Modeling Cardiac Disease Mechanisms Using Induced Pluripotent Stem Cell-Derived Cardiomyocytes: Progress, Promises and Challenges
Abstract
:1. Introduction
2. Human Pluripotent Stem Cells: Applications, Power, and Limitations
3. iPSCs and Genome Editing
4. Differentiation of Cardiac Cells from iPSCs
5. Drug Discovery and Personalized Medicine
6. Cardiac Regenerative Medicine
7. iPSCs in Cardiac Disease Modeling
7.1. Long QT Syndrome
7.2. Leopard Syndrome
7.3. Catecholaminergic Polymorphic Ventricular Tachycardia
7.4. Arrhythmogenic Right Ventricular Cardiomyopathy
7.5. Restrictive Cardiomyopathy
7.6. Dilated Cardiomyopathy
7.7. Left Ventricular Non-Compaction
7.8. Hypertrophic Cardiomyopathy
8. 3D Platforms for hiPSC-Cardiomyocytes-Based Cardiac Model Disease
9. Conclusions
Author Contributions
Funding
Conflicts of Interest
Abbreviations
CVD | (Cardiovascular Disease) |
iPSCs | (Induced Pluripotent Stem Cells) |
HF | (Heart Failure) |
I/R | (Ischemic/Reperfusion) |
MI | (Myocardial Infarction) |
hPSCs | (Human Pluripotent Stem Cells) |
ESCs | (Embryonic Stem Cells) |
CMs | (Cardiomyocytes) |
iPSC-CMs | (Induced Pluripotent Stem Cells-derived Cardiomyocytes) |
ICM | (Inner Cell Mass) |
ZNFs | (Zinc-Finger Nucleases) |
TALENs | (Transcription-Activator Like Effector Nucleases) |
CRISPR-Cas9 | (Clustered Regularly Interspaced Short Palindromic Repeats-associated protein 9) |
DSBs | (Double-Strand DNA Breaks) |
NHEJ | (Nonhomologous End Joining) |
HR | (Homologous Recombination) |
DCM | (Dilated Cardiomyopathy) |
BTHS | (Barth Syndrome) |
LQTS | (Long QT Syndrome) |
BS | (Brugada Syndrome) |
LVNC | (Left Ventricular Non-Compaction) |
END-2 | (Endoderm-like cells) |
EB | (Embryod Body) |
BMPs | (Bone Morphogenetic Proteins) |
CPCs | (Cardiac Progenitor Cells) |
SMCs | (Smooth Muscle Cells) |
CAR-T | (Chimeric Antigen Receptor T cell) |
GWAS | (Genome-Wide Association Studies) |
NGS | (Next Generation Sequencing) |
ALS | (Amyotrophic Lateral Sclerosis) |
TKIs | (Twenty-one Tyrosine Kinase Inhibitors) |
VEGF | (Vascular Endothelial Growth Factor) |
PDGF | (Platelet-Derived Growth Factor) |
HLA | (Human Leukocyte Antigen) |
References
- Flora, G.D.; Nayak, M.K. A Brief Review of Cardiovascular Diseases, Associated Risk Factors and Current Treatment Regimes. Curr. Pharm. Des. 2019, 25, 4063–4084. [Google Scholar] [CrossRef] [PubMed]
- Chiong, M.; Wang, Z.V.; Pedrozo, Z.; Cao, D.J.; Troncoso, R.; Ibacache, M.; Criollo, A.; Nemchenko, A.; Hill, J.A.; Lavandero, S. Cardiomyocyte death: Mechanisms and translational implications. Cell Death Dis. 2011, 2, e244. [Google Scholar] [CrossRef] [PubMed]
- Bergmann, O.; Bhardwaj, R.D.; Bernard, S.; Zdunek, S.; Barnabé-Heider, F.; Walsh, S.; Zupicich, J.; Alkass, K.; Buchholz, B.A.; Druid, H.; et al. Evidence for Cardiomyocyte Renewal in Humans. Science 2009, 324, 98–102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Justice, M.J.; Dhillon, P. Using the mouse to model human disease: Increasing validity and reproducibility. Dis. Model. Mech. 2016, 9, 101–103. [Google Scholar] [CrossRef] [Green Version]
- Dvash, T.; Sharon, N.; Yanuka, O.; Benvenisty, N. Molecular Analysis of LEFTY-Expressing Cells in Early Human. Stem Cells 2007, 25, 465–472. [Google Scholar] [CrossRef]
- Parrotta, E.I.; Scalise, S.; Scaramuzzino, L.; Cuda, G. Stem Cells: The Game Changers of Human Cardiac Disease Modelling and Regenerative Medicine. Int. J. Mol. Sci. 2019, 20, 5760. [Google Scholar] [CrossRef] [Green Version]
- Takahashi, K.; Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006, 126, 663–676. [Google Scholar] [CrossRef] [Green Version]
- Takahashi, K.; Tanabe, K.; Ohnuki, M.; Narita, M.; Ichisaka, T.; Tomoda, K. Induction of Pluripotent Stem Cells from Adult Human Fibroblasts by Defined Factors. Cell 2007, 131, 861–872. [Google Scholar] [CrossRef] [Green Version]
- Yu, J.; Vodyanik, M.A.; Smuga-Otto, K.; Antosiewicz-Bourget, J.; Frane, J.L.; Tian, S.; Nie, J.; Jonsdottir, G.A.; Ruotti, V.; Stewart, R.; et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 2007, 318, 1917–1920. [Google Scholar] [CrossRef] [PubMed]
- Park, I.H.; Arora, N.; Huo, H.; Maherali, N.; Ahfeldt, T.; Shimamura, A.; Lensch, M.W.; Cowan, C.; Hochedlinger, K.; Daley, G.Q. Disease-Specific Induced Pluripotent Stem (iPS) Cells. Cell 2008, 134, 877–886. [Google Scholar] [CrossRef] [Green Version]
- Shi, Y.; Inoue, H.; Wu, J.C.; Yamanaka, S. Induced pluripotent stem cell technology: A decade of progress. Nat. Rev. Drug Discov. 2017, 16, 115–130. [Google Scholar] [CrossRef] [PubMed]
- Thomson, J.A. Embryonic stem cell lines derived from human blastocysts. Science 1998, 282, 1145–1147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wobus, A.M.; Boheler, K.R. Embryonic Stem Cells: Prospects for Developmental Biology and Cell Therapy. Phiyiological Rev. 2005, 85, 635–678. [Google Scholar] [CrossRef] [PubMed]
- Pickering, S. Generation of a human embryonic stem cell line encoding the cystic fibrosis mutation ∆ F508, using preimplantation genetic diagnosis. Reprod. Biomed. Online 2005, 10, 390–397. [Google Scholar] [CrossRef]
- Parrotta, E.; De Angelis, M.T.; Scalise, S.; Candeloro, P.; Santamaria, G.; Paonessa, M.; Coluccio, M.L.; Perozziello, G.; De Vitis, S.; Sgura, A.; et al. Two sides of the same coin? Unraveling subtle differences between human embryonic and induced pluripotent stem cells by Raman spectroscopy. Stem Cell Res. Ther. 2017, 8, 271. [Google Scholar] [CrossRef] [Green Version]
- Chin, M.H.; Mason, M.J.; Xie, W.; Volinia, S.; Singer, M.; Peterson, C.; Ambartsumyan, G.; Aimiuwu, O.; Richter, L.; Zhang, J.; et al. Induced pluripotent stem cells and embryonic stem cells are distinguished by gene expression signatures. Cell Stem Cell 2009, 5, 111–123. [Google Scholar] [CrossRef] [Green Version]
- Parrotta, E.I.; Scalise, S.; Taverna, D.; De Angelis, M.T.; Sarro, G.; Gaspari, M.; Santamaria, G.; Cuda, G. Comprehensive proteogenomic analysis of human embryonic and induced pluripotent stem cells. J. Cell. Mol. Med. 2019, 23, 5440–5453. [Google Scholar] [CrossRef] [Green Version]
- Mayshar, Y.; Ben-david, U.; Lavon, N.; Biancotti, J.; Yakir, B.; Clark, A.T.; Plath, K.; Lowry, W.E.; Benvenisty, N. Identification and Classification of Chromosomal Aberrations in Human Induced Pluripotent Stem Cells. Stem Cell 2010, 7, 521–531. [Google Scholar] [CrossRef] [Green Version]
- Polo, J.M.; Liu, S.; Figueroa, M.E.; Kulalert, W.; Eminli, S.; Tan, K.Y.; Apostolou, E.; Stadtfeld, M.; Li, Y.; Shioda, T.; et al. Cell type of origin influences the molecular and functional properties of mouse induced pluripotent stem cells. Nat. Biotechnol. 2010, 28, 848–857. [Google Scholar] [CrossRef] [Green Version]
- Guha, P.; Morgan, J.W.; Mostoslavsky, G.; Rodrigues, N.P.; Boyd, A.S. Lack of Immune Response to Differentiated Cells Derived from Syngeneic Induced Pluripotent Stem Cells. Stem Cell 2013, 12, 407–412. [Google Scholar] [CrossRef] [Green Version]
- Ramalingam, S.; London, V.; Kandavelou, K.; Cebotaru, L.; Guggino, W.; Civin, C.; Chandrasegaran, S. Generation and Genetic Engineering of hiPSCs Using Designed Zinc Finger Nucleases. Stem Cells Dev. 2013, 22, 595–610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ramalingam, S.; Annaluru, N.; Kandavelou, K.; Chandrasegaran, S. TALEN-mediated Generation and Genetic Correction of Disease-Specific Human Induced Pluripotent Stem Cells. Curr. Gene Ther. 2014, 14, 461–472. [Google Scholar] [CrossRef] [PubMed]
- Bruntraeger, M.; Byrne, M.; Long, K.; Bassett, A.R. Editing the genome of Human Induced Pluripotent Stem Cells using CRISPR/Cas9 Ribonucleoprotein Complexes. Methods Mol. Biol. 2019, 1961, 153–183. [Google Scholar] [PubMed]
- McDermott-Roe, C.; Lv, W.; Maximova, T.; Wada, S.; Bukowy, J.; Marquez, M.; Lai, S.; Shehu, A.; Benjamin, I.; Geurts, A.; et al. Investigation of a dilated cardiomyopathy–associated variant in BAG3 using genome-edited iPSC-derived cardiomyocytes. JCI Insight 2019, 4, e128799. [Google Scholar] [CrossRef] [PubMed]
- Wang, G.; McCain, M.L.; Yang, L.; He, A.; Pasqualini, F.S.; Agarwal, A.; Yuan, H.; Jiang, D.; Zhang, D.; Zangi, L.; et al. Modeling the mitochondrial cardiomyopathy of Barth syndrome with iPSC and heart-on-chip technologies. J. Musculoskelet. Neuronal Interact. 2014, 15, 1–9. [Google Scholar]
- Garg, P.; Oikonomopoulos, A.; Chen, H.; Li, Y.; Lam, C.K.; Sallam, K.; Perez, M.; Lux, R.L.; Sanguinetti, M.C.; Wu, J.C. Genome Editing of Induced Pluripotent Stem Cells to Decipher Cardiac Channelopathy Variant. J. Am. Coll. Cardiol. 2018, 72, 62–75. [Google Scholar] [CrossRef]
- De la Roche, J.; Angsutararux, P.; Kempf, H.; Janan, M.; Bolesani, E.; Thiemann, S.; Wojciechowski, D.; Co, M.; Franke, A.; Schwanke, K.; et al. Comparing human iPSC- cardiomyocytes versus HEK293T cells unveils disease-causing effects of Brugada mutation A735V of NaV 1.5 sodium channels. Sci. Rep. 2019, 9, 11173. [Google Scholar] [CrossRef] [Green Version]
- Kodo, K.; Ong, S.G.; Jahanbani, F.; Termglinchan, V.; Hirono, K.; InanlooRahatloo, K.; Ebert, A.D.; Shukla, P.; Abilez, O.J.; Churko, J.M.; et al. iPSC-derived cardiomyocytes reveal abnormal TGF-β signalling in left ventricular non-compaction cardiomyopathy. Nat. Cell Biol. 2016, 18, 1031–1042. [Google Scholar] [CrossRef] [Green Version]
- Rodriguez, E. Ethical Issues in Genome Editing using Crispr/Cas9 System. Clin. Res. Bioeth. 2016, 7, 2. [Google Scholar]
- Brokowski, C.; Adli, M. CRISPR Ethics: Moral Considerations for Applications of a Powerful Tool. J. Mol. Biol. 2018, 431, 88–101. [Google Scholar] [CrossRef]
- Yang, L.; Soonpaa, M.H.; Adler, E.D.; Roepke, T.K.; Kattman, S.J.; Kennedy, M.; Henckaerts, E.; Bonham, K.; Abbott, G.W.; Linden, R.M.; et al. Human cardiovascular progenitor cells develop from a KDR+ embryonic-stem-cell-derived population. Nature 2008, 453, 524–528. [Google Scholar] [CrossRef] [PubMed]
- Arai, A.; Yamamoto, K.; Toyama, J. Murine Cardiac Progenitor Cells Require Visceral Embryonic Endoderm and Primitive Streak. Dev. Dyn. 1997, 210, 344–353. [Google Scholar] [CrossRef]
- Mummery, C.; Oostwaard, D.W.; Doevendans, P.; Spijker, R.; Van Den Brink, S.; Hassink, R.; Van Der Heyden, M.; Opthof, T.; Pera, M.; Brutel, A.; et al. Differentiation of Human Embryonic Stem Cells to Cardiomyocytes. Role of Coculture With Visceral Endoderm-Like Cells. Circulation 2002, 107, 2733–2740. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Freund, C.; Davis, R.P.; Gkatzis, K.; Oostwaard, D.W.; Mummery, C.L. The first reported generation of human induced pluripotent stem cells (iPS cells) and iPS cell-derived cardiomyocytes in the Netherlands. Netherlands Hear. J. 2010, 18, 51–54. [Google Scholar]
- Passier, R.; Oostwaard, D.W.; Snapper, J.; Kloots, J.; Hassink, R.J.; Kuijk, E.; Roelen, B.; De, B. Increased Cardiomyocyte Differentiation from Human Embryonic Stem Cells in Serum-Free Cultures. Stem Cells 2005, 23, 772–780. [Google Scholar] [CrossRef] [PubMed]
- Doetschman, T.C.; Eistetter, H.; Katz, M.; Schmidt, W.; Kemler, R. The in vitro development of blastocyst-derived embryonic stem cell lines: Formation of visceral yolk sac, blood islands and myocardium. Development 1985, 87, 27–45. [Google Scholar] [PubMed]
- Osafune, K.; Caron, L.; Borowiak, M.; Martinez, R.J.; Fitz-gerald, C.S.; Sato, Y.; Cowan, C.A.; Chien, K.R.; Melton, D.A. Marked differences in differentiation propensity among human embryonic stem cell lines. Nat. Biotechnol. 2008, 26, 313–315. [Google Scholar] [CrossRef]
- Wang, H.; Hao, J.; Hong, C.C. Cardiac Induction of Embryonic Stem Cells by a Small Molecule Inhibitor of Wnt/β-Catenin Signaling. ACS Chem. Biol. 2011, 6, 192–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Laflamme, M.A.; Chen, K.Y.; Naumova, A.V.; Muskheli, V.; Fugate, J.A.; Dupras, S.K.; Reinecke, H.; Xu, C.; Hassanipour, M.; Police, S.; et al. Cardiomyocytes derived from human embryonic stem cells in pro-survival factors enhance function of infarcted rat hearts. Nat. Biotechnol. 2007, 25, 1015–1024. [Google Scholar] [CrossRef] [PubMed]
- Chau, M.D.L.; Tuft, R.; Fogarty, K.; Bao, Z. Notch signaling plays a key role in cardiac cell differentiation. Mech. Dev. 123 2006, 123, 626–640. [Google Scholar] [CrossRef] [PubMed]
- Bastakoty, D.; Saraswati, S.; Joshi, P.; Atkinson, J.; Liu, J.; Harris, J.L.; Young, P.P. Temporary, Systemic Inhibition of the WNT/β-Catenin Pathway promotes Regenerative Cardiac Repair following Myocardial Infarct. Cell Stem Cell 2016, 2. [Google Scholar] [CrossRef] [PubMed]
- Abad, M.; Hashimoto, H.; Zhou, H.; Morales, M.G.; Chen, B.; Bassel-duby, R.; Olson, E.N. Notch Inhibition Enhances Cardiac Reprogramming by Increasing MEF2C Transcriptional Activity. Stem Cell Rep. 2017, 8, 548–560. [Google Scholar] [CrossRef]
- Bhattacharya, S.; Burridge, P.W.; Kropp, E.M.; Chuppa, S.L.; Kwok, W.-M.; Wu, J.C.; Boheler, K.R.; Gundry, R.L. High Efficiency Differentiation of Human Pluripotent Stem Cells to Cardiomyocytes and Characterization by Flow Cytometry. J. Vis. Exp. 2014, 23, 52010. [Google Scholar] [CrossRef] [Green Version]
- Paige, S.L.; Osugi, T.; Afanasiev, O.K.; Pabon, L.; Reinecke, H.; Murry, C.E. Endogenous Wnt/b-Catenin Signaling Is Required for Cardiac Differentiation in Human Embryonic Stem Cells. PLoS ONE 2010, 5, e11134. [Google Scholar] [CrossRef] [PubMed]
- Kadari, A.; Mekala, S.; Wagner, N.; Malan, D.; Köth, J.; Sasse, P.; Herzig, S.; Brüstle, O.; Ergün, S.; Edenhofer, F. Robust Generation of Cardiomyocytes from Human iPS Cells Requires Precise Modulation of BMP and WNT Signaling. Stem Cell Rev. Rep. 2015, 11, 560–569. [Google Scholar] [CrossRef] [PubMed]
- Burridge, P.W.; Matsa, E.; Shukla, P.; Lin, Z.C.; Churko, J.M.; Ebert, A.D.; Lan, F.; Diecke, S.; Huber, B.; Mordwinkin, N.M.; et al. Chemically defined generation of human cardiomyocytes. Nat. Methods 2014, 11, 855–860. [Google Scholar] [CrossRef] [Green Version]
- Zhang, J.; Klos, M.; Wilson, G.F.; Herman, A.M.; Lian, X.; Raval, K.K.; Barron, M.R.; Hou, L.; Soerens, A.G.; Yu, J.; et al. Extracellular Matrix Promotes Highly Efficient Cardiac Differentiation of Human Pluripotent Stem Cells: The Matrix Sandwich Method. Circ. Res. 2013, 111, 1125–1136. [Google Scholar] [CrossRef]
- Kattman, S.J.; Witty, A.D.; Gagliardi, M.; Dubois, N.C.; Niapour, M.; Hotta, A.; Ellis, J.; Keller, G. Stage-Specific Optimization of Activin / Nodal and BMP Signaling Promotes Cardiac Differentiation of Mouse and Human Pluripotent Stem Cell Lines. Cell Stem Cell 2011, 8, 228–240. [Google Scholar] [CrossRef] [Green Version]
- Cao, N.; Liang, H.; Huang, J.; Wang, J.; Chen, Y.; Chen, Z.; Yang, H. Highly efficient induction and long-term maintenance of multipotent cardiovascular progenitors from human pluripotent stem cells under defined conditions. Cell Res. 2013, 23, 1119–1132. [Google Scholar] [CrossRef]
- Lalit, P.A.; Salick, M.R.; Nelson, D.O.; Squirrell, J.M.; Christina, M.; Patel, N.G.; Saeed, I.; Schmuck, E.G.; Markandeya, Y.S.; Wong, R.; et al. Lineage Reprogramming of Fibroblasts to Proliferative Induced Cardiac Progenitor Cells by Defined Factors. Cell Stem Cell 2017, 18, 354–367. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.; Cao, N.; Huang, Y.; Spencer, C.I.; Fu, J.; Yu, C.; Liu, K.; Nie, B.; Xu, T.; Li, K.; et al. Expandable Cardiovascular Progenitor Cells Reprogrammed from Fibroblasts. Cell Stem Cell 2018, 18, 368–381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lian, X.; Hsiao, C.; Wilson, G.; Zhu, K.; Hazeltine, L.B.; Azarin, S.M.; Raval, K.K. Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling. Proc. Natl. Acad. Sci. USA 2012, 109, E1848–E1857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Veerman, C.C.; Kosmidis, G.; Mummery, C.L.; Casini, S.; Verkerk, A.O.; Bellin, M. Immaturity of Human Stem-Cell-Derived Cardiomyocytes in Culture: Fatal Flaw or Soluble Problem? Stem Cells Dev. 2015, 24, 1035–1052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dubois, N.C.; Craft, A.M.; Sharma, P.; Elliott, D.A.; Stanley, E.G.; Elefanty, A.G.; Gramolini, A.; Keller, G. SIRPA is a specific cell-surface marker for isolating cardiomyocytes derived from human pluripotent stem cells. Nat. Biotechnol. 2011, 29, 1011–1019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Elliot, D.A.; Braam, S.R.; Koutsis, K.; Ng, E.S.; Jenny, R.; Lagerqvist, E.L.; Biben, C.; Hatzistavrou, T.; Hirst, C.E.; Yu, Q.C.; et al. NKX2-5eGFP/w hESCs for isolation of human cardiac progenitors and cardiomyocytes. Nat. Methods 2011, 8, 1037–1040. [Google Scholar] [CrossRef]
- Uosaki, H.; Fukushima, H.; Takeuchi, A.; Matsuoka, S.; Nakatsuji, N.; Yamanaka, S.; Yamashita, J.K. Efficient and Scalable Purification of Cardiomyocytes from Human Embryonic and Induced Pluripotent Stem Cells by VCAM1 Surface Expression. PLoS ONE 2011, 6, e23657. [Google Scholar] [CrossRef]
- Weng, Z.; He, J.; Zi, M.; Chow, Y.; Mok, C.F.; Keung, W.; Chow, H.; Leung, A.Y.H.; Hajjar, R.J.; Li, R.A.; et al. A Simple, Cost-Effective but Highly Efficient System for Deriving Ventricular Cardiomyocytes from Human Pluripotent Stem Cells. Stem Cells Dev. 2014, 23, 1704–1716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cyganek, L.; Tiburcy, M.; Sekeres, K.; Gerstenberg, K.; Bohnenberger, H.; Lenz, C.; Henze, S.; Stauske, M.; Salinas, G.; Zimmermann, W.; et al. Deep phenotyping of human induced pluripotent stem cell—Derived atrial and ventricular cardiomyocytes. JCI Insight 2018, 3, e99941. [Google Scholar] [CrossRef] [Green Version]
- Argenziano, M.; Lambers, E.; Hong, L.; Sridhar, A.; Zhang, M.; Chalazan, B.; Menon, A.; Savio-galimberti, E.; Wu, J.C.; Rehman, J. Electrophysiologic Characterization of Calcium Handling in Human Induced Pluripotent Stem Cell-Derived Atrial Cardiomyocytes. Stem Cell Rep. 2018, 10, 1867–1878. [Google Scholar] [CrossRef]
- Zhang, J.; Wilson, G.F.; Soerens, A.G.; Koonce, C.H.; Yu, J.; Sean, P.; Thomson, J.A.; Kamp, T.J. Functional Cardiomyocytes Derived from Human Induced Pluripotent Stem Cells. Circ. Res. 2009, 104, e30–e41. [Google Scholar] [CrossRef] [Green Version]
- Schweizer, P.A.; Darche, F.F.; Ullrich, N.D.; Geschwill, P.; Greber, B.; Rivinius, R.; Seyler, C.; Müller-decker, K.; Draguhn, A.; Utikal, J.; et al. Subtype-specific differentiation of cardiac pacemaker cell clusters from human induced pluripotent stem cells. Stem Cell Res. Ther. 2017, 8, 229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Machiraju, P.; Greenway, S.C. Current methods for the maturation of induced pluripotent stem cell derived cardiomyocytes. World J. Stem Cells 2019, 11, 33–43. [Google Scholar] [CrossRef] [PubMed]
- Lundy, S.D.; Zhu, W.-Z.; Regnier, M.; Laflamme, M.A. Structural and Functional Maturation of Cardiomyocytes Derived From Human Pluripotent Stem Cells. Stem Cells Dev. 2013, 22, 1991–2002. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ebert, A.; Joshi, A.U.; Andorf, S.; Dai, Y.; Sampathkumar, S.; Chen, H.; Li, Y.; Garg, P.; Toischer, K.; Hasenfuss, G.; et al. Proteasome-Dependent Regulation of Distinct Metabolic States During Long-Term Culture of Human. Circ. Res. 2019, 125, 90–103. [Google Scholar] [CrossRef]
- Yang, X.; Rodriguez, M.; Pabon, L.; Fischer, K.A.; Reinecke, H.; Regnier, M.; Sniadecki, N.J.; Ruohola-baker, H.; Murry, C.E. Tri-iodo-L-thyronine promotes the maturation of human cardiomyocytes-derived from induced pluripotent stem cells. J. Mol. Cell. Cardiol. 2014, 72, 296–304. [Google Scholar] [CrossRef] [Green Version]
- Parikh, S.S.; Blackwell, D.J.; Gomez-hurtado, N.; Frisk, M.; Wang, L.; Kim, K.; Dahl, C.P.; Fiane, A.; Tønnessen, T.; Kryshtal, D.O.; et al. Thyroid and Glucocorticoid Hormones Promote Functional T-Tubule Development in Human-Induced Pluripotent Stem Cell–Derived Cardiomyocytes. Circ. Res. 2017, 121, 1323–1330. [Google Scholar] [CrossRef]
- Ruan, J.-L.; Tulloch, N.L.; Razumova, M.V.; Saiget, M.; Muskheli, V.; Pabon, L.; Reinecke, H.; Regnier, M.; Murry, C.E. Mechanical stress conditioning and electrical stimulation Promote contractility and Force Maturation of induced Pluripotent stem cell-Derived human cardiac tissue. Circulation 2016, 134, 1557–1567. [Google Scholar] [CrossRef]
- Kadota, S.; Pabon, L.; Reinecke, H.; Murry, C.E. In Vivo Maturation of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes in Neonatal and Adult Rat Hearts Shin. Stem Cell Rep. 2017, 8, 278–289. [Google Scholar] [CrossRef]
- Cho, G.-S.; Lee, D.I.; Tampakakis, E.; Murphy, S.; Andersen, P.; Uosaki, H.; Chelko, S.; Chakir, K.; Hong, I.; Seo, K.; et al. Neonatal Transplantation Confers Maturation of PSC-Derived Cardiomyocytes Conducive to Modeling Cardiomyopathy. Cell Rep. 2017, 18, 571–582. [Google Scholar] [CrossRef] [Green Version]
- Chandarana, M.; Curtis, A.; Hoskins, C. The use of nanotechnology in cardiovascular disease. Appl. Nanosci. 2018, 8, 1607–1619. [Google Scholar] [CrossRef] [Green Version]
- Dattola, E.; Parrotta, I.; Scalise, S.; Perozziello, G.; Limongi, T.; Candeloro, P.; Coluccio, L.; Maletta, C.; Bruno, L.; De Angelis, T.; et al. Development of 3D PVA scaffolds for cardiac tissue engineering and cell screening applications. RSC Adv. 2019, 9, 4246–4257. [Google Scholar] [CrossRef] [Green Version]
- Khorshidi, S.; Solouk, A.; Mirzadeh, H.; Mazinani, S.; Lagaron, J.M.; Shari, S.; Ramakrishna, S. A review of key challenges of electrospun scaffolds for tissue-engineering applications. J. Tissue Eng. Regen. Med. 2016, 10, 715–738. [Google Scholar] [CrossRef] [PubMed]
- Stout, D.A.; Basu, B.; Webster, T.J. Poly (lactic—co-glycolic acid): Carbon nanofiber composites for myocardial tissue engineering applications. Acta Biomater. 2011, 7, 3101–3112. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.; Zeng, D.; Ding, L.; Li, X.; Liu, X.; Li, W.; Wei, T.; Yan, S.; Xie, J.; Wei, L.; et al. Three-dimensional poly-(ε-caprolactone) nanofibrous scaffolds directly promote the cardiomyocyte differentiation of murine- induced pluripotent stem cells through Wnt/β-catenin signaling. BMC Cell Biol. 2015, 16, 22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fong, A.H.; Romero-Lopez, M.; Heylman, C.M.; Keating, M.; Tran, D.; Sobrino, A.; Tran, A.Q.; Pham, H.H.; Fimbres, C.; Gershon, P.D.; et al. Three-Dimensional Adult Cardiac Extracellular Matrix Promotes Maturation of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Tissue Eng. 2016, 22, 1016–1025. [Google Scholar] [CrossRef] [Green Version]
- Kroll, K.; Chabria, M.; Wang, K.; Hausermann, F.; Schuler, F.; Polonchuk, L. Electro-mechanical conditioning of human iPSC-derived cardiomyocytes for translational research. Prog. Biophys. Mol. Biol. 2017, 130, 212–222. [Google Scholar] [CrossRef]
- Gerbin, K.A.; Murry, C.E. The winding road to regenerating the human heart. Cardiovasc. Pathol. 2016, 24, 133–140. [Google Scholar] [CrossRef] [Green Version]
- Zweigerdt, R.; Olmer, R.; Singh, H.; Haverich, A.; Martin, U. Scalable expansion of human pluripotent stem cells in suspension culture. Nat. Protoc. 2011, 6, 689–700. [Google Scholar] [CrossRef]
- Ismadi, M.; Gupta, P.; Fouras, A.; Verma, P.; Jadhav, S. Flow Characterization of a Spinner Flask for Induced Pluripotent Stem Cell Culture Application. PLoS ONE 2014, 9, e106493. [Google Scholar] [CrossRef]
- Chen, V.C.; Ye, J.; Shukla, P.; Hua, G.; Chen, D.; Lin, Z.; Liu, J.; Chai, J.; Gold, J.; Wu, J.; et al. Development of a scalable suspension culture for cardiac differentiation from human pluripotent stem cells. Stem Cell Rep. 2015, 15, 365–375. [Google Scholar] [CrossRef]
- Badenes, S.M.; Fernandes, T.G.; Cordeiro, C.S.M.; Boucher, S.; Kuninger, D.; Vemuri, M.C.; Diogo, M.M.; Cabral, J.M.S. Defined Essential 8 TM Medium and Vitronectin Efficiently Support Scalable Xeno-Free Expansion of Human Induced Pluripotent Stem Cells in Stirred Microcarrier Culture Systems. PLoS ONE 2016, 11, e0151264. [Google Scholar]
- Laco, F.; Lam, A.T.; Woo, T.; Tong, G.; Ho, V.; Soong, P.; Grishina, E.; Lin, K.; Reuveny, S.; Oh, S.K. Selection of human induced pluripotent stem cells lines optimization of cardiomyocytes differentiation in an integrated suspension microcarrier bioreactor. Stem Cell Res. Ther. 2020, 11, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Herrmann, J. Adverse cardiac effects of cancer therapies: Cardiotoxicity and arrhythmia [published online ahead of print, 2020 Mar 30]. Nat. Rev. Cardiol. 2020. [Google Scholar] [CrossRef]
- Kernik, D.C.; Morotti, S.; Wu, H.; Garg, P.; Duff, H.J.; Kurokawa, J. A computational model of induced pluripotent stem-cell derived cardiomyocytes incorporating experimental variability from multiple data sources. J. Physiol. 2019, 597, 4533–4564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wainger, B.J.; Kiskinis, E.; Mellin, C.; Wiskow, O.; Han, S.S.W.; Sandoe, J.; Perez, N.P.; Williams, L.A.; Lee, S.; Boulting, G.; et al. Intrinsic Membrane Hyperexcitability of Amyotrophic Lateral Sclerosis Patient-Derived Motor Neurons. Cell Rep. 2014, 7, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fiedler, L.R.; Chapman, K.; Xie, M.; Tralau-stewart, C.; Perrior, T.; Schneider, M.D. MAP4K4 Inhibition Promotes Survival of Human Stem Cell-Derived Cardiomyocytes and Reduces Infarct Size In Vivo Article MAP4K4 Inhibition Promotes Survival of Human Stem Cell-Derived Cardiomyocytes and Reduces Infarct Size In Vivo. Cell Stem Cell 2019, 24, 579–591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Obergrussberger, A.; Haarmann, C.; Stölzle-feix, S.; Becker, N.; Ohtsuki, A.; Brüggemann, A.; George, M.; Fertig, N. Automated Patch Clamp Recordings of Human Stem Cell-Derived Cardiomyocytes. In Stem Cell-Derived Models in Toxicology; Springer: New York, NY, USA, 2019; pp. 57–82. ISBN 9781493966615. [Google Scholar]
- Asakura, K.; Hayashi, S.; Ojima, A.; Taniguchi, T.; Miyamoto, N.; Nakamor, C.; Nagasawa, C.; Kitamur, T.; Osada, T.; Honda, Y.; et al. Improvement of acquisition and analysis methods in multi-electrode array experiments with iPS cell-derived cardiomyocytes. J. Pharmacol. Toxicol. Methods 2015, 75, 17–26. [Google Scholar] [CrossRef] [Green Version]
- Sirenko, O.; Crittenden, C.; Callamaras, N.; Hesley, J.; Chen, Y.; Funes, C.; Rusyn, I.; Anson, B.; Cromwell, E.F. Multiparameter In Vitro Assessment of Compound Effects on Cardiomyocyte Physiology Using iPSC Cells. J. Biomol. Screen. 2013, 18, 39–53. [Google Scholar] [CrossRef] [Green Version]
- Sheehy, S.P.; Pasqualini, F.; Grosberg, A.; Park, S.J.; Aratyn-Schaus, Y.; Parker, K.K. Quality Metrics for Stem Cell-Derived Cardiac Myocytes. Stem Cell Rep. 2014, 2, 282–294. [Google Scholar] [CrossRef] [Green Version]
- Banerjee, I.; Carrion, K.; Serrano, R.; Dyo, J.; Sasik, R.; Lund, S.; Willems, E.; Aceves, S.; Meili, R.; Mercola, M.; et al. Cyclic stretch of Embryonic Cardiomyocytes Increases Proliferation, Growth, and Expression While Repressing Tgf-β Signaling. J. Mol. Cell. Cardiol. 2015, 79, 133–144. [Google Scholar] [CrossRef] [Green Version]
- Sharma, A.; McKeithan, W.L.; Serrano, R.; Kitani, T.; Burridge, P.W.; Álamo, J.C.; Mercola, M.; Wu, J.C. Use of human induced pluripotent stem cell-derived cardiomyocytes to assess drug cardiotoxicity. Nat. Protoc. 2019, 13, 3018–3041. [Google Scholar] [CrossRef] [PubMed]
- Burridge, P.W.; Li, Y.F.; Matsa, E.; Wu, H.; Ong, S.; Sharma, A.; Holmström, A.; Chang, A.C.; Coronado, M.J.; Ebert, A.D.; et al. Human Induced Pluripotent Stem Cell–Derived Cardiomyocytes Recapitulate the Predilection of Breast Cancer Patients to Doxorubicin–Induced Cardiotoxicity. Nat. Med. 2016, 22, 547–556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chaudhari, U.; Nemade, H.; Wagh, V.; Gaspar, J.A.; Ellis, J.K.; Srinivasan, S.P.; Spitkovski, D.; Nguemo, F.; Louisse, J.; Bremer, S.; 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, 2763–2777. [Google Scholar] [CrossRef] [PubMed]
- Sharma, A.; Burridge, P.W.; Mckeithan, W.L.; Serrano, R.; Shukla, P.; Sayed, N.; Churko, J.M.; Kitani, T.; Wu, H.; Holmström, A.; et al. High-Throughput Screening of Tyrosine Kinase Inhibitor Cardiotoxicity with Human Induced Pluripotent Stem Cells. Sci. Transl. Med. 2017, 9, eaaf2584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ewer, M.S.; Vooletich, M.T.; Durand, J.; Woods, M.L.; Davis, J.R.; Valero, V.; Lenihan, D.J. Reversibility of Trastuzumab-Related Cardiotoxicity: New Insights Based on Clinical Course and Response to Medical Treatment. J. Clin. Oncol. 2005, 23, 7820–7826. [Google Scholar] [CrossRef]
- Kitani, T.; Ong, S.-G.; Lam, C.K.; Rhee, J.-W.; Zhang, J.Z.; Oikonomopoulos, A.; Ma, N.; Tian, L.; Lee, J.; Telli, M.L.; et al. Human-Induced Pluripotent Stem Cell Model of Trastuzumab-Induced Cardiac Dysfunction in Patients With Breast Cancer. Circulation 2019, 139, 2451–2465. [Google Scholar] [CrossRef]
- Mehta, A.; Ramachandra, C.J.A.; Singh, P.; Chitre, A.; Lua, C.H.; Mura, M.; Crotti, L.; Wong, P.; Schwartz, P.J.; Gnecchi, M.; et al. Identification of a targeted and testable antiarrhythmic therapy for long-QT syndrome type 2 using a patient-specific cellular model. Eur. Heart J. 2018, 39, 1446–1455. [Google Scholar] [CrossRef]
- Paik, D.T.; Chandy, M.; Wu, J.C. Patient and disease–specific induced pluripotent stem cells for discovery of personalized cardiovascular drugs and therapeutics. Pharmacol. Rev. 2020, 72, 320–342. [Google Scholar] [CrossRef] [Green Version]
- Solomon, S.; Pitossi, F.; Rao, M.S. Banking on iPSC- Is it Doable and is it Worthwhile. Stem Cell Rev. Rep. 2015, 11, 1–10. [Google Scholar] [CrossRef] [Green Version]
- Poolman, R.A.; Brooks, G. Expressions and Activities of Cell Cycle Regulatory Molecules During the Transition from Myocyte Hyperplasia to Hypertrophy. J. Mol. Cell. Cardiol. 1998, 30, 2121–2135. [Google Scholar] [CrossRef]
- Tambara, K.; Sakakibara, Y.; Sakaguchi, G.; Lu, F.; Premaratne, G.U.; Lin, X.; Nishimura, K.; Komeda, M. Transplanted Skeletal Myoblasts Can Fully Replace the Infarcted Myocardium When They Survive in the Host in Large Numbers. Circulation 2003, 108, 259–263. [Google Scholar] [CrossRef] [Green Version]
- Tse, H.; Siu, C.; Zhu, S.; Songyan, L.; Zhang, Q.; Lai, W.-H.; Kwong, Y.-L.; Nicholls, J.; Lau, C.-P. Paracrine effects of direct intramyocardial implantation of bone marrow derived cells to enhance neovascularization in chronic ischaemic myocardium. Eur. J. Heart Fail. 2007, 9, 747–753. [Google Scholar] [CrossRef]
- Kawamoto, A.; Losordo, D.W. Endothelial Progenitor Cells for Cardiovascular Regeneration. Trends Cardiovasc. Med. 2008, 18, 33–37. [Google Scholar] [CrossRef] [Green Version]
- Chen, S.; Fang, W.; Qian, J.; Ye, F.; Liu, Y.; Shan, S.; Zhang, J.; Lin, S.; Liao, L.; Zhao, R.C.H. Improvement of Cardiac Function After Transplantation of Autologous Bone Marrow Mesenchymal Stem Cells in Patients With Acute Myocardial Infarction. Chin. Med. J. Engl. 2004, 117, 1443–1448. [Google Scholar]
- Valina, C.; Pinkernell, K.; Song, Y.; Bai, X.; Sadat, S.; Campeau, R.J.; Le Jemtel, T.H.; Alt, E. Intracoronary administration of autologous adipose tissue-derived stem cells improves left ventricular function, perfusion, and remodelling after acute myocardial infarction. Eur. Heart J. 2007, 28, 2667–2677. [Google Scholar] [CrossRef]
- Makkar, R.R.; Smith, R.R.; Cheng, K.; Malliaras, K.; Thomson, L.E.J.; Berman, D.; Czer, L.S.C.; Marbán, L.; Mendizabal, A.; Johnston, P.V.; et al. Intracoronary cardiosphere-derived cells for heart regeneration after myocardial infarction (CADUCEUS): A prospective, randomised phase 1 trial. Lancet 2012, 379, 895–904. [Google Scholar] [CrossRef] [Green Version]
- Yamada, S.; Nelson, T.J.; Crespo-Diaz, R.J.; Perez-Terzic, C.; Liu, X.-K.; Miki, T.; Seino, S.; Behfar, A.; Terzic, A. Embryonic Stem Cell Therapy of Heart Failure in Genetic Cardiomyopathy. Stem Cells 2008, 26, 2644–2653. [Google Scholar] [CrossRef] [Green Version]
- Weinberger, F.; Breckwoldt, K.; Pecha, S.; Kelly, A.; Geertz, B.; Starbatty, J.; Yorgan, T.; Cheng, K.; Lessmann, K.; Stolen, T.; et al. Cardiac repair in guinea pigs with human engineered heart tissue from induced pluripotent stem cells. Sci. Transl. Med. 2016, 8, 363ra148. [Google Scholar] [CrossRef]
- Kishino, Y.; Fujita, J.; Tohyama, S.; Okada, M.; Tanosaki, S.; Someya, S.; Fukuda, K. Toward the realization of cardiac regenerative medicine using pluripotent stem cells. Inflamm. Regen. 2020, 40, 4–9. [Google Scholar] [CrossRef]
- Kawamura, M.; Miyagawa, S.; Miki, K.; Saito, A.; Fukushima, S.; Higuchi, T.; Kawamura, T.; Kuratani, T.; Daimon, T.; Shimizu, T.; et al. Feasibility, safety, and therapeutic efficacy of human induced pluripotent stem cell-derived cardiomyocyte sheets in a porcine ischemic cardiomyopathy model. Circulation 2012, 126, 29–37. [Google Scholar] [CrossRef] [Green Version]
- Shiba, Y.; Fernandes, S.; Zhu, W.; Filice, D.; Muskheli, V.; Kim, J.; Palpant, N.J.; Gantz, J.; Moyes, K.W.; Reinecke, H.; et al. Human ES-cell-derived cardiomyocytes electrically couple and suppress arrhythmias in injured hearts. Nature 2012, 489, 322–325. [Google Scholar] [CrossRef]
- Chong, J.J.H.; Yang, X.; Don, C.W.; Minami, E.; Liu, Y.W.; Weyers, J.J.; Mahoney, W.M.; Van Biber, B.; Cook, S.M.; Palpant, N.J.; et al. Human embryonic-stem-cell-derived cardiomyocytes regenerate non-human primate hearts. Nature 2014, 510, 273–277. [Google Scholar] [CrossRef]
- Shiba, Y.; Gomibuchi, T.; Seto, T.; Wada, Y.; Ichimura, H.; Tanaka, Y.; Ogasawara, T.; Okada, K.; Shiba, N.; Sakamoto, K.; et al. Allogeneic transplantation of iPS cell-derived cardiomyocytes regenerates primate hearts. Nature 2016, 538, 388–391. [Google Scholar] [CrossRef]
- Ma, J.; Guo, L.; Fiene, S.J.; Anson, B.D.; Thomson, J.A.; Kamp, T.J.; Kolaja, K.L.; Swanson, B.J.; January, C.T. High purity human-induced pluripotent stem cell-derived cardiomyocytes: Electrophysiological properties of action potentials and ionic currents. Am. J. Physiol. Hear. Circ. Physiol. 2011, 301, H2006–H2017. [Google Scholar] [CrossRef]
- Masumoto, H.; Matsuo, T.; Yamamizu, K.; Uosaki, H.; Narazaki, G.; Katayama, S.; Marui, A.; Shimizu, T.; Ikeda, T.; Okano, T.; 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 2012, 30, 1196–1205. [Google Scholar] [CrossRef] [Green Version]
- Jiang, Y.; Park, P.; Hong, S.; Ban, K. Maturation of Cardiomyocytes Derived from Human Pluripotent Stem Cells: Current Strategies and Limitations. Mol. Cells 2018, 41, 613–621. [Google Scholar]
- Caspi, O.; Huber, I.; Kehat, I.; Habib, M.; Arbel, G.; Gepstein, A.; Yankelson, L.; Aronson, D.; Beyar, R.; Gepstein, L. Transplantation of human embryonic stem cell-derived cardiomyocytes improves myocardial performance in infarcted rat hearts. J. Am. Coll. Cardiol. 2007, 50, 1884–1893. [Google Scholar] [CrossRef]
- Van Laake, L.W.; Passier, R.; Monshouwer-kloots, J.; Verkleij, A.J.; Lips, D.J.; Freund, C.; Den Ouden, K.; Oostwaard, D.W.; Korving, J.; Tertoolen, L.G.; et al. Human embryonic stem cell-derived cardiomyocytes survive and mature in the mouse heart and transiently improve function after myocardial infarction. Stem Cell Res. 2007, 1, 9–24. [Google Scholar] [CrossRef] [Green Version]
- Kawamura, M.; Miyagawa, S.; Fukushima, S.; Saito, A.; Miki, K.; Funakoshi, S.; Yoshida, Y.; Yamanaka, S.; Shimizu, T.; Okano, T.; et al. Enhanced Therapeutic Effects of Human iPS Cell Derived- Cardiomyocyte by Combined Cell-Sheets with Omental Flap Technique in Porcine Ischemic Cardiomyopathy Model. Sci. Rep. 2017, 7, 8824. [Google Scholar] [CrossRef]
- Cyranoski, D. Reprogrammed stem cells approved to mend human hearts for the first time. Nature 2018, 557, 619–620. [Google Scholar] [CrossRef] [Green Version]
- Savoji, H.; Mohammadi, M.H.; Rafatian, N.; Toroghi, M.K.; Wang, E.Y.; Zhao, Y.; Korolj, A.; Ahadian, S.; Radisic, M. Cardiovascular disease models: A game changing paradigm in drug discovery and screening. Biomaterials 2019, 198, 3–26. [Google Scholar] [CrossRef] [PubMed]
- Sala, L.; Gnecchi, M.; Schwartz, P.J. Long QT Syndrome Modelling with Cardiomyocytes Derived from Human-induced Pluripotent Stem Cells. Arrhythmia Electrophysiol. Rev. 2019, 8, 105–110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carvajal-Vergara, X.; Sevilla, A.; Dsouza, S.L.; Ang, Y.S.; Schaniel, C.; Lee, D.F.; Yang, L.; Kaplan, A.D.; Adler, E.D.; Rozov, R.; et al. Patient-specific induced pluripotent stem-cell-derived models of LEOPARD syndrome. Nature 2010, 465, 808–812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liang, P.; Sallam, K.; Wu, H.; Li, Y.; Itzhaki, I.; Garg, P.; Zhang, Y.; Termglichan, V.; Lan, F.; Gu, M.; et al. Patient-Specific and Genome-Edited Induced Pluripotent Stem Cell–Derived Cardiomyocytes Elucidate Single-Cell Phenotype of Brugada Syndrome. J. Am. Coll. Cardiol. 2016, 68, 2086–2096. [Google Scholar] [CrossRef] [PubMed]
- Itzhaki, I.; Maizels, L.; Huber, I.; Gepstein, A.; Arbel, G.; Caspi, O.; Miller, L.; Belhassen, B.; Nof, E.; Glikson, M.; et al. Modeling of Catecholaminergic Polymorphic Ventricular Tachycardia with Patient-Specific Human-Induced Pluripotent Stem Cells. J. Am. Coll. Cardiol. 2012, 60, 990–1000. [Google Scholar] [CrossRef] [Green Version]
- Caspi, O.; Huber, I.; Gepstein, A.; Arbel, G.; Maizels, L.; Boulos, M.; Gepstein, L. Modeling of arrhythmogenic right ventricular cardiomyopathy with human induced pluripotent stem cells. Circ. Cardiovasc. Genet. 2013, 6, 557–568. [Google Scholar] [CrossRef] [Green Version]
- Shah, D.; Virtanen, L.; Prajapati, C.; Kiamehr, M.; Kallio, P.; Taimen, P.; Aalto-Setälä, K. Modeling of LMNA-Related Dilated Cardiomyopathy Using Human Induced Pluripotent Stem Cells. Cells 2019, 8, 594. [Google Scholar] [CrossRef] [Green Version]
- Li, S.; Pan, H.; Tan, C.; Sun, Y.; Song, Y.; Zhang, X.; Yang, W.; Wang, X.; Li, D.; Dai, Y.; et al. Mitochondrial Dysfunctions Contribute to Hypertrophic Cardiomyopathy in Patient iPSC-Derived Cardiomyocytes with MT-RNR2 Mutation. Stem Cell Rep. 2018, 10, 808–821. [Google Scholar] [CrossRef] [Green Version]
- Kuroda, Y.; Yuasa, S.; Watanabe, Y.; Ito, S.; Egashira, T.; Seki, T.; Hattori, T.; Ohno, S.; Kodaira, M.; Suzuki, T.; et al. Flecainide ameliorates arrhythmogenicity through NCX flux in Andersen-Tawil syndrome-iPS cell-derived cardiomyocytes. Biochem. Biophys. Rep. 2017, 9, 245–256. [Google Scholar] [CrossRef]
- Yazawa, M.; Hsueh, B.; Jia, X.; Pasca, A.M.; Jonathan, A.; Bernstein, J.H.; Dolmetsch, R.E. Using iPS cells to investigate cardiac phenotypes in patients with Timothy Syndrome. Nature 2011, 471, 230–234. [Google Scholar] [CrossRef]
- Hick, A.; Wattenhofer-Donzé, M.; Chintawar, S.; Tropel, P.; Simard, J.P.; Vaucamps, N.; Gall, D.; Lambot, L.; André, C.; Reutenauer, L.; et al. Neurons and cardiomyocytes derived from induced pluripotent stem cells as a model for mitochondrial defects in Friedreich’s ataxia. Dis. Model. Mech. 2013, 6, 608–621. [Google Scholar] [CrossRef] [Green Version]
- Fatica, E.M.; DeLeonibus, G.A.; House, A.; Kodger, J.V.; Pearce, R.W.; Shah, R.R.; Levi, L.; Sandlers, Y. Barth Syndrome: Exploring Cardiac Metabolism with Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Metabolites 2019, 9, 306. [Google Scholar] [CrossRef] [Green Version]
- Sato, Y.; Kobayashi, H.; Higuchi, T.; Shimada, Y.; Era, T.; Kimura, S.; Ohashi, T.; Eto, Y.; Ida, H. Disease modeling and lentiviral gene transfer in patient-specific induced pluripotent stem cells from late-onset Pompe disease patient. Mol. Ther. Methods Clin. Dev. 2015, 2, 15023. [Google Scholar] [CrossRef] [PubMed]
- Moss, A.J. Long QT Syndrome. J. Am. Med. Assoc. 2003, 289, 2041–2044. [Google Scholar] [CrossRef]
- Tester, D.J.; Ackerman, M.J. Genetics of Long QT Syndrome. Methodist Debakey Cardiovasc. J. 2014, 10, 29–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moretti, A.; Bellin, M.; Welling, A.; Jung, C.B.; Lam, J.T.; Bott-Flügel, L.; Dorn, T.; Goedel, A.; Höhnke, C.; Hofmann, F.; et al. Patient-Specific Induced Pluripotent Stem-Cell Models for Long-QT Syndrome. N. Engl. J. Med. 2010, 363, 1397–1409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Egashira, T.; Yuasa, S.; Suzuki, T.; Aizawa, Y.; Yamakawa, H.; Matsuhashi, T.; Ohno, Y.; Tohyama, S.; Okata, S.; Seki, T.; et al. Disease characterization using LQTS-specific induced pluripotent stem cells. Cardiovasc. Res. 2012, 95, 419–429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ma, D.; Wei, H.; Lu, J.; Huang, D.; Liu, Z.; Loh, L.J.; Islam, O.; Liew, R.; Shim, W.; Cook, S.A. Characterization of a novel KCNQ1 mutation for type 1 long QT syndrome and assessment of the therapeutic potential of a novel IKs activator using patient-specific induced pluripotent stem cell-derived cardiomyocytes. Stem Cell Res. Ther. 2015, 6, 39. [Google Scholar] [CrossRef]
- Wang, Y.; Liang, P.; Lan, F.; Wu, H.; Lisowski, L.; Gu, M.; Hu, S.; Kay, M.A.; Urnov, F.D.; Shinnawi, R.; et al. Genome editing of isogenic human induced pluripotent stem cells recapitulates long QT phenotype for drug testing. J. Am. Coll. Cardiol. 2014, 64, 451–459. [Google Scholar] [CrossRef] [Green Version]
- Keller, D.I.; Grenier, J.; Christé, G.; Dubouloz, F.; Osswald, S.; Brink, M.; Ficker, E.; Chahine, M. Characterization of novel KCNH2 mutations in type 2 long QT syndrome manifesting as seizures. Can. J. Cardiol. 2009, 25, 455–462. [Google Scholar] [CrossRef] [Green Version]
- Bellin, M.; Casini, S.; Davis, R.P.; Aniello, C.D.; Haas, J.; Oostwaard, D.W.; Tertoolen, L.G.J.; Jung, C.B.; Elliott, D.A.; Welling, A.; et al. Isogenic human pluripotent stem cell pairs reveal the role of a KCNH2 mutation in long-QT syndrome. EMBO J. 2013, 32, 3161–3175. [Google Scholar] [CrossRef]
- Itzhaki, I.; Maizels, L.; Huber, I.; Zwi-Dantsis, L.; Caspi, O.; Winterstern, A.; Feldman, O.; Gepstein, A.; Arbel, G.; Hammerman, H.; et al. Modelling the long QT syndrome with induced pluripotent stem cells. Nature 2011, 471, 225–229. [Google Scholar] [CrossRef] [PubMed]
- Matsa, E.; Rajamohan, D.; Dick, E.; Young, L.; Mellor, I.; Staniforth, A.; Denning, C. Drug evaluation in cardiomyocytes derived from human induced pluripotent stem cells carrying a long QT syndrome type 2 mutation. Eur. Heart J. 2011, 32, 952–962. [Google Scholar] [CrossRef] [Green Version]
- Van Mil, A.; Balk, G.M.; Neef, K.; Buikema, J.W.; Asselbergs, F.W.; Wu, S.M.; Doevendans, P.A.; Sluijter, J.P.G. Modelling inherited cardiac disease using human induced pluripotent stem cell-derived cardiomyocytes: Progress, pitfalls, and potential. Cardiovasc. Res. 2018, 114, 1828–1842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Smith, J.L.; Anderson, C.L.; Burgess, D.E.; Elayi, C.S.; January, C.T.; Delisle, B.P. Molecular pathogenesis of long QT syndrome type 2. J. Arrhythmia 2016, 32, 373–380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Spencer, C.I.; Baba, S.; Nakamura, K.; Hua, E.A.; Sears, M.A.F.; Fu, C.C.; Zhang, J.; Balijepalli, S.; Tomoda, K.; Hayashi, Y.; et al. Calcium transients closely reflect prolonged action potentials in iPSC models of inherited cardiac arrhythmia. Stem Cell Rep. 2014, 3, 269–281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Refsgaard, L.; Holst, A.G.; Sadjadieh, G.; Haunsø, S.; Nielsen, J.B.; Olesen, M.S. High prevalence of genetic variants previously associated with LQT syndrome in new exome data. Eur. J. Hum. Genet. 2012, 20, 905–908. [Google Scholar] [CrossRef]
- Gellens, M.E.; George, A.L.; Chen, L.; Chahine, M.; Hornt, R.; Barchi, R.L.; Kallent, R.G. Primary structure and functional expression of the human cardiac tetrodotoxin-insensitive voltage-dependent sodium channel. Proc. Natl. Acad. Sci. USA 1992, 89, 554–558. [Google Scholar] [CrossRef] [Green Version]
- Napolitano, C.; Priori, S.G.; Schwartz, P.J. Torsade de Pointes Mechanisms and Management. Drugs 1994, 47, 51–65. [Google Scholar] [CrossRef]
- Fatima, A.; Kaifeng, S.; Dittmann, S.; Xu, G.; Gupta, M.K.; Linke, M.; Nguemo, F.; Milting, H.; Farr, M.; Hescheler, J.; et al. The Disease-Specific Phenotype in Cardiomyocytes Derived from Induced Pluripotent Stem Cells of Two Long QT Syndrome Type 3 Patients. PLoS ONE 2013, 8, e83005. [Google Scholar] [CrossRef] [Green Version]
- Ma, D.; Wei, H.; Zhao, Y.; Lu, J.; Li, G.; Binte, N.; Sahib, E.; Hong, T.; Yean, K.; Shim, W.; et al. Modeling type 3 long QT syndrome with cardiomyocytes derived from patient-specific induced pluripotent stem cells. Int. J. Cardiol. 2013, 168, 5277–5286. [Google Scholar] [CrossRef]
- Terrenoire, C.; Wang, K.; Tung, K.W.C.; Chung, W.K.; Pass, R.H.; Lu, J.T.; Jean, J.; Omari, A.; Sampson, K.J.; Kotton, D.N.; et al. Induced pluripotent stem cells used to reveal drug actions in a long QT syndrome family with complex genetics. J. Gen. Physiol. 2013, 141, 61–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Malan, D.; Zhang, M.; Stallmeyer, B.; Muller, J.; Fleischmann, B.K.; Schulze-Bahr, E.; Sasse, P.; Greber, B. Human iPS cell model of type 3 long QT syndrome recapitulates drug-based phenotype correction. Basic Res. Cardiol. 2016, 111, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wilde, A.A.M.; Bezzina, C.R. Genetics of cardiac arrhythmias. Heart 2005, 91, 1352–1358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Remme, C.A.; Scicluna, B.P.; Verkerk, A.O.; Amin, A.S.; Van Brunschot, S.; Beekman, L.; Deneer, V.H.M.; Chevalier, C.; Oyama, F.; Miyazaki, H.; et al. Genetically Determined Differences in Sodium Current Characteristics Modulate Conduction Disease Severity in Mice With Cardiac Sodium Channelopathy. Circ. Res. 2009, 1283–1292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Veerapandiyan, A.; Statland, J.M.; Tawil, R. Andersen-Tawil Syndrome. In GeneReviews; Adam, M.P., Ardinger, H.H., Pagon, R.A., Wallace, S.E., Bean, L.J.H., Stephens, K., Amemiya, A., Eds.; University of Washington: Seattle, WA, USA, 2004; pp. 1993–2020. [Google Scholar]
- Dhamoon, A.S.; Jalife, J. The inward rectifier current (IK1) controls cardiac excitability and is involved in arrhythmogenesis. Hear. Rhythm 2005, 2, 316–324. [Google Scholar] [CrossRef]
- Plaster, N.M.; Tawil, R.; Tristani-Firouzi, M.; Canún, S.; Bendahhou, S.; Tsunoda, A.; Donaldson, M.R.; Iannaccone, S.T.; Brunt, E.; Barohn, R.; et al. Mutations in Kir2.1 cause the developmental and episodic electrical phenotypes of Andersen’s syndrome. Cell 2001, 105, 511–519. [Google Scholar] [CrossRef] [Green Version]
- Walsh, M.A.; Turner, C.; Timothy, K.W.; Seller, N.; Hares, D.L.; James, A.F.; Hancox, J.C.; Uzun, O.; Boyce, D.; Stuart, A.G.; et al. A multicentre study of patients with Timothy syndrome. Europace 2018, 20, 377–385. [Google Scholar] [CrossRef] [Green Version]
- Gez, L.S.; Hagalili, Y.; Shainberg, A.; Atlas, D. Voltage-driven Ca2+ binding at the L-type Ca2+ channel triggers cardiac excitation-contraction coupling prior to Ca2+ influx. Biochemistry 2012, 51, 9658–9666. [Google Scholar] [CrossRef]
- Crotti, L.; Ph, D.; Johnson, C.N.; Graf, E.; Sc, M.; De Ferrari, M.; Cuneo, B.F.; Ovadia, M.; Papagiannis, J.; Feldkamp, M.D.; et al. Calmodulin Mutations Associated with Recurrent Cardiac Arrest in Infants. Circulation 2014, 127, 1009–1017. [Google Scholar] [CrossRef] [Green Version]
- Rocchetti, M.; Sala, L.; Dreizehnter, L.; Crotti, L.; Sinnecker, D.; Mura, M.; Pane, L.S.; Altomare, C.; Torre, E.; Mostacciuolo, G.; et al. Elucidating arrhythmogenic mechanisms of long-QT syndrome CALM1-F142L mutation in patient-specific induced pluripotent stem cell-derived cardiomyocytes. Cardiovasc. Res. 2017, 113, 531–541. [Google Scholar] [CrossRef]
- Limpitikul, W.B.; Dick, I.E.; Tester, D.J.; Boczek, N.J.; Limphong, P.; Yang, W.; Choi, M.H.; Babich, J.; DiSilvestre, D.; Kanter, R.J.; et al. A Precision Medicine Approach to the Rescue of Function on Malignant Calmodulinopathic Long QT Syndrome. Circ. Res. 2017, 120, 39–48. [Google Scholar] [CrossRef]
- Grossmann, K.S.; Rosário, M.; Birchmeier, C.; Birchmeier, W. The tyrosine phosphatase Shp2 in development and cancer. Adv. Cancer Res. 2010, 106, 53–89. [Google Scholar]
- Swan, H.; Piippo, K.; Viitasalo, M.; Heikkilä, P.; Paavonen, T.; Kainulainen, K.; Kere, J.; Keto, P.; Kontula, K.; Toivonen, L. Arrhythmic disorder mapped to chromosome 1q42-q43 causes malignant polymorphic ventricular tachycardia in structurally normal hearts. J. Am. Coll. Cardiol. 1999, 34, 2035–2042. [Google Scholar] [CrossRef] [Green Version]
- Roston, T.M.; Van Petegem, F.; Sanatani, S. Catecholaminergic polymorphic ventricular tachycardia: A model for genotype-specific therapy. Curr. Opin. Cardiol. 2017, 32, 78–85. [Google Scholar] [CrossRef] [PubMed]
- Jung, C.B.; Moretti, A.; Mederos, M.; Iop, L.; Storch, U.; Bellin, M.; Dorn, T.; Ruppenthal, S.; Pfeiffer, S.; Goedel, A.; et al. Dantrolene rescues arrhythmogenic RYR2 defect in a patient-specific stem cell model of catecholaminergic polymorphic ventricular tachycardia. EMBO Mol. Med. 2012, 4, 180–191. [Google Scholar] [CrossRef] [PubMed]
- Kujala, K.; Paavola, J.; Lahti, A.; Larsson, K.; Pekkanen-mattila, M.; Viitasalo, M.; Lahtinen, A.M.; Toivonen, L.; Kontula, K.; Swan, H.; et al. Cell Model of Catecholaminergic Polymorphic Ventricular Tachycardia Reveals Early and Delayed Afterdepolarizations. PLoS ONE 2012, 7, e44660. [Google Scholar] [CrossRef] [PubMed]
- Sasaki, K.; Makiyama, T.; Yoshida, Y.; Wuriyanghai, Y.; Kamakura, T.; Nishiuchi, S.; Hayano, M.; Harita, T.; Yamamoto, Y.; Kohjitani, H.; et al. Patient-Specific Human Induced Pluripotent Stem Cell Model Assessed with Electrical Pacing Validates S107 as a Potential Therapeutic Agent for Catecholaminergic Polymorphic Ventricular Tachycardia. PLoS ONE 2016, 11, e0164795. [Google Scholar] [CrossRef] [PubMed]
- Preininger, M.K.; Jha, R.; Maxwell, J.T.; Wu, Q.; Singh, M.; Wang, B.; Dalal, A.; Mceachin, Z.T.; Rossoll, W.; Hales, C.M.; et al. A human pluripotent stem cell model of catecholaminergic polymorphic ventricular tachycardia recapitulates patient-specific drug responses. Dis. Model. Mech. 2016, 9, 927–939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thiene, G.; Corrado, D.; Basso, C. Arrhythmogenic right ventricular cardiomyopathy/dysplasia. Orphanet J. Rare Dis. 2007, 2, 45. [Google Scholar] [CrossRef]
- Kim, C.; Wong, J.; Wen, J.; Wang, S.; Wang, C.; Spiering, S.; Kan, N.G.; Forcales, S.; Puri, P.L.; Leone, T.C.; et al. Studying arrhythmogenic right ventricular dysplasia with patient-specific iPSCs. Nature 2013, 494, 105–110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dorn, T.; Kornherr, J.; Parrotta, E.I.; Zawada, D.; Ayetey, H.; Santamaria, G.; Iop, L.; Mastantuono, E.; Sinnecker, D.; Goedel, A.; et al. Interplay of cell-cell contacts and RhoA/MRTF-A signaling regulates cardiomyocyte identity. EMBO J. 2018, 37, e98133. [Google Scholar] [CrossRef]
- Muchtar, E.; Blauwet, L.A.; Gertz, M.A. Restrictive cardiomyopathy: Genetics, pathogenesis, clinical manifestations, diagnosis, and therapy. Circ. Res. 2017, 121, 819–837. [Google Scholar] [CrossRef]
- Rammos, A.; Meladinis, V.; Vovas, G.; Patsouras, D. Restrictive Cardiomyopathies: The Importance of Noninvasive Cardiac Imaging Modalities in Diagnosis and Treatment—A Systematic Review. Radiol. Res. Pract. 2017, 2017, 2874902. [Google Scholar] [CrossRef] [Green Version]
- Mogensen, J.; Kubo, T.; Duque, M.; Uribe, W.; Shaw, A.; Murphy, R.; Gimeno, J.R.; Elliott, P.; McKenna, W.J. Idiopathic restrictive cardiomyopathy is part of the clinical expression of cardiac troponin I mutations. J. Clin. Investig. 2003, 111, 209–216. [Google Scholar] [CrossRef] [Green Version]
- Kaski, J.P.; Syrris, P.; Burch, M.; Tomé Esteban, M.T.; Fenton, M.; Christiansen, M.; Andersen, P.S.; Sebire, N.; Ashworth, M.; Deanfield, J.E.; et al. Idiopathic restrictive cardiomyopathy in children is caused by mutations in cardiac sarcomere protein genes. Heart 2008, 94, 1478–1484. [Google Scholar] [CrossRef]
- Kostareva, A.; Kiselev, A.; Gudkova, A.; Frishman, G.; Ruepp, A.; Frishman, D.; Smolina, N.; Tarnovskaya, S.; Nilsson, D.; Zlotina, A.; et al. Genetic spectrum of idiopathic restrictive cardiomyopathy uncovered by next-generation sequencing. PLoS ONE 2016, 11, e0163362. [Google Scholar] [CrossRef] [Green Version]
- Pruszczyk, P.; Kostera-Pruszczyk, A.; Shatunov, A.; Goudeau, B.; Dramiñska, A.; Takeda, K.; Sambuughin, N.; Vicart, P. Restrictive cardiomyopathy with atrioventricular conduction block resulting from a desmin mutation. Int. J. Cardiol. 2007, 117, 244–253. [Google Scholar] [CrossRef]
- Wu, W.; Lu, C.X.; Wang, Y.N.; Liu, F.; Chen, W.; Liu, Y.T.; Han, Y.C.; Cao, J.; Zhang, S.Y.; Zhang, X. Novel phenotype-genotype correlations of restrictive cardiomyopathy with myosin-binding protein c (mybpc3) gene mutations tested by next-generation sequencing. J. Am. Heart Assoc. 2015, 4, e001879. [Google Scholar] [CrossRef] [Green Version]
- Greenway, S.C.; Wilson, G.J.; Wilson, J.; George, K.; Kantor, P.F. Sudden death in an infant with angina, restrictive cardiomyopathy, and coronary artery bridging an unusual phenotype for a β-myosin heavy chain (MYH7) sarcomeric protein mutation. Circ. Hear. Fail. 2012, 5, 92–93. [Google Scholar]
- Huby, A.C.; Mendsaikhan, U.; Takagi, K.; Martherus, R.; Wansapura, J.; Gong, N.; Osinska, H.; James, J.F.; Kramer, K.; Saito, K.; et al. Disturbance in Z-disk mechanosensitive proteins induced by a persistent mutant myopalladin causes familial restrictive cardiomyopathy. J. Am. Coll. Cardiol. 2014, 64, 2765–2776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brodehl, A.; Ferrier, R.A.; Hamilton, S.J.; Greenway, S.C.; Brundler, M.A.; Yu, W.; Gibson, W.T.; Mckinnon, M.L.; Mcgillivray, B.; Alvarez, N.; et al. Mutations in FLNC are Associated with Familial Restrictive Cardiomyopathy. Hum. Mutat. 2016, 37, 269–279. [Google Scholar] [CrossRef] [PubMed]
- Tucker, N.R.; McLellan, M.A.; Hu, D.; Ye, J.; Parsons, V.A.; Mills, R.W.; Clauss, S.; Dolmatova, E.; Shea, M.A.; Milan, D.J.; et al. Novel Mutation in FLNC (Filamin C) Causes Familial Restrictive Cardiomyopathy. Circ. Cardiovasc. Genet. 2017, 10, e001780. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brodehl, A.; Pour Hakimi, S.A.; Stanasiuk, C.; Ratnavadivel, S.; Hendig, D.; Gaertner, A.; Gerull, B.; Gummert, J.; Paluszkiewicz, L.; Milting, H. Restrictive Cardiomyopathy is Caused by a Novel Homozygous Desmin (DES) Mutation p.Y122H Leading to a Severe Filament Assembly Defect. Genes 2019, 10, 918. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Klauke, B.; Gaertner-rommel, A.; Schulz, U.; Kassner, A.; Laser, T.; Kececioglu, D.; Paluszkiewicz, L.; Blanz, U.; Sandica, E.; Van Den Bogaerdt, A.J.; et al. High proportion of genetic cases in patients with advanced cardiomyopathy including a novel homozygous Plakophilin 2-gene mutation. PLoS ONE 2017, 12, e0189489. [Google Scholar] [CrossRef] [Green Version]
- Gerull, B.; Gramlich, M.; Atherton, J.; McNabb, M.; Trombitás, K.; Sasse-Klaassen, S.; Seidman, J.G.; Seidman, C.; Granzier, H.; Labeit, S.; et al. Mutations of TTN, encoding the giant muscle filament titin, cause familial dilated cardiomyopathy. Nat. Genet. 2002, 30, 201–204. [Google Scholar] [CrossRef]
- Gramlich, M.; Pane, L.S.; Zhou, Q.; Chen, Z.; Murgia, M.; Schötterl, S.; Goedel, A.; Metzger, K.; Brade, T.; Parrotta, E.; et al. Antisense-mediated exon skipping: A therapeutic strategy for titin-based dilated cardiomyopathy. EMBO Mol. Med. 2015, 7, 562–576. [Google Scholar] [CrossRef]
- Lin, B.; Li, Y.; Han, L.; Kaplan, A.D.; Ao, Y.; Kalra, S.; Bett, G.C.L.; Rasmusson, R.L.; Denning, C.; Yang, L. Modeling and study of the mechanism of dilated cardiomyopathy using induced pluripotent stem cells derived from individuals with Duchenne muscular dystrophy. Dis Model. Mech. 2015, 8, 457–466. [Google Scholar] [CrossRef] [Green Version]
- Wu, H.; Lee, J.; Vincent, L.G.; Wang, Q.; Gu, M.; Lan, F.; Churko, J.M.; Sallam, K.I.; Matsa, E.; Sharma, A.; et al. Epigenetic Regulation of Phosphodiesterases 2A and 3A Underlies Compromised β-Adrenergic Signaling in an iPSC Model of Dilated Cardiomyopathy. Cell Stem Cell 2015, 17, 89–100. [Google Scholar] [CrossRef] [Green Version]
- Yang, K.-C.; Breitbart, A.; De Lange, W.J.; Hofsteen, P.; Futakuchi-Tsuchida, A.; Xu, J.; Schopf, C.; Razumova, M.V.; Jiao, A.; Boucek, R.; et al. Novel Adult-Onset Systolic Cardiomyopathy Due to MYH7 E848G Mutation in Patient-Derived Induced Pluripotent Stem Cells. JACC Basic Transl. Sci. 2018, 3, 728–740. [Google Scholar] [CrossRef]
- Lee, Y.; Lau, Y.; Cai, Z.; Lai, W.; Wong, L.; Tse, H.; Ng, K.; Siu, C. Modeling Treatment Response for Lamin A/C Related Dilated Cardiomyopathy in Human Induced Pluripotent Stem Cells. J. Am. Heart Assoc. 2017, 6, e005677. [Google Scholar] [CrossRef] [PubMed]
- Norton, N.; Li, D.; Rieder, M.J.; Siegfried, J.D.; Rampersaud, E.; Mangos, S.; Gonzalez-quintana, J.; Wang, L.; Mcgee, S.; Reiser, J.; et al. Genome-wide Studies of Copy Number Variation and Exome Sequencing Identify Rare Variants in BAG3 as a Cause of Dilated Cardiomyopathy. Am. J. Hum. Genet. 2011, 88, 273–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hishiya, A.; Kitazawa, T.; Takayama, S. BAG3 and Hsc70 Interact With Actin Capping Protein CapZ to Maintain Myofibrillar Integrity Under Mechanical Stress. Circ. Res. 2010, 107, 1220–1231. [Google Scholar] [CrossRef] [Green Version]
- Arndt, V.; Dick, N.; Tawo, R.; Dreiseidler, M.; Wenzel, D.; Hesse, M.; Saftig, P.; Saint, R.; Fu, D.O.; Fleischmann, B.K.; et al. Chaperone-Assisted Selective Autophagy Is Essential for Muscle Maintenance. Curr. Biol. 2010, 20, 143–148. [Google Scholar] [CrossRef]
- Dai, Y.; Amenov, A.; Ignatyeva, N.; Koschinski, A.; Xu, H.; Soong, P.L.; Tiburcy, M.; Linke, W.A.; Zaccolo, M.; Hasenfuss, G.; et al. Troponin destabilization impairs sarcomere-cytoskeleton interactions in iPSC-derived cardiomyocytes from dilated cardiomyopathy patients. Sci. Rep. 2020, 10, 209. [Google Scholar] [CrossRef] [Green Version]
- Towbin, J.A. Left Ventricular Noncompaction: A New Form of Heart Failure. Heart Fail. Clin. 2010, 6, 453–469. [Google Scholar] [CrossRef]
- Richard, P.; Charron, P.; Carrier, L.; Ledeuil, C.; Cheav, T.; Benaiche, A.; Isnard, R.; Dubourg, O.; Burban, M.; Millaire, A.; et al. Hypertrophic Cardiomyopathy: Distribution of Disease Genes, Spectrum of Mutations, and Implications for a Molecular Diagnosis Strategy. Circulation 2003, 107, 2227–2232. [Google Scholar] [CrossRef] [PubMed]
- Cohn, R.; Thakar, K.; Lowe, A.; Ladha, F.A.; Pettinato, A.M.; Romano, R.; Meredith, E.; Chen, Y.S.; Atamanuk, K.; Huey, B.D.; et al. A Contraction Stress Model of Hypertrophic Cardiomyopathy due to Sarcomere Mutations. Stem Cell Rep. 2019, 12, 71–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Camacho, P.; Fan, H.; Liu, Z.; He, J. Small mammalian animal models of heart disease. Am. J. Cardiovasc. Dis. 2016, 6, 70–80. [Google Scholar]
- Chaicharoenaudomrung, N.; Kunhorm, P.; Noisa, P. Three-dimensional cell culture systems as an in vitro platform for cancer and stem cell modeling. World J. Stem Cells 2019, 11, 1065–1083. [Google Scholar] [CrossRef] [PubMed]
- Figtree, G.A.; Bubb, K.J.; Tang, O.; Kizana, E.; Gentile, C. Vascularized Cardiac Spheroids as Novel 3D in vitro Models to Study Cardiac Fibrosis. Cells Tissues Organs 2017, 204, 191–198. [Google Scholar] [CrossRef] [PubMed]
- Archer, C.R.; Sargeant, R.; Basak, J.; Pilling, J.; Barnes, J.R.; Pointon, A. Characterization and Validation of a Human 3D Cardiac Microtissue for the Assessment of Changes in Cardiac Pathology. Sci. Rep. 2018, 8, 10160. [Google Scholar] [CrossRef] [PubMed]
- Al-Haque, S.; Miklas, J.W.; Feric, N.; Chiu, L.L.Y.; Li, W.; Chen, K.; Simmons, C.A.; Radisic, M. Hydrogel Substrate Stiffness and Topography Interact to Induce Contact Guidance in Cardiac Fibroblasts. Macromol. Biosci. 2012, 12, 1342–1353. [Google Scholar] [CrossRef]
- Zuppinger, C. 3D Cardiac Cell Culture: A Critical Review of Current Technologies and Applications. Front. Cardiovasc. Med. 2019, 6, 87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mosqueira, D.; Mannhardt, I.; Bhagwan, J.R.; Lis-slimak, K.; Katili, P.; Scott, E.; Hassan, M.; Prondzynski, M.; Harmer, S.C.; Tinker, A.; et al. CRISPR/Cas9 editing in human pluripotent stem cell-cardiomyocytes highlights arrhythmias, hypocontractility, and energy depletion as potential therapeutic targets for hypertrophic cardiomyopathy. Eur. Heart J. 2018, 44, 3879–3892. [Google Scholar] [CrossRef]
- Zhao, Y.; Rafatian, N.; Feric, N.T.; Cox, B.J.; Aschar-Sobbi, R.; Wang, E.Y.; Aggarwal, P.; Zhang, B.; Conant, G.; Ronaldson-Bouchard, K.; et al. A Platform for Generation of Chamber-Specific Cardiac Tissues and Disease Modeling. Cell 2019, 176, 913–927.e18. [Google Scholar] [CrossRef] [Green Version]
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Parrotta, E.I.; Lucchino, V.; Scaramuzzino, L.; Scalise, S.; Cuda, G. Modeling Cardiac Disease Mechanisms Using Induced Pluripotent Stem Cell-Derived Cardiomyocytes: Progress, Promises and Challenges. Int. J. Mol. Sci. 2020, 21, 4354. https://doi.org/10.3390/ijms21124354
Parrotta EI, Lucchino V, Scaramuzzino L, Scalise S, Cuda G. Modeling Cardiac Disease Mechanisms Using Induced Pluripotent Stem Cell-Derived Cardiomyocytes: Progress, Promises and Challenges. International Journal of Molecular Sciences. 2020; 21(12):4354. https://doi.org/10.3390/ijms21124354
Chicago/Turabian StyleParrotta, Elvira Immacolata, Valeria Lucchino, Luana Scaramuzzino, Stefania Scalise, and Giovanni Cuda. 2020. "Modeling Cardiac Disease Mechanisms Using Induced Pluripotent Stem Cell-Derived Cardiomyocytes: Progress, Promises and Challenges" International Journal of Molecular Sciences 21, no. 12: 4354. https://doi.org/10.3390/ijms21124354
APA StyleParrotta, E. I., Lucchino, V., Scaramuzzino, L., Scalise, S., & Cuda, G. (2020). Modeling Cardiac Disease Mechanisms Using Induced Pluripotent Stem Cell-Derived Cardiomyocytes: Progress, Promises and Challenges. International Journal of Molecular Sciences, 21(12), 4354. https://doi.org/10.3390/ijms21124354