307
Views
0
CrossRef citations to date
0
Altmetric
Review

Induced pluripotent stem cell derived cardiac models: effects of Thymosin β4

, &
Pages 111-120 | Received 30 Dec 2017, Accepted 02 May 2018, Published online: 31 Jul 2018

References

  • Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663–676.
  • Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131(5):861–872.
  • Yu J, Vodyanik MA, Smuga-Otto K, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007;318(5858):1917–1920.
  • Chambers SM, Fasano CA, Papapetrou EP, et al. Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nat Biotechnol. 2009;27(3):275–280.
  • Choi SM, Kim Y, Shim JS, et al. Efficient drug screening and gene correction for treating liver disease using patient-specific stem cells. Hepatology. 2013;57(6):2458–2468.
  • Mizuno Y, Chang H, Umeda K, et al. Generation of skeletal muscle stem/progenitor cells from murine induced pluripotent stem cells. Faseb J. 2010;24(7):2245–2253.
  • Moretti A, Bellin M, Welling A, et al. Patient-specific induced pluripotent stem-cell models for long-QT syndrome. N Engl J Med. 2010;363(15):1397–409.
  • Gramlich M, Pane LS, Zhou Q, et al. Antisense-mediated exon skipping: a therapeutic strategy for titin-based dilated cardiomyopathy. EMBO Mol Med. 2015;7(5):562–576.
  • Lian X, Zhang J, Azarin SM, et al. Directed cardiomyocyte differentiation from human pluripotent stem cells by modulating Wnt/beta-catenin signaling under fully defined conditions. Nat Protoc. 2013;8(1):162–175.
  • Zhang J, Wilson GF, Soerens AG, et al. Functional cardiomyocytes derived from human induced pluripotent stem cells. Circ Res. 2009;104(4):e30–41.
  • Burridge PW, Li YF, Matsa E, et al. Human induced pluripotent stem cell-derived cardiomyocytes recapitulate the predilection of breast cancer patients to doxorubicin-induced cardiotoxicity. Nat Med. 2016;22(5):547–556.
  • Kyrychenko, V, Kyrychenko S, Tiburcy M, et al. Functional correction of dystrophin actin binding domain mutations by genome editing. JCI Insight. 2017;2(18):e95918.
  • Chong JJH, Yang X, Don CW, et al. Human embryonic-stem-cell-derived cardiomyocytes regenerate non-human primate hearts. Nature. 2014;510(7504):273–277.
  • Weinberger F, Breen M, Choyke P, et al. Cardiac repair in guinea pigs with human engineered heart tissue from induced pluripotent stem cells. Sci Transl Med. 2016;8(363):363ra148.
  • Bowles J, Schepers G, Koopman P. Phylogeny of the SOX family of developmental transcription factors based on sequence and structural indicators. Dev Biol. 2000;227(2):239–255.
  • Schepers GE, Teasdale RD, Koopman P. Twenty pairs of sox: extent, homology, and nomenclature of the mouse and human sox transcription factor gene families. Dev Cell. 2002;3(2):167–170.
  • Kidder BL, Palmer S, Knott JG. SWI/SNF-Brg1 regulates self-renewal and occupies core pluripotency-related genes in embryonic stem cells. Stem Cells. 2009;27(2):317–328.
  • McMahon SB, Van Buskirk HA, Dugan KA, et al. The novel ATM-related protein TRRAP is an essential cofactor for the c-Myc and E2F oncoproteins. Cell. 1998;94(3):363–374.
  • Nakagawa M, Takizawa N, Narita M, et al. Promotion of direct reprogramming by transformation-deficient Myc. Proc Natl Acad Sci U S A. 2010;107(32):14152–14157.
  • Niwa H, Ogawa K, Shimosato D, et al. A parallel circuit of LIF signalling pathways maintains pluripotency of mouse ES cells. Nature. 2009;460(7251):118–122.
  • Maekawa M, Yamaguchi K, Nakamura T, et al. Direct reprogramming of somatic cells is promoted by maternal transcription factor Glis1. Nature. 2011;474(7350):225–229.
  • Yoshida Y, Takahashi K, Okita K, et al. Hypoxia enhances the generation of induced pluripotent stem cells. Cell Stem Cell. 2009;5(3):237–241.
  • Huangfu D, Maehr R, Guo W, et al. Induction of pluripotent stem cells by defined factors is greatly improved by small-molecule compounds. Nat Biotechnol. 2008;26(7):795–797.
  • Shi Y, Desponts C, Do JT, et al. Induction of pluripotent stem cells from mouse embryonic fibroblasts by Oct4 and Klf4 with small-molecule compounds. Cell Stem Cell. 2008;3(5):568–574.
  • Lin T, Ambasudhan R, Yuan X, et al. A chemical platform for improved induction of human iPSCs. Nat Methods. 2009;6(11):805–808.
  • Judson RL, Babiarz JE, Venere M, et al. Embryonic stem cell-specific microRNAs promote induced pluripotency. Nat Biotechnol. 2009;27(5):459–461.
  • Cheng L, Hansen NF, Zhao L, et al. Low incidence of DNA sequence variation in human induced pluripotent stem cells generated by nonintegrating plasmid expression. Cell Stem Cell. 2012;10(3):337–344.
  • Hussein SM, Batada NN, Vuoristo S, et al. Copy number variation and selection during reprogramming to pluripotency. Nature. 2011;471(7336):58–62.
  • Ji J, Ng SH, Sharma V, et al. Elevated coding mutation rate during the reprogramming of human somatic cells into induced pluripotent stem cells. Stem Cells. 2012;30(3):435–440.
  • Okita K, Nakagawa M, Hyenjong H, et al. Generation of mouse induced pluripotent stem cells without viral vectors. Science. 2008;322(5903):949–953.
  • Seki T, Yuasa S, Oda M, et al. Generation of induced pluripotent stem cells from human terminally differentiated circulating T cells. Cell Stem Cell. 2010;7(1):11–14.
  • Woltjen K, Michael IP, Mohseni P, et al. piggyBac transposition reprograms fibroblasts to induced pluripotent stem cells. Nature. 2009;458(7239):766–770.
  • Malik N, Rao MS. A review of the methods for human iPSC derivation. Methods Mol Biol. 2013;997:23–33.
  • Steichen C, Luce E, Maluenda J, et al. Messenger RNA- versus retrovirus-based induced pluripotent stem cell reprogramming strategies: analysis of genomic integrity. Stem Cells Transl Med. 2014;3(6):686–691.
  • Bouma MJ, Van Iterson M, Janssen B, et al. Differentiation-defective human induced pluripotent stem cells reveal strengths and limitations of the teratoma assay and in vitro pluripotency assays. Stem Rep. 2017;8(5):1340–1353.
  • Müller F-J, Schuldt BM, Williams R, et al. A bioinformatic assay for pluripotency in human cells. Nat Methods. 2011;8(4):315–317.
  • Tsankov AM, Akopian V, Pop R, et al. A qPCR scoreCard quantifies the differentiation potential of human pluripotent stem cells. Nat Biotechnol. 2015;33(11):1182–1192.
  • Luu, P.-L, Gerovska D, Schöler HR, et al. Rules governing the mechanism of epigenetic reprogramming memory. Vol. 10. 2018;10(2):149–174.
  • Chen Y, Li X, Eswarakumar VP, et al. Fibroblast growth factor (FGF) signaling through PI 3-kinase and Akt/PKB is required for embryoid body differentiation. Oncogene. 2000;19(33):3750–3756.
  • Nostro MC, Cheng X, Keller GM, et al. Wnt, activin, and BMP signaling regulate distinct stages in the developmental pathway from embryonic stem cells to blood. Cell Stem Cell. 2008;2(1):60–71.
  • Bellin M, Casini S, Davis RP, et al. Isogenic human pluripotent stem cell pairs reveal the role of a KCNH2 mutation in long-QT syndrome. Embo J. 2013;32(24):3161–3175.
  • Jung CB, Moretti A, Mederos Y Schnitzler M, et al. Dantrolene rescues arrhythmogenic RYR2 defect in a patient-specific stem cell model of catecholaminergic polymorphic ventricular tachycardia. EMBO Mol Med. 2012;4(3):180–191.
  • Costello I, Pimeisl I-M, Dräger S, et al. The T-box transcription factor Eomesodermin acts upstream of Mesp1 to specify cardiac mesoderm during mouse gastrulation. Nat Cell Biol. 2011;13(9):1084–1091.
  • Faial T, Bernardo AS, Mendjan S, et al. Brachyury and SMAD signalling collaboratively orchestrate distinct mesoderm and endoderm gene regulatory networks in differentiating human embryonic stem cells. Development. 2015;142(12):2121–2135.
  • Bondue A, Blanpain C. Mesp1: a key regulator of cardiovascular lineage commitment. Circ Res. 2010;107(12):1414–1427.
  • David R, Brenner C, Stieber J, et al. MesP1 drives vertebrate cardiovascular differentiation through Dkk-1-mediated blockade of Wnt-signalling. Nat Cell Biol. 2008;10(3):338–345.
  • Tanaka M, Chen Z, Bartunkova S, et al. The cardiac homeobox gene Csx/Nkx2.5 lies genetically upstream of multiple genes essential for heart development. Development. 1999;126(6):1269–1280.
  • Drouin E, Charpentier F, Gauthier C, et al. Electrophysiologic characteristics of cells spanning the left ventricular wall of human heart: evidence for presence of M cells. J Am Coll Cardiol. 1995;26(1):185–192.
  • Saggin L, Gorza L, Ausoni S, et al. Troponin I switching in the developing heart. J Biol Chem. 1989;264(27):16299–16302.
  • Zwi L, Caspi O, Arbel G, et al. Cardiomyocyte differentiation of human induced pluripotent stem cells. Circulation. 2009;120(15):1513–1523.
  • Chen Z, Xian W, Bellin M, et al. Subtype-specific promoter-driven action potential imaging for precise disease modelling and drug testing in hiPSC-derived cardiomyocytes. Eur Heart J. 2017;38(4):292–301.
  • Yang L, Soonpaa MH, Adler ED, et al. Human cardiovascular progenitor cells develop from a KDR+ embryonic-stem-cell-derived population. Nature. 2008;453(7194):524–528.
  • Parikh SS, Blackwell DJ, Gomez-Hurtado N, et al. Thyroid and glucocorticoid hormones promote functional T-tubule development in human-induced pluripotent stem cell-derived cardiomyocytes. Circ Res. 2017;121(12):1323–1330.
  • Yang X, Rodriguez M, Pabon L, et al. Tri-iodo-l-thyronine promotes the maturation of human cardiomyocytes-derived from induced pluripotent stem cells. J Mol Cell Cardiol. 2014;72:296–304.
  • Kroll K, Chabria M, Wang K, et al. Electro-mechanical conditioning of human iPSC-derived cardiomyocytes for translational research. Prog Biophys Mol Biol. 2017;130(Pt B):212–222.
  • Mummery C, Ward-Van Oostwaard D, Doevendans P, et al. Differentiation of human embryonic stem cells to cardiomyocytes: role of coculture with visceral endoderm-like cells. Circulation. 2003;107(21):2733–2740.
  • Huethorst E, Hortigon M, Zamora-Rodriguez V, et al. Enhanced human-induced pluripotent stem cell derived cardiomyocyte maturation using a dual microgradient substrate. ACS Biomater Sci Eng. 2016;2(12):2231–2239.
  • Moss AJ, Shimizu W, Wilde AAM, et al. Clinical aspects of type-1 long-QT syndrome by location, coding type, and biophysical function of mutations involving the KCNQ1 gene. Circulation. 2007;115(19):2481–2489.
  • Herman DS, Lam L, Taylor MRG, et al. Truncations of titin causing dilated cardiomyopathy. N Engl J Med. 2012;366(7):619–628.
  • Sharma, A, Burridge PW, McKeithan WL, et al. High-throughput screening of tyrosine kinase inhibitor cardiotoxicity with human induced pluripotent stem cells. Sci Transl Med. 2017;9(377).
  • Mandai M, Watanabe A, Kurimoto Y, et al. Autologous induced stem-cell-derived retinal cells for macular degeneration. N Engl J Med. 2017;376(11):1038–1046.
  • Huang Y-C, Khait L, Birla RK. Contractile three-dimensional bioengineered heart muscle for myocardial regeneration. J Biomed Mater Res A. 2007;80(3):719–731.
  • Radisic M, Park H, Shing H, et al. Functional assembly of engineered myocardium by electrical stimulation of cardiac myocytes cultured on scaffolds. Proc Natl Acad Sci U S A. 2004;101(52):18129–18134.
  • Zimmermann W-H, Schneiderbanger K, Schubert P, et al. Tissue engineering of a differentiated cardiac muscle construct. Circ Res. 2002;90(2):223–230.
  • Herron T, McDonald KS. Small amounts of alpha-myosin heavy chain isoform expression significantly increase power output of rat cardiac myocyte fragments. Circ Res. 2002;90(11):1150–1152.
  • Zimmermann W-H, Melnychenko I, Wasmeier G, et al. Engineered heart tissue grafts improve systolic and diastolic function in infarcted rat hearts. Nat Med. 2006;12(4):452–458.
  • Zimmermann W-H, Didié M, Wasmeier GH, et al. Cardiac grafting of engineered heart tissue in syngenic rats. Circulation. 2002;106(12 Suppl 1):I151–I157.
  • Mannhardt I, Breckwoldt K, Letuffe-Brenière D, et al. Human engineered heart tissue: analysis of contractile force. Stem Rep. 2016;7(1):29–42.
  • Iseoka, H, Miyagawa S, Fukushima S, et al. Pivotal role of non-cardiomyocytes in electromechanical and therapeutic potential of induced pluripotent stem cell-derived engineered cardiac tissue. Tissue Eng Part A. 2018;24(3-4):287–300.
  • Weinberger F, Breckwoldt K, Pecha S, et al. Cardiac repair in guinea pigs with human engineered heart tissue from induced pluripotent stem cells. Sci Transl Med. 2016;8(363):363ra148–363ra148.
  • Xue B, Leyrat C, Grimes JM, et al. Structural basis of thymosin-β4/profilin exchange leading to actin filament polymerization. Proc Natl Acad Sci U S A. 2014;111(43):E4596–E4605.
  • Wang D-Z, Li S, Hockemeyer D, et al. Potentiation of serum response factor activity by a family of myocardin-related transcription factors. Proc Natl Acad Sci U S A. 2002;99(23):14855–14860.
  • Bock-Marquette I, Saxena A, White MD, et al. Thymosin beta4 activates integrin-linked kinase and promotes cardiac cell migration, survival and cardiac repair. Nature. 2004;432(7016):466–472.
  • Gomez-Marquez J, Franco Del Amo F, Carpintero P, et al. High levels of mouse thymosin beta4 mRNA in differentiating P19 embryonic cells and during development of cardiovascular tissues. Biochim Biophys Acta. 1996;1306(2–3):187–193.
  • Rossdeutsch A, Smart N, Dubé KN, et al. Essential role for thymosin beta4 in regulating vascular smooth muscle cell development and vessel wall stability. Circ Res. 2012;111(4):e89–102.
  • Smart N, Risebro CA, Melville AAD, et al. Thymosin beta4 induces adult epicardial progenitor mobilization and neovascularization. Nature. 2007;445(7124):177–182.
  • Yan B, Xu H, Yan Q, et al. Regulation of PTEN/Akt pathway enhances cardiomyogenesis and attenuates adverse left ventricular remodeling following thymosin beta4 Overexpressing embryonic stem cell transplantation in the infarcted heart. PLoS One. 2013;8(9):e75580.
  • Smart N, Riegler J, Turtle CW, et al. Aberrant developmental titin splicing and dysregulated sarcomere length in Thymosin beta4 knockout mice. J Mol Cell Cardiol. 2017;102:94–107.
  • Hinkel R, Ball HL, DiMaio JM, et al. C-terminal variable AGES domain of Thymosin beta4: the molecule’s primary contribution in support of post-ischemic cardiac function and repair. J Mol Cell Cardiol. 2015;87:113–125.
  • Hertzog M, Van Heijenoort C, Didry D, et al. The beta-thymosin/WH2 domain; structural basis for the switch from inhibition to promotion of actin assembly. Cell. 2004;117(5):611–623.
  • Hinkel R, Hamm JM, Page AF, et al. MRTF-A controls vessel growth and maturation by increasing the expression of CCN1 and CCN2. Nat Commun. 2014;5:3970.
  • Miralles F, Posern G, Zaromytidou A-I, et al. Actin dynamics control SRF activity by regulation of its coactivator MAL. Cell. 2003;113(3):329–342.
  • Nelson TJ, Balza R, Xiao Q, et al. SRF-dependent gene expression in isolated cardiomyocytes: regulation of genes involved in cardiac hypertrophy. J Mol Cell Cardiol. 2005;39(3):479–489.
  • Niu Z, Yu W, Zhang SX, et al. Conditional mutagenesis of the murine serum response factor gene blocks cardiogenesis and the transcription of downstream gene targets. J Biol Chem. 2005;280(37):32531–32538.
  • Nam YJ, Song K, Luo X, et al. Reprogramming of human fibroblasts toward a cardiac fate. Proc Natl Acad Sci U S A. 2013;110(14):5588–5593.
  • Naito AT, Tominaga A, Oyamada M, et al. Early stage-specific inhibitions of cardiomyocyte differentiation and expression of Csx/Nkx-2.5 and GATA-4 by phosphatidylinositol 3-kinase inhibitor LY294002. Exp Cell Res. 2003;291(1):56–69.
  • Karakikes I, Senyei GD, Hansen J, et al. Small molecule-mediated directed differentiation of human embryonic stem cells toward ventricular cardiomyocytes. Stem Cells Transl Med. 2014;3(1):18–31.
  • Li L, Guleria RS, Thakur S, et al. Thymosin beta4 prevents angiotensin II-induced cardiomyocyte growth by regulating Wnt/WISP signaling. J Cell Physiol. 2016;231(8):1737–1744.
  • Hinkel R, Howe A, Renner S, et al. Diabetes mellitus-induced microvascular destabilization in the myocardium. J Am Coll Cardiol. 2017;69(2):131–143.
  • Philp D, Huff T, Gho YS, et al. The actin binding site on thymosin beta4 promotes angiogenesis. Faseb J. 2003;17(14):2103–2105.
  • Ziegler T, Hinkel R, Stöhr A, et al. Thymosin beta4 improves differentiation and vascularization of EHTs. Stem Cells Int. 2017;2017:6848271.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.