011.
[144]
S.-L. Lin, D. C. Chang, C.-H. Lin, S.-Y. Ying, D. Leu, and D. T. S. Wu, “Regulation of somatic cell reprogramming through inducible mir-302 expression,” Nucleic Acids Research, vol. 39, no. 3, pp. 1054–1065, 2011.
[145]
N. Pfaff, J. Fiedler, A. Holzmann et al., “MiRNA screening reveals a new miRNA family stimulating iPS cell generation via regulation of Meox2,” EMBO Reports, vol. 12, no. 11, pp. 1153–1159, 2011.
[146]
Z. Li, C.-S. Yang, K. Nakashima, and T. M. Rana, “Small RNA-mediated regulation of iPS cell generation,” EMBO Journal, vol. 30, no. 5, pp. 823–834, 2011.
[147]
C.-H. Kuo and S.-Y. Ying, “MicroRNA-mediated somatic cell reprogramming,” Journal of Cellular Biochemistry, vol. 114, no. 2, pp. 275–281, 2013.
[148]
N. Xu, T. Papagiannakopoulos, G. Pan, J. A. Thomson, and K. S. Kosik, “MicroRNA-145 regulates OCT4, SOX2, and KLF4 and represses pluripotency in human embryonic stem cells,” Cell, vol. 137, no. 4, pp. 647–658, 2009.
[149]
C.-S. Yang, Z. Li, and T. M. Rana, “microRNAs modulate iPS cell generation,” RNA, vol. 17, no. 8, pp. 1451–1460, 2011.
[150]
Y. J. Choi, C.-P. Lin, J. J. Ho et al., “MiR-34 miRNAs provide a barrier for somatic cell reprogramming,” Nature Cell Biology, vol. 13, no. 11, pp. 1353–1360, 2011.
[151]
Y. L. Lee, Q. Peng, S. W. Fong et al., “Sirtuin 1 facilitates generation of induced pluripotent stem cells from mouse embryonic fibroblasts through the miR-34a and p53 pathways,” PLoS ONE, vol. 7, no. 9, Article ID e45633, 2012.
[152]
K. A. Worringer, T. A. Rand, Y. Hayashi et al., “The let-7/LIN-41 pathway regulates reprogramming to human induced pluripotent stem cells by controlling expression of prodifferentiation genes,” Cell Stem Cell, vol. 14, no. 1, pp. 40–52, 2014.
[153]
K. Takahashi and S. Yamanaka, “Induced pluripotent stem cells in medicine and biology,” Development, vol. 140, no. 12, pp. 2457–2461, 2013.
[154]
O. Iglesias-García, B. Pelacho, and F. Prósper, “Induced pluripotent stem cells as a new strategy for cardiac regeneration and disease modeling,” Journal of Molecular and Cellular Cardiology, vol. 62, pp. 43–50, 2013.
[155]
H. Okano, M. Nakamura, K. Yoshida et al., “Steps toward safe cell therapy using induced pluripotent stem cells,” Circulation Research, vol. 112, no. 3, pp. 523–533, 2013.
[156]
J. Liang and Y. Wang, “Stem/progenitor cell based therapies for repair of myocardial infarction: current developments in methods of cell delivery,” Surgery: Current Research, vol. 3, no. 2, p. 131, 2013.
[157]
L.-C. Hsiao, C. Carr, K.-C. Chang, S.-Z. Lin, and K. Clarke, “Stem cell-based therapy for ischemic heart disease,” Cell Transplantation, vol. 22, no. 4, pp. 663–675, 2013.
[158]
M. A. Laflamme and C. E. Murry, “Heart regeneration,” Nature, vol. 473, no. 7347, pp. 326–335, 2011.
[159]
E. C. Perin, J. T. Willerson, C. J. Pepine et al., “Effect of transendocardial delivery of autologous bone marrow mononuclear cells on functional capacity, left ventricular function, and perfusion in chronic heart failure: the FOCUS-CCTRN trial,” Journal of the American Medical Association, vol. 307, no. 16, pp. 1717–1726, 2012.
[160]
A. R. Chugh, G. M. Beache, J. H. Loughran et al., “Administration of cardiac stem cells in patients with ischemic cardiomyopathy: the SCIPIO trial: surgical aspects and interim analysis of myocardial function and viability by magnetic resonance,” Circulation, vol. 126, no. 11, pp. S54–S64, 2012.
[161]
A. Leri and P. Anversa, “Bone-marrow-derived cells and heart failure-the debate goes on,” Nature Reviews Cardiology, vol. 10, no. 7, pp. 372–373, 2013.
[162]
M.-M. Zaruba, M. Soonpaa, S. Reuter, and L. J. Field, “Cardiomyogenic potential of C-kit+-expressing cells derived from neonatal and adult mouse hearts,” Circulation, vol. 121, no. 18, pp. 1992–2000, 2010.
[163]
I. Chimenti, R. R. Smith, T.-S. Li et al., “Relative roles of direct regeneration versus paracrine effects of human cardiosphere-derived cells transplanted into infarcted mice,” Circulation Research, vol. 106, no. 5, pp. 971–980, 2010.
[164]
J. Beers, D. R. Gulbranson, N. George et al., “Passaging and colony expansion of human pluripotent stem cells by enzyme-free dissociation in chemically defined culture conditions,” Nature Protocols, vol. 7, no. 11, pp. 2029–2040, 2012.
[165]
G. Chen, D. R. Gulbranson, Z. Hou et al., “Chemically defined conditions for human iPSC derivation and culture,” Nature Methods, vol. 8, no. 5, pp. 424–429, 2011.
[166]
S. Yamanaka, “Induced pluripotent stem cells: past, present, and future,” Cell Stem Cell, vol. 10, no. 6, pp. 678–684, 2012.
[167]
J. Zhang, G. F. Wilson, A. G. Soerens et al., “Functional cardiomyocytes derived from human induced pluripotent stem cells,” Circulation Research, vol. 104, no. 4, pp. e30–e41, 2009.
[168]
S. Yamanaka, “Patient-Specific pluripotent stem cells become even more accessible,” Cell Stem Cell, vol. 7, no. 1, pp. 1–2, 2010.
[169]
K. Yusa, S. T. Rashid, H. Strick-Marchand et al., “Targeted gene correction of α1-antitrypsin deficiency in induced pluripotent stem cells,” Nature, vol. 478, no. 7369, pp. 391–394, 2011.
[170]
A. Filareto, S. Parker, R. Darabi et al., “An ex vivo gene therapy approach to treat muscular dystrophy using inducible pluripotent stem cells,” Nature Communications, vol. 4, article 1549, 2013.
[171]
C. Mauritz, K. Schwanke, M. Reppel et al., “Generation of functional murine cardiac myocytes from induced pluripotent stem cells,” Circulation, vol. 118, no. 5, pp. 507–517, 2008.
[172]
K. Schenke-Layland, K. E. Rhodes, E. Angelis et al., “Reprogrammed mouse fibroblasts differentiate into cells of the cardiovascular and hematopoietic lineages,” Stem Cells, vol. 26, no. 6, pp. 1537–1546, 2008.
[173]
J. Zhang, M. Klos, G. F. Wilson et al., “Extracellular matrix promotes highly efficient cardiac differentiation of human pluripotent stem cells: the matrix sandwich method,” Circulation Research, vol. 111, no. 9, pp. 1125–1136, 2012.
[174]
G. Narazaki, H. Uosaki, M. Teranishi et al., “Directed and systematic differentiation of cardiovascular cells from mouse induced pluripotent stem cells,” Circulation, vol. 118, no. 5, pp. 498–506, 2008.
[175]
E. N. Olson, “Gene regulatory networks in the evolution and development of the heart,” Science, vol. 313, no. 5795, pp. 1922–1927, 2006.
[176]
E. N. Olson and M. D. Schneider, “Sizing up the heart: development redux in disease,” Genes and Development, vol. 17, no. 16, pp. 1937–1956, 2003.
[177]
S. Martin-Puig, Z. Wang, and K. R. Chien, “Lives of a heart cell: tracing the origins of cardiac progenitors,” Cell Stem Cell, vol. 2, no. 4, pp. 320–331, 2008.
[178]
C. L. Mummery, J. Zhang, E. S. Ng, D. A. Elliott, A. G. Elefanty, and T. J. Kamp, “Differentiation of human embryonic stem cells and induced pluripotent stem cells to cardiomyocytes: a methods overview,” Circulation Research, vol. 111, no. 3, pp. 344–358, 2012.
[179]
T. J. Nelson, A. Martinez-Fernandez, S. Yamada, C. Perez-Terzic, Y. Ikeda, and A. Terzic, “Repair of acute myocardial infarction with induced pluripotent stem cells induced by human stemness factors,” Circulation, vol. 120, no. 5, pp. 408–416, 2009.
[180]
L. Zwi-Dantsis, I. Huber, M. Habib et al., “Derivation and cardiomyocyte differentiation of induced pluripotent stem cells from heart failure patients,” European Heart Journal, vol. 34, no. 21, pp. 1575–1586, 2013.
[181]
P. W. Burridge and E. T. Zambidis, “Highly efficient directed differentiation of human induced pluripotent stem cells into cardiomyocytes,” Methods in Molecular Biology, vol. 997, pp. 149–161, 2013.
[182]
X. Lian, J. Zhang, S. M. Azarin et al., “Directed cardiomyocyte differentiation from human pluripotent stem cells by modulating Wnt/β-catenin signaling under fully defined conditions,” Nature Protocols, vol. 8, no. 1, pp. 162–175, 2013.
[183]
X. Lian, C. Hsiao, G. Wilson et al., “Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling,” Proceedings of the National Academy of Sciences of the United States of America, vol. 109, no. 27, pp. E1848–E1857, 2012.
[184]
Y. Shiba, S. Fernandes, W.-Z. Zhu et al., “Human ES-cell-derived cardiomyocytes electrically couple and suppress arrhythmias in injured hearts,” Nature, vol. 489, no. 7415, pp. 322–325, 2012.
[185]
M. Rubach, R. Adelmann, M. Haustein et al., et al., “Mesenchymal stem cells and their conditioned medium improve integration of purified induced pluripotent stem cell-derived cardiomyocyte clusters into myocardial tissue,” Stem Cells and Development, vol. 23, no. 6, pp. 643–653, 2014.
[186]
L. Chang, M. Noseda, M. Higginson et al., “Differentiation of vascular smooth muscle cells from local precursors during embryonic and adult arteriogenesis requires Notch signaling,” Proceedings of the National Academy of Sciences of the United States of America, vol. 109, no. 18, pp. 6993–6998, 2012.
[187]
C. Cochain, K. M. Channon, and J.-S. Silvestre, “Angiogenesis in the infarcted myocardium,” Antioxidants and Redox Signaling, vol. 18, no. 9, pp. 1100–1113, 2013.
[188]
L. Carpenter, C. Carr, C. T. Yang, D. J. Stuckey, K. Clarke, and S. M. Watt, “Efficient differentiation of human induced pluripotent stem cells generates cardiac cells that provide protection following myocardial infarction in the rat,” Stem Cells and Development, vol. 21, no. 6, pp. 977–986, 2012.
[189]
W.-H. Lai, J. C. Y. Ho, Y.-C. Chan et al., “Attenuation of hind-limb ischemia in mice with endothelial-like cells derived from different sources of human stem cells,” PLoS ONE, vol. 8, no. 3, Article ID e57876, 2013.
[190]
T. S. Park, I. Bhutto, L. Zimmerlin et al., et al., “Vascular progenitors from cord blood-derived iPSC possess augmented capacity for regenerating ischemic retinal vasculature,” Circulation, vol. 129, no. 3, pp. 359–372, 2013.
[191]
A. J. Rufaihah, N. F. Huang, J. Kim et al., “Human induced pluripotent stem cell-derived endothelial cells exhibit functional heterogeneity,” American Journal of Translational Research, vol. 5, no. 1, pp. 21–35, 2013.
[192]
J. Fujita, Y. Itabashi, T. Seki et al., “Myocardial cell sheet therapy and cardiac function,” American Journal of Physiology: Heart and Circulatory Physiology, vol. 303, no. 10, pp. H1169–H1182, 2012.
[193]
H. Masumoto, T. Matsuo, K. Yamamizu et al., “Pluripotent stem cell-engineered cell sheets reassembled with defined cardiovascular populations ameliorate reduction in infarct heart function through cardiomyocyte-mediated neovascularization,” Stem Cells, vol. 30, no. 6, pp. 1196–1205, 2012.
[194]
M. Kawamura, S. Miyagawa, K. Miki et al., “Feasibility, safety, and therapeutic efficacy of human induced pluripotent stem cell-derived cardiomyocyte sheets in a porcine ischemic cardiomyopathy model,” Circulation, vol. 126, no. 11, pp. S29–S37, 2012.
[195]
B. Dai, W. Huang, M. Xu et al., “Reduced collagen deposition in infarcted myocardium facilitates induced pluripotent stem cell engraftment and angiomyogenesis for improvement of left ventricular function,” Journal of the American College of Cardiology, vol. 58, no. 20, pp. 2118–2127, 2011.
[196]
W. Huang, B. Dai, Z. Wen et al., “Molecular Strategy to Reduce In Vivo Collagen Barrier Promotes Entry of NCX1 Positive Inducible Pluripotent Stem Cells (iPSCNCX1+) into Ischemic (or Injured) Myocardium,” PLoS ONE, vol. 8, no. 8, Article ID e70023, 2013.
[197]
M. Kawamura, S. Miyagawa, S. Fukushima et al., et al., “Enhanced survival of transplanted human induced pluripotent stem cell-derived cardiomyocytes by the combination of cell sheets with the pedicled omental flap technique in a porcine heart,” Circulation, vol. 128, 1, pp. S87–S94, 2013.
[198]
M. Halbach, G. Peinkofer, S. Baumgartner et al., et al., “Electrophysiological integration and action potential properties of transplanted cardiomyocytes derived from induced pluripotent stem cells,” Cardiovascular Research, vol. 100, no. 3, pp. 432–440, 2013.
[199]
M. Ieda, J.-D. Fu, P. Delgado-Olguin et al., “Direct reprogramming of fibroblasts into functional cardiomyocytes by defined factors,” Cell, vol. 142, no. 3, pp. 375–386, 2010.
[200]
J. A. Efe, S. Hilcove, J. Kim et al., “Conversion of mouse fibroblasts into cardiomyocytes using a direct reprogramming strategy,” Nature Cell Biology, vol. 13, no. 3, pp. 215–222, 2011.
[201]
K. Inagawa, K. Miyamoto, H. Yamakawa et al., “Induction of cardiomyocyte-like cells in infarct hearts by gene transfer of Gata4, Mef2c, and Tbx5,” Circulation Research, vol. 111, no. 9, pp. 1147–1156, 2012.
[202]
L. Qian, Y. Huang, C. I. Spencer et al., “In vivo reprogramming of murine cardiac fibroblasts into induced cardiomyocytes,” Nature, vol. 485, no. 7400, pp. 593–598, 2012.
[203]
L. Qian, E. C. Berry, J.-D. Fu, M. Ieda, and D. Srivastava, “Reprogramming of mouse fibroblasts into cardiomyocyte-like cells in vitro,” Nature Protocols, vol. 8, no. 6, pp. 1204–1215, 2013.
[204]
K. Song, Y.-J. Nam, X. Luo et al., “Heart repair by reprogramming non-myocytes with cardiac transcription factors,” Nature, vol. 485, no. 7400, pp. 599–604, 2012.
[205]
J. X. Chen, M. Krane, M. A. Deutsch et al., “Inefficient reprogramming of fibroblasts into cardiomyocytes using Gata4, Mef2c, and Tbx5,” Circulation Research, vol. 111, no. 1, pp. 50–55, 2012.
[206]
M. Mathison, R. P. Gersch, A. Nasser et al., “In vivo cardiac cellular reprogramming efficacy is enhanced by angiogenic preconditioning of the infarcted myocardium with vascular endothelial growth factor,” Journal of the American Heart Association, vol. 1, no. 6, 2012.
[207]
R. Wada, N. Muraoka, K. Inagawa et al., “Induction of human cardiomyocyte-like cells from fibroblasts by defined factors,” Proceedings of the National Academy of Sciences of the United States of America, vol. 110, no. 31, pp. 12667–12672, 2013.
[208]
A. Bird, “Perceptions of epigenetics,” Nature, vol. 447, no. 7143, pp. 396–398, 2007.
[209]
Y.-J. Nam, K. Song, and E. N. Olson, “Heart repair by cardiac reprogramming,” Nature Medicine, vol. 19, no. 4, pp. 413–415, 2013.
[210]
L. Qian and D. Srivastava, “Direct cardiac reprogramming: from developmental biology to cardiac regeneration,” Cardiovascular Research, vol. 113, no. 7, pp. 915–921, 2013.
[211]
J. Chen, H. Liu, J. Liu et al., “H3K9 methylation is a barrier during somatic cell reprogramming into iPSCs,” Nature Genetics, vol. 45, no. 1, pp. 34–42, 2013.
[212]
M. Vecellio, V. Meraviglia, S. Nanni et al., “In vitro epigenetic reprogramming of human cardiac mesenchymal stromal cells into functionally competent cardiovascular precursors,” PLoS ONE, vol. 7, no. 12, Article ID e51694, 2012.
[213]
A. Margariti, B. Winkler, E. Karamariti et al., “Direct reprogramming of fibroblasts into endothelial cells capable of angiogenesis and reendothelialization in tissue-engineered vessels,” Proceedings of the National Academy of Sciences of the United States of America, vol. 109, no. 34, pp. 13793–13798, 2012.
[214]
J. Li, N. F. Huang, J. Zou et al., “Conversion of human fibroblasts to functional endothelial cells by defined factors,” Arteriosclerosis, Thrombosis, and Vascular Biology, vol. 33, no. 6, pp. 1366–1375, 2013.
[215]
T. M. Jayawardena, B. Egemnazarov, E. A. Finch et al., “MicroRNA-mediated in vitro and in vivo direct reprogramming of cardiac fibroblasts to cardiomyocytes,” Circulation Research, vol. 110, no. 11, pp. 1465–1473, 2012.
[216]
D. A. Robinton and G. Q. Daley, “The promise of induced pluripotent stem cells in research and therapy,” Nature, vol. 481, no. 7381, pp. 295–305, 2012.
[217]
F. T. Merkle and K. Eggan, “Modeling human disease with pluripotent stem cells: from genome association to function,” Cell Stem Cell, vol. 12, no. 6, pp. 656–668, 2013.
[218]
L. M. R. Ferreira and M. A. Mostajo-Radji, “How induced pluripotent stem cells are redefining personalized medicine,” Gene, vol. 520, no. 1, pp. 1–6, 2013.
[219]
M. A. G. Van Der Heyden and M. K. B. Jonsson, “Personalized medicine and the role of induced pluripotent stem cells,” Cardiovascular Research, vol. 95, no. 4, pp. 395–396, 2012.
[220]
S. J. Engle and D. Puppala, “Integrating human pluripotent stem cells into drug development,” Cell Stem Cell, vol. 12, no. 6, pp. 699–712, 2013.
[221]
J. T. Dimos, K. T. Rodolfa, K. K. Niakan et al., “Induced pluripotent stem cells generated from patients with ALS can be differentiated into motor neurons,” Science, vol. 321, no. 5893, pp. 1218–1221, 2008.
[222]
I.-H. Park, N. Arora, H. Huo et al., “Disease-Specific Induced Pluripotent Stem Cells,” Cell, vol. 134, no. 5, pp. 877–886, 2008.
[223]
A. D. Ebert, J. Yu, F. F. Rose Jr. et al., “Induced pluripotent stem cells from a spinal muscular atrophy patient,” Nature, vol. 457, no. 7227, pp. 277–280, 2009.
[224]
M. Grskovic, A. Javaherian, B. Strulovici, and G. Q. Daley, “Induced pluripotent stem cells—opportunities for disease modelling and drug discovery,” Nature Reviews Drug Discovery, vol. 10, no. 12, pp. 915–929, 2011.
[225]
C. Dambrot, R. Passier, D. Atsma, and C. L. Mummery, “Cardiomyocyte differentiation of pluripotent stem cells and their use as cardiac disease models,” Biochemical Journal, vol. 434, no. 1, pp. 25–35, 2011.
[226]
X. Carvajal-Vergara, A. Sevilla, S. L. Dsouza et al., “Patient-specific induced pluripotent stem-cell-derived models of LEOPARD syndrome,” Nature, vol. 465, no. 7299, pp. 808–812, 2010.
[227]
A. Moretti, M. Bellin, A. Welling et al., “Patient-specific induced pluripotent stem-cell models for long-QT syndrome,” The New England Journal of Medicine, vol. 363, no. 15, pp. 1397–1409, 2010.
[228]
I. Itzhaki, L. Maizels, I. Huber et al., “Modelling the long QT syndrome with induced pluripotent stem cells,” Nature, vol. 471, no. 7337, pp. 225–230, 2011.
[229]
M. Yazawa, B. Hsueh, X. Jia et al., “Using induced pluripotent stem cells to investigate cardiac phenotypes in Timothy syndrome,” Nature, vol. 471, no. 7337, pp. 230–236, 2011.
[230]
F. Lan, A. S. Lee, P. Liang et al., “Abnormal calcium handling properties underlie familial hypertrophic cardiomyopathy pathology in patient-specific induced pluripotent stem cells,” Cell Stem Cell, vol. 12, no. 1, pp. 101–113, 2013.
[231]
N. Sun, M. Yazawa, J. Liu et al., “Patient-specific induced pluripotent stem cells as a model for familial dilated cardiomyopathy,” Science Translational Medicine, vol. 4, no. 130, Article ID 130ra47, pp. 130r–147r, 2012.
[232]
D. Ma, H. Wei, J. Lu et al., “Generation of patient-specific induced pluripotent stem cell-derived cardiomyocytes as a cellular model of arrhythmogenic right ventricular cardiomyopathy,” European Heart Journal, vol. 34, no. 15, pp. 1122–1133, 2013.
[233]
R. P. Davis, S. Casini, C. W. Van Den Berg et al., “Cardiomyocytes derived from pluripotent stem cells recapitulate electrophysiological characteristics of an overlap syndrome of cardiac sodium channel disease,” Circulation, vol. 125, no. 25, pp. 3079–3091, 2012.
[234]
D. Malan, S. Friedrichs, B. K. Fleischmann, and P. Sasse, “Cardiomyocytes obtained from induced pluripotent stem cells with Long-QT syndrome 3 recapitulate typical disease-specific features in vitro,” Circulation Research, vol. 109, no. 8, pp. 841–847, 2011.
[235]
M. Bellin, S. Casini, R. P. Davis et al., et al., “Isogenic human pluripotent stem cell pairs reveal the role of a KCNH2 mutation in long-QT syndrome,” The EMBO Journal, vol. 32, no. 24, pp. 3161–3175, 2013.
[236]
D. Sinnecker, A. Goedel, K.-L. Laugwitz, and A. Moretti, “Induced pluripotent stem cell-derived cardiomyocytes: a versatile tool for arrhythmia research,” Circulation Research, vol. 112, no. 6, pp. 961–968, 2013.
[237]
C. B. Jung, A. Moretti, M. Mederos y Schnitzler et al., “Dantrolene rescues arrhythmogenic RYR2 defect in a patient-specific stem cell model of catecholaminergic polymorphic ventricular tachycardia,” EMBO Molecular Medicine, vol. 4, no. 3, pp. 180–191, 2012.
[238]
H. Zhou, M. Xu, Q. Huang et al., “Genome-scale RNAi screen for host factors required for HIV replication,” Cell Host and Microbe, vol. 4, no. 5, pp. 495–504, 2008.
[239]
C. W. Scott, M. F. Peters, and Y. P. Dragan, “Human induced pluripotent stem cells and their use in drug discovery for toxicity testing,” Toxicology Letters, vol. 219, no. 1, pp. 49–58, 2013.
[240]
J. C. Davila, G. G. Cezar, M. Thiede, S. Strom, T. Miki, and J. Trosko, “Use and application of stem cells in toxicology,” Toxicological Sciences, vol. 79, no. 2, pp. 214–223, 2004.
[241]
T. Seki, S. Yuasa, M. Oda et al., “Generation of induced pluripotent stem cells from human terminally differentiated circulating t cells,” Cell Stem Cell, vol. 7, no. 1, pp. 11–13, 2010.
[242]
P. Liang, F. Lan, A. S. Lee et al., “Drug screening using a library of human induced pluripotent stem cell-derived cardiomyocytes reveals disease-specific patterns of cardiotoxicity,” Circulation, vol. 127, no. 16, pp. 1677–1691, 2013.
[243]
H. Lal, K. L. Kolaja, and T. Force, “Cancer genetics and the cardiotoxicity of the therapeutics,” Journal of the American College of Cardiology, vol. 61, no. 3, pp. 267–274, 2013.
[244]
M. Mercola, A. Colas, and E. Willems, “Induced pluripotent stem cells in cardiovascular drug discovery,” Circulation Research, vol. 112, no. 3, pp. 534–548, 2013.
[245]
H. Kamao, M. Mandai, S. Okamoto et al., “Characterization of human induced pluripotent stem cell-derived retinal pigment epithelium cell sheets aiming for clinical application,” Stem Cell Reports, vol. 2, no. 2, pp. 205–218, 2014.
[246]
D. Cyranoski, “Stem cells cruise to clinic,” Nature, vol. 494, no. 7438, p. 413, 2013.
[247]
F. Hattori, H. Chen, H. Yamashita et al., “Nongenetic method for purifying stem cell-derived cardiomyocytes,” Nature Methods, vol. 7, no. 1, pp. 61–66, 2010.
[248]
H. Uosaki, H. Fukushima, A. Takeuchi et al., “Efficient and scalable purification of cardiomyocytes from human embryonic and induced pluripotent stem cells by VCAM1 surface expression,” PLoS ONE, vol. 6, no. 8, Article ID e23657, 2011.
[249]
D. A. Elliott, S. R. Braam, K. Koutsis et al., “ hESCs for isolation of human cardiac progenitors and cardiomyocytes,” Nature Methods, vol. 8, no. 12, pp. 1037–1043, 2011.
[250]
L. W. Van Laake, L. Qian, P. Cheng et al., “Reporter-based isolation of induced pluripotent stem cell-and embryonic stem cell-derived cardiac progenitors reveals limited gene expression variance,” Circulation Research, vol. 107, no. 3, pp. 340–347, 2010.
[251]
G. Blin, D. Nury, S. Stefanovic et al., “A purified population of multipotent cardiovascular progenitors derived from primate pluripotent stem cells engrafts in postmyocardial infarcted nonhuman primates,” The Journal of Clinical Investigation, vol. 120, no. 4, pp. 1125–1139, 2010.
[252]
K. Ban, B. Wile, S. Kim et al., et al., “Purification of cardiomyocytes from differentiating pluripotent stem cells using molecular beacons that target cardiomyocyte-specific mRNA,” Circulation, vol. 128, no. 17, pp. 1897–1909, 2013.
[253]
S. Tohyama, F. Hattori, M. Sano et al., “Distinct metabolic flow enables large-scale purification of mouse and human pluripotent stem cell-derived cardiomyocytes,” Cell Stem Cell, vol. 12, no. 1, pp. 127–137, 2013.
[254]
U. Ben-David and N. Benvenisty, “The tumorigenicity of human embryonic and induced pluripotent stem cells,” Nature Reviews Cancer, vol. 11, no. 4, pp. 268–277, 2011.
[255]
K. C. Wollert, G. P. Meyer, J. Lotz et al., “Intracoronary autologous bone-marrow cell transfer after myocardial infarction: the BOOST randomised controlled clinical trial,” The Lancet, vol. 364, no. 9429, pp. 141–148, 2004.
[256]
Y. Mayshar, U. Ben-David, N. Lavon et al., “Identification and classification of chromosomal aberrations in human induced pluripotent stem cells,” Cell Stem Cell, vol. 7, no. 4, pp. 521–531, 2010.
[257]
E. Vanneste, T. Voet, C. Le Caignec et al., “Chromosome instability is common in human cleavage-stage embryos,” Nature Medicine, vol. 15, no. 5, pp. 577–583, 2009.
[258]
N. Hyka-Nouspikel, J. Desmarais, P. J. Gokhale et al., “Deficient DNA damage response and cell cycle checkpoints lead to accumulation of point mutations in human embryonic stem cells,” Stem Cells, vol. 30, no. 9, pp. 1901–1910, 2012.
[259]
M. X. Doss, J. M. Di Diego, R. J. Goodrow et al., “Maximum diastolic potential of human induced pluripotent stem cell-derived cardiomyocytes depends critically on IKr,” PLoS ONE, vol. 7, no. 7, Article ID e40288, 2012.
[260]
A. Kuzmenkin, H. Liang, G. Xu et al., “Functional characterization of cardiomyocytes derived from murine induced pluripotent stem cells in vitro,” FASEB Journal, vol. 23, no. 12, pp. 4168–4180, 2009.
[261]
J. M. Cordeiro, V. V. Nesterenko, S. Sicouri et al., “Identification and characterization of a transient outward K+ current in human induced pluripotent stem cell-derived cardiomyocytes,” Journal of Molecular and Cellular Cardiology, vol. 60, no. 1, pp. 36–46, 2013.
[262]
C. Y. Ivashchenko, G. C. Pipes, I. M. Lozinskaya et al., et al., “Human-induced pluripotent stem cell-derived cardiomyocytes exhibit temporal changes in phenotype,” American Journal of Physiology, vol. 305, no. 6, pp. H913–H922, 2013.
[263]
P. Jakob and U. Landmesser, “Role of microRNAs in stem/progenitor cells and cardiovascular repair,” Cardiovascular Research, vol. 93, no. 4, pp. 614–622, 2012.
[264]
M. Quattrocelli, G. Palazzolo, I. Agnolin et al., “Synthetic sulfonyl-hydrazone-1 positively regulates cardiomyogenic microRNA expression and cardiomyocyte differentiation of induced pluripotent stem cells,” Journal of Cellular Biochemistry, vol. 112, no. 8, pp. 2006–2014, 2011.
[265]
N. Cao, Z. Liu, Z. Chen et al., “Ascorbic acid enhances the cardiac differentiation of induced pluripotent stem cells through promoting the proliferation of cardiac progenitor cells,” Cell Research, vol. 22, no. 1, pp. 219–236, 2012.
[266]
S. S. Nunes, J. W. Miklas, J. Liu et al., “Biowire: a platform for maturation of human pluripotent stem cell-derived cardiomyocytes,” Nature Methods, vol. 10, no. 8, pp. 781–787, 2013.
[267]
A. Nsair, K. Schenke-Layland, B. van Handel et al., “Characterization and therapeutic potential of induced pluripotent stem cell-derived cardiovascular progenitor cells,” PLoS ONE, vol. 7, no. 10, Article ID e45603, 2012.
[268]
P. E. De Almeida, J. D. Ransohoff, A. Nahid, and J. C. Wu, “Immunogenicity of pluripotent stem cells and their derivatives,” Circulation Research, vol. 112, no. 3, pp. 549–561, 2013.
[269]
T. Zhao, Z.-N. Zhang, Z. Rong, and Y. Xu, “Immunogenicity of induced pluripotent stem cells,” Nature, vol. 474, no. 7350, pp. 212–216, 2011.
[270]
Z. Liu, X. Wen, H. Wang et al., “Molecular imaging of induced pluripotent stem cell immunogenicity with in vivo development in ischemic myocardium,” PLoS ONE, vol. 8, no. 6, Article ID e66369, 2013.
[271]
S. Kaneko and S. Yamanaka, “To be immunogenic, or not to be: that's the iPSC question,” Cell Stem Cell, vol. 12, no. 4, pp. 385–386, 2013.
[272]
R. Araki, M. Uda, Y. Hoki et al., “Negligible immunogenicity of terminally differentiated cells derived from induced pluripotent or embryonic stem cells,” Nature, vol. 494, no. 7435, pp. 100–104, 2013.
[273]
P. Guha, J. W. Morgan, G. Mostoslavsky, N. P. Rodrigues, and A. S. Boyd, “Lack of immune response to differentiated cells derived from syngeneic induced pluripotent stem cells,” Cell Stem Cell, vol. 12, no. 4, pp. 407–412, 2013.
[274]
K. M. Dhodapkar, D. Feldman, P. Matthews et al., “Natural immunity to pluripotency antigen OCT4 in humans,” Proceedings of the National Academy of Sciences of the United States of America, vol. 107, no. 19, pp. 8718–8723, 2010.
[275]
M. Rao, L. Ahrlund-Richter, and D. S. Kaufman, “Concise review: cord blood banking, transplantation and induced pluripotent stem cell: success and opportunities,” Stem Cells, vol. 30, no. 1, pp. 55–60, 2012.
[276]
C. J. Taylor, E. M. Bolton, and J. A. Bradley, “Immunological considerations for embryonic and induced pluripotent stem cell banking,” Philosophical Transactions of the Royal Society B: Biological Sciences, vol. 366, no. 1575, pp. 2312–2322, 2011.
[277]
K. Okita, Y. Matsumura, Y. Sato et al., “A more efficient method to generate integration-free human iPS cells,” Nature Methods, vol. 8, no. 5, pp. 409–412, 2011.
[278]
C. J. Taylor, S. Peacock, A. N. Chaudhry, J. A. Bradley, and E. M. Bolton, “Generating an iPSC bank for HLA-matched tissue transplantation based on known donor and recipient hla types,” Cell Stem Cell, vol. 11, no. 2, pp. 147–152, 2012.
[279]
J. I. Pearl, A. S. Lee, D. B. Leveson-Gower et al., “Short-term immunosuppression promotes engraftment of embryonic and induced pluripotent stem cells,” Cell Stem Cell, vol. 8, no. 3, pp. 309–317, 2011.