Skip to main content

Patient-Derived Induced Pluripotent Stem Cells Provide a Regenerative Medicine Platform for Duchenne Muscular Dystrophy Heart Failure

  • Chapter
  • First Online:
Regenerative Medicine for Degenerative Muscle Diseases

Part of the book series: Stem Cell Biology and Regenerative Medicine ((STEMCELL))

  • 841 Accesses

Abstract

Mutations in the dystrophin gene cause Duchenne muscular dystrophy, a hereditary condition that leads to degeneration of both skeletal and cardiac muscle. All DMD patients inevitably develop heart disease (cardiomyopathy), and 30 % of them succumb to congestive heart failure. The mechanism of DMD heart failure is still elusive, partly due to the hard-to-access heart cells from patients. Because no effective treatment exists for DMD heart disease, patients with DMD represent an unmet medical need. To address this need, we modeled heart disease in DMD using a regenerative medicine technology called cellular reprogramming. Reprogramming involves converting an adult somatic cell into a pluripotent stem cell, called induced pluripotent stem cell (iPSC). IPSCs are further transformed or differentiated into heart cells for study. Reprogramming technology provides vast amounts of heart cells, offering an unprecedented opportunity to study genetic heart diseases like DMD heart failure. Establishing this regenerative medicine platform will not only facilitate exploration of disease etiology, but also provide unlimited personalized material to screen therapeutic compounds for individual patients.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 84.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 109.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Hermans MC, Pinto YM, Merkies IS, de Die-Smulders CE, Crijns HJ, Faber CG. Hereditary muscular dystrophies and the heart. Neuromuscul Disord. 2010;20:479–92.

    Article  CAS  PubMed  Google Scholar 

  2. Armstrong SC, Latham CA, Shivell CL, Ganote CE. Ischemic loss of sarcolemmal dystrophin and spectrin: correlation with myocardial injury. J Mol Cell Cardiol. 2001;33:1165–79.

    Article  CAS  PubMed  Google Scholar 

  3. Kido M, Otani H, Kyoi S, Sumida T, Fujiwara H, Okada T, Imamura H. Ischemic preconditioning-mediated restoration of membrane dystrophin during reperfusion correlates with protection against contraction-induced myocardial injury. Am J Physiol Heart Circ Physiol. 2004;287:H81–90.

    Article  CAS  PubMed  Google Scholar 

  4. Lee GH, Badorff C, Knowlton KU. Dissociation of sarcoglycans and the dystrophin carboxyl terminus from the sarcolemma in enteroviral cardiomyopathy. Circ Res. 2000;87:489–95.

    Article  CAS  PubMed  Google Scholar 

  5. Burelle Y, Khairallah M, Ascah A, Allen BG, Deschepper CF, Petrof BJ, Des Rosiers C. Alterations in mitochondrial function as a harbinger of cardiomyopathy: lessons from the dystrophic heart. J Mol Cell Cardiol. 2010;48:310–21.

    Article  CAS  PubMed  Google Scholar 

  6. Hoffman EP, Brown Jr RH, Kunkel LM. Dystrophin: the protein product of the Duchenne muscular dystrophy locus. Cell. 1987;51:919–28.

    Article  CAS  PubMed  Google Scholar 

  7. Muntoni F, Torelli S, Ferlini A. Dystrophin and mutations: one gene, several proteins, multiple phenotypes. Lancet Neurol. 2003;2:731–40.

    Article  CAS  PubMed  Google Scholar 

  8. Percival JM, Adamo CM, Beavo JA, Froehner SC. Evaluation of the therapeutic utility of phosphodiesterase 5A inhibition in the mdx mouse model of duchenne muscular dystrophy. Handb Exp Pharmacol. 2011;323–344.

    Google Scholar 

  9. Spurney C, Shimizu R, Morgenroth LP, Kolski H, Gordish-Dressman H, Clemens PR, Investigators C. Cooperative International Neuromuscular Research Group Duchenne Natural History Study demonstrates insufficient diagnosis and treatment of cardiomyopathy in Duchenne muscular dystrophy. Muscle Nerve. 2014;50:250–6.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Nigro G, Comi LI, Politano L, Bain RJ. The incidence and evolution of cardiomyopathy in Duchenne muscular dystrophy. Int J Cardiol. 1990;26:271–7.

    Article  CAS  PubMed  Google Scholar 

  11. Townsend D, Yasuda S, Metzger J. Cardiomyopathy of Duchenne muscular dystrophy: pathogenesis and prospect of membrane sealants as a new therapeutic approach. Expert Rev Cardiovasc Ther. 2007;5:99–109.

    Article  CAS  PubMed  Google Scholar 

  12. Ishikawa Y, Bach JR, Minami R. Cardioprotection for Duchenne's muscular dystrophy. Am Heart J. 1999;137:895–902.

    Article  CAS  PubMed  Google Scholar 

  13. Matsumura T, Tamura T, Kuru S, Kikuchi Y, Kawai M. Carvedilol can prevent cardiac events in Duchenne muscular dystrophy. Intern Med. 2010;49:1357–63.

    Article  CAS  PubMed  Google Scholar 

  14. Rhodes J, Margossian R, Darras BT, Colan SD, Jenkins KJ, Geva T, Powell AJ. Safety and efficacy of carvedilol therapy for patients with dilated cardiomyopathy secondary to muscular dystrophy. Pediatr Cardiol. 2008;29:343–51.

    Article  CAS  PubMed  Google Scholar 

  15. Schram G, Fournier A, Leduc H, Dahdah N, Therien J, Vanasse M, Khairy P. All-cause mortality and cardiovascular outcomes with prophylactic steroid therapy in Duchenne muscular dystrophy. J Am Coll Cardiol. 2013;61:948–54.

    Article  PubMed  Google Scholar 

  16. Fayssoil A, Orlikowski D, Nardi O, Annane D. Complete atrioventricular block in Duchenne muscular dystrophy. Europace. 2008;10:1351–2.

    Article  CAS  PubMed  Google Scholar 

  17. Fayssoil A, Orlikowski D, Nardi O, Annane D. Pacemaker implantation for sinus node dysfunction in a young patient with Duchenne muscular dystrophy. Congest Heart Fail. 2010;16:127–8.

    Article  PubMed  Google Scholar 

  18. Takano N, Honke K, Hasui M, Ohno I, Takemura H. A case of pacemaker implantation for complete atrioventricular block associated with Duchenne muscular dystrophy. No To Hattatsu. 1997;29:476–80.

    CAS  PubMed  Google Scholar 

  19. Amodeo A, Adorisio R. Left ventricular assist device in Duchenne cardiomyopathy: can we change the natural history of cardiac disease? Int J Cardiol. 2012;161, e43.

    Article  PubMed  Google Scholar 

  20. Davies JE, Winokur TS, Aaron MF, Benza RL, Foley BA, Holman WL. Cardiomyopathy in a carrier of Duchenne's muscular dystrophy. J Heart Lung Transplant. 2001;20:781–4.

    Article  CAS  PubMed  Google Scholar 

  21. Smith MC, Arabia FA, Tsau PH, Smith RG, Bose RK, Woolley DS, Rhenman BE, Sethi GK, Copeland JG. CardioWest total artificial heart in a moribund adolescent with left ventricular thrombi. Ann Thorac Surg. 2005;80:1490–2.

    Article  PubMed  Google Scholar 

  22. Webb ST, Patil V, Vuylsteke A. Anaesthesia for non-cardiac surgery in patient with Becker's muscular dystrophy supported with a left ventricular assist device. Eur J Anaesthesiol. 2007;24:640–2.

    Article  CAS  PubMed  Google Scholar 

  23. Andrikopoulos G, Kourouklis S, Trika C, Tzeis S, Rassias I, Papademetriou C, Katsivas A, Theodorakis G. Cardiac resynchronization therapy in Becker muscular dystrophy. Hellenic J Cardiol. 2013;54:227–9.

    PubMed  Google Scholar 

  24. Fayssoil A, Abasse S. Cardiac resynchronization therapy in Becker muscular dystrophy: for which patients? Hellenic J Cardiol. 2010;51:377–8.

    PubMed  Google Scholar 

  25. Stollberger C, Finsterer J. Left ventricular synchronization by biventricular pacing in Becker muscular dystrophy as assessed by tissue Doppler imaging. Heart Lung. 2005;34:317–20.

    Article  PubMed  Google Scholar 

  26. Leung DG, Herzka DA, Thompson WR, He B, Bibat G, Tennekoon G, Russell SD, Schuleri KH, Lardo AC, Kass DA, Thompson RE, Judge DP, Wagner KR. Sildenafil does not improve cardiomyopathy in Duchenne/Becker muscular dystrophy. Ann Neurol. 2014;76(4):541–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Childers MK, Okamura CS, Bogan DJ, Bogan JR, Petroski GF, McDonald K, Kornegay JN. Eccentric contraction injury in dystrophic canine muscle. Arch Phys Med Rehabil. 2002;83:1572–8.

    Article  PubMed  Google Scholar 

  28. Childers MK, Okamura CS, Bogan DJ, Bogan JR, Sullivan MJ, Kornegay JN. Myofiber injury and regeneration in a canine homologue of Duchenne muscular dystrophy. Am J Phys Med Rehabil. 2001;80:175–81.

    Article  CAS  PubMed  Google Scholar 

  29. Childers MK, Staley JT, Kornegay JN, McDonald KS. Skinned single fibers from normal and dystrophin-deficient dogs incur comparable stretch-induced force deficits. Muscle Nerve. 2005;31:768–71.

    Article  PubMed  Google Scholar 

  30. McNeil PL, Khakee R. Disruptions of muscle fiber plasma membranes. Role in exercise-induced damage. Am J Pathol. 1992;140:1097–109.

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Petrof BJ, Shrager JB, Stedman HH, Kelly AM, Sweeney HL. Dystrophin protects the sarcolemma from stresses developed during muscle contraction. Proc Natl Acad Sci U S A. 1993;90:3710–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Mokri B, Engel AG. Duchenne dystrophy: electron microscopic findings pointing to a basic or early abnormality in the plasma membrane of the muscle fiber. Neurology. 1975;25:1111–20.

    Article  CAS  PubMed  Google Scholar 

  33. Townsend D, Turner I, Yasuda S, Martindale J, Davis J, Shillingford M, Kornegay JN, Metzger JM. Chronic administration of membrane sealant prevents severe cardiac injury and ventricular dilatation in dystrophic dogs. J Clin Invest. 2010;120:1140–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Yasuda S, Townsend D, Michele DE, Favre EG, Day SM, Metzger JM. Dystrophic heart failure blocked by membrane sealant poloxamer. Nature. 2005;436:1025–9.

    Article  CAS  PubMed  Google Scholar 

  35. Allen DG, Whitehead NP. Duchenne muscular dystrophy--what causes the increased membrane permeability in skeletal muscle? Int J Biochem Cell Biol. 2011;43:290–4.

    Article  CAS  PubMed  Google Scholar 

  36. Prosser BL, Ward CW, Lederer WJ. X-ROS signaling: rapid mechano-chemo transduction in heart. Science. 2011;333:1440–5.

    Article  CAS  PubMed  Google Scholar 

  37. Ullrich ND, Fanchaouy M, Gusev K, Shirokova N, Niggli E. Hypersensitivity of excitation-contraction coupling in dystrophic cardiomyocytes. Am J Physiol Heart Circ Physiol. 2009;297:H1992–2003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Williams IA, Allen DG. Intracellular calcium handling in ventricular myocytes from mdx mice. Am J Physiol Heart Circ Physiol. 2007;292:H846–55.

    Article  CAS  PubMed  Google Scholar 

  39. Kyrychenko S, Polakova E, Kang C, Pocsai K, Ullrich ND, Niggli E, Shirokova N. Hierarchical accumulation of RyR post-translational modifications drives disease progression in dystrophic cardiomyopathy. Cardiovasc Res. 2013;97:666–75.

    Article  CAS  PubMed  Google Scholar 

  40. Jung C, Martins AS, Niggli E, Shirokova N. Dystrophic cardiomyopathy: amplification of cellular damage by Ca2+ signalling and reactive oxygen species-generating pathways. Cardiovasc Res. 2008;77:766–73.

    Article  CAS  PubMed  Google Scholar 

  41. Cheng YJ, Lang D, Caruthers SD, Efimov IR, Chen J, Wickline SA. Focal but reversible diastolic sheet dysfunction reflects regional calcium mishandling in dystrophic mdx mouse hearts. Am J Physiol Heart Circ Physiol. 2012;303:H559–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Koenig X, Dysek S, Kimbacher S, Mike AK, Cervenka R, Lukacs P, Nagl K, Dang XB, Todt H, Bittner RE, Hilber K. Voltage-gated ion channel dysfunction precedes cardiomyopathy development in the dystrophic heart. PLoS ONE. 2011;6, e20300.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Shin JH, Bostick B, Yue Y, Hajjar R, Duan D. SERCA2a gene transfer improves electrocardiographic performance in aged mdx mice. J Transl Med. 2011;9:132.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Whitehead NP, Yeung EW, Allen DG. Muscle damage in mdx (dystrophic) mice: role of calcium and reactive oxygen species. Clin Exp Pharmacol Physiol. 2006;33:657–62.

    Article  CAS  PubMed  Google Scholar 

  45. Franco Jr A, Lansman JB. Calcium entry through stretch-inactivated ion channels in mdx myotubes. Nature. 1990;344:670–3.

    Article  CAS  PubMed  Google Scholar 

  46. Squire S, Raymackers JM, Vandebrouck C, Potter A, Tinsley J, Fisher R, Gillis JM, Davies KE. Prevention of pathology in mdx mice by expression of utrophin: analysis using an inducible transgenic expression system. Hum Mol Genet. 2002;11:3333–44.

    Article  CAS  PubMed  Google Scholar 

  47. Fanchaouy M, Polakova E, Jung C, Ogrodnik J, Shirokova N, Niggli E. Pathways of abnormal stress-induced Ca2+ influx into dystrophic mdx cardiomyocytes. Cell Calcium. 2009;46:114–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Teichmann MD, Wegner FV, Fink RH, Chamberlain JS, Launikonis BS, Martinac B, Friedrich O. Inhibitory control over Ca(2+) sparks via mechanosensitive channels is disrupted in dystrophin deficient muscle but restored by mini-dystrophin expression. PLoS ONE. 2008;3, e3644.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Lorin C, Vögeli I, Niggli E. Dystrophic cardiomyopathy - role of TRPV2 channels in stretch-induced cell damage. Cardiovasc Res. 2015;106(1):153–62.

    Article  CAS  PubMed  Google Scholar 

  50. Sadeghi A, Doyle AD, Johnson BD. Regulation of the cardiac L-type Ca2+ channel by the actin-binding proteins alpha-actinin and dystrophin. Am J Physiol Cell Physiol. 2002;282:C1502–11.

    Article  CAS  PubMed  Google Scholar 

  51. Friedrich O, von Wegner F, Chamberlain JS, Fink RH, Rohrbach P. L-type Ca2+ channel function is linked to dystrophin expression in mammalian muscle. PLoS ONE. 2008;3, e1762.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Koenig X, Rubi L, Obermair GJ, Cervenka R, Dang XB, Lukacs P, Kummer S, Bittner RE, Kubista H, Todt H, Hilber K. Enhanced currents through L-type calcium channels in cardiomyocytes disturb the electrophysiology of the dystrophic heart. Am J Physiol Heart Circ Physiol. 2014;306:H564–73.

    Article  CAS  PubMed  Google Scholar 

  53. Woolf PJ, Lu S, Cornford-Nairn R, Watson M, Xiao XH, Holroyd SM, Brown L, Hoey AJ. Alterations in dihydropyridine receptors in dystrophin-deficient cardiac muscle. Am J Physiol Heart Circ Physiol. 2006;290:H2439–45.

    Article  CAS  PubMed  Google Scholar 

  54. Cohn RD, Durbeej M, Moore SA, Coral-Vazquez R, Prouty S, Campbell KP. Prevention of cardiomyopathy in mouse models lacking the smooth muscle sarcoglycan-sarcospan complex. J Clin Invest. 2001;107:R1–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Phillips MF, Quinlivan R. Calcium antagonists for Duchenne muscular dystrophy. Cochrane Database Syst Rev. 2008, CD004571.

    Google Scholar 

  56. Fauconnier J, Thireau J, Reiken S, Cassan C, Richard S, Matecki S, Marks AR, Lacampagne A. Leaky RyR2 trigger ventricular arrhythmias in Duchenne muscular dystrophy. Proc Natl Acad Sci U S A. 2010;107:1559–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Menazza S, Blaauw B, Tiepolo T, Toniolo L, Braghetta P, Spolaore B, Reggiani C, Di Lisa F, Bonaldo P, Canton M. Oxidative stress by monoamine oxidases is causally involved in myofiber damage in muscular dystrophy. Hum Mol Genet. 2010;19:4207–15.

    Article  CAS  PubMed  Google Scholar 

  58. Rando TA. Oxidative stress and the pathogenesis of muscular dystrophies. Am J Phys Med Rehabil. 2002;81:S175–86.

    Article  PubMed  Google Scholar 

  59. Spurney CF, Knoblach S, Pistilli EE, Nagaraju K, Martin GR, Hoffman EP. Dystrophin-deficient cardiomyopathy in mouse: expression of Nox4 and Lox are associated with fibrosis and altered functional parameters in the heart. Neuromuscul Disord. 2008;18:371–81.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Williams IA, Allen DG. The role of reactive oxygen species in the hearts of dystrophin-deficient mdx mice. Am J Physiol Heart Circ Physiol. 2007;293:H1969–77.

    Article  CAS  PubMed  Google Scholar 

  61. Nediani C, Raimondi L, Borchi E, Cerbai E. Nitric oxide/reactive oxygen species generation and nitroso/redox imbalance in heart failure: from molecular mechanisms to therapeutic implications. Antioxid Redox Signal. 2011;14:289–331.

    Article  CAS  PubMed  Google Scholar 

  62. Gao WD, Liu Y, Marban E. Selective effects of oxygen free radicals on excitation-contraction coupling in ventricular muscle. Implications for the mechanism of stunned myocardium. Circulation. 1996;94:2597–604.

    Article  CAS  PubMed  Google Scholar 

  63. Aikawa R, Komuro I, Yamazaki T, Zou Y, Kudoh S, Tanaka M, Shiojima I, Hiroi Y, Yazaki Y. Oxidative stress activates extracellular signal-regulated kinases through Src and Ras in cultured cardiac myocytes of neonatal rats. J Clin Invest. 1997;100:1813–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Zima AV, Blatter LA. Redox regulation of cardiac calcium channels and transporters. Cardiovasc Res. 2006;71:310–21.

    Article  CAS  PubMed  Google Scholar 

  65. Momose M, Iguchi N, Imamura K, Usui H, Ueda T, Miyamoto K, Inaba S. Depressed myocardial fatty acid metabolism in patients with muscular dystrophy. Neuromuscul Disord. 2001;11:464–9.

    Article  CAS  PubMed  Google Scholar 

  66. Perloff JK, Henze E, Schelbert HR. Alterations in regional myocardial metabolism, perfusion, and wall motion in Duchenne muscular dystrophy studied by radionuclide imaging. Circulation. 1984;69:33–42.

    Article  CAS  PubMed  Google Scholar 

  67. Quinlivan RM, Lewis P, Marsden P, Dundas R, Robb SA, Baker E, Maisey M. Cardiac function, metabolism and perfusion in Duchenne and Becker muscular dystrophy. Neuromuscul Disord. 1996;6:237–46.

    Article  CAS  PubMed  Google Scholar 

  68. Khairallah M, Labarthe F, Bouchard B, Danialou G, Petrof BJ, Des RC. Profiling substrate fluxes in the isolated working mouse heart using 13C-labeled substrates: focusing on the origin and fate of pyruvate and citrate carbons. Am J Physiol Heart Circ Physiol. 2004;286:H1461–70.

    Article  CAS  PubMed  Google Scholar 

  69. Wehling M, Spencer MJ, Tidball JG. A nitric oxide synthase transgene ameliorates muscular dystrophy in mdx mice. J Cell Biol. 2001;155:123–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Xu KY, Huso DL, Dawson TM, Bredt DS, Becker LC. Nitric oxide synthase in cardiac sarcoplasmic reticulum. Proc Natl Acad Sci U S A. 1999;96:657–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Elfering SL, Sarkela TM, Giulivi C. Biochemistry of mitochondrial nitric-oxide synthase. J Biol Chem. 2002;277:38079–86.

    Article  CAS  PubMed  Google Scholar 

  72. Wehling-Henricks M, Jordan MC, Roos KP, Deng B, Tidball JG. Cardiomyopathy in dystrophin-deficient hearts is prevented by expression of a neuronal nitric oxide synthase transgene in the myocardium. Hum Mol Genet. 2005;14:1921–33.

    Article  CAS  PubMed  Google Scholar 

  73. Ascah A, Khairallah M, Daussin F, Bourcier-Lucas C, Godin R, Allen BG, Petrof BJ, Des Rosiers C, Burelle Y. Stress-induced opening of the permeability transition pore in the dystrophin-deficient heart is attenuated by acute treatment with sildenafil. Am J Physiol Heart Circ Physiol. 2011;300:H144–53.

    Article  CAS  PubMed  Google Scholar 

  74. Khairallah M, Khairallah RJ, Young ME, Allen BG, Gillis MA, Danialou G, Deschepper CF, Petrof BJ, Des RC. Sildenafil and cardiomyocyte-specific cGMP signaling prevent cardiomyopathic changes associated with dystrophin deficiency. Proc Natl Acad Sci U S A. 2008;105:7028–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Adamo CM, Dai DF, Percival JM, Minami E, Willis MS, Patrucco E, Froehner SC, Beavo JA. Sildenafil reverses cardiac dysfunction in the mdx mouse model of Duchenne muscular dystrophy. Proc Natl Acad Sci U S A. 2010;107:19079–83.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Briggs R, King TJ. Nuclear transplantation studies on the early gastrula (Rana pipiens). I. Nuclei of presumptive endoderm. Dev Biol. 1960;2:252–70.

    Article  CAS  PubMed  Google Scholar 

  77. Gurdon JB, Elsdale TR, Fischberg M. Sexually mature individuals of Xenopus laevis from the transplantation of single somatic nuclei. Nature. 1958;182:64–5.

    Article  CAS  PubMed  Google Scholar 

  78. Miller RA, Ruddle FH. Pluripotent teratocarcinoma-thymus somatic cell hybrids. Cell. 1976;9:45–55.

    Article  CAS  PubMed  Google Scholar 

  79. Blau HM, Chiu CP, Webster C. Cytoplasmic activation of human nuclear genes in stable heterocaryons. Cell. 1983;32:1171–80.

    Article  CAS  PubMed  Google Scholar 

  80. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663–76.

    Article  CAS  PubMed  Google Scholar 

  81. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, Yamanaka S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861–72.

    Article  CAS  PubMed  Google Scholar 

  82. Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, Nie J, Jonsdottir GA, Ruotti V, Stewart R, Slukvin II, Thomson JA. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007;318:1917–20.

    Article  CAS  PubMed  Google Scholar 

  83. Kim K, Doi A, Wen B, Ng K, Zhao R, Cahan P, Kim J, Aryee MJ, Ji H, Ehrlich LI, Yabuuchi A, Takeuchi A, Cunniff KC, Hongguang H, McKinney-Freeman S, Naveiras O, Yoon TJ, Irizarry RA, Jung N, Seita J, Hanna J, Murakami P, Jaenisch R, Weissleder R, Orkin SH, Weissman IL, Feinberg AP, Daley GQ. Epigenetic memory in induced pluripotent stem cells. Nature. 2010;467:285–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Kim K, Zhao R, Doi A, Ng K, Unternaehrer J, Cahan P, Hongguang H, Loh YH, Aryee MJ, Lensch MW, Li H, Collins JJ, Feinberg AP, Daley GQ. Donor cell type can influence the epigenome and differentiation potential of human induced pluripotent stem cells. Nat Biotechnol. 2011;29(12):1117–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Marchetto MC, Yeo GW, Kainohana O, Marsala M, Gage FH, Muotri AR. Transcriptional signature and memory retention of human-induced pluripotent stem cells. PLoS ONE. 2009;4, e7076.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  86. Ohi Y, Qin H, Hong C, Blouin L, Polo JM, Guo T, Qi Z, Downey SL, Manos PD, Rossi DJ, Yu J, Hebrok M, Hochedlinger K, Costello JF, Song JS, Ramalho-Santos M. Incomplete DNA methylation underlies a transcriptional memory of somatic cells in human iPS cells. Nat Cell Biol. 2011;13:541–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Polo JM, Liu S, Figueroa ME, Kulalert W, Eminli S, Tan KY, Apostolou E, Stadtfeld M, Li Y, Shioda T, Natesan S, Wagers AJ, Melnick A, Evans T, Hochedlinger K. Cell type of origin influences the molecular and functional properties of mouse induced pluripotent stem cells. Nat Biotechnol. 2010;28:848–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Zhao XY, Li W, Lv Z, Liu L, Tong M, Hai T, Hao J, Guo CL, Ma QW, Wang L, Zeng F, Zhou Q. iPS cells produce viable mice through tetraploid complementation. Nature. 2009;461:86–90.

    Article  CAS  PubMed  Google Scholar 

  89. Kim JB, Greber B, Arauzo-Bravo MJ, Meyer J, Park KI, Zaehres H, Scholer HR. Direct reprogramming of human neural stem cells by OCT4. Nature. 2009;461:649–53.

    Article  CAS  PubMed  Google Scholar 

  90. Liu H, Ye Z, Kim Y, Sharkis S, Jang YY. Generation of endoderm-derived human induced pluripotent stem cells from primary hepatocytes. Hepatology. 2010;51:1810–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Mukamel Z, Hagai T, Gilad S, Amann-Zalcenstein D, Tanay A, Amit I, Novershtern N, Hanna JH. Deterministic direct reprogramming of somatic cells to pluripotency. Nature. 2013;502:65–70.

    Article  PubMed  CAS  Google Scholar 

  92. Yang R, Zheng Y, Li L, Liu S, Burrows M, Wei Z, Nace A, Herlyn M, Cui R, Guo W, Cotsarelis G, Xu X. Direct conversion of mouse and human fibroblasts to functional melanocytes by defined factors. Nat Commun. 2014;5:5807.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Lemper M, Leuckx G, Heremans Y, German MS, Heimberg H, Bouwens L, Baeyens L. Reprogramming of human pancreatic exocrine cells to beta-like cells. Cell Death Differ. 2014;22(7):1117–30.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Pulecio J, Nivet E, Sancho-Martinez I, Vitaloni M, Guenechea G, Xia Y, Kurian L, Dubova I, Bueren J, Laricchia-Robbio L, Izpisua Belmonte JC. Conversion of human fibroblasts into monocyte-like progenitor cells. Stem Cells. 2014;32:2923–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Kim YJ, Lim H, Li Z, Oh Y, Kovlyagina I, Choi IY, Dong X, Lee G. Generation of multipotent induced neural crest by direct reprogramming of human postnatal fibroblasts with a single transcription factor. Cell Stem Cell. 2014;15:497–506.

    Article  CAS  PubMed  Google Scholar 

  96. Sandler VM, Lis R, Liu Y, Kedem A, James D, Elemento O, Butler JM, Scandura JM, Rafii S. Reprogramming human endothelial cells to haematopoietic cells requires vascular induction. Nature. 2014;511:312–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Huang P, Zhang L, Gao Y, He Z, Yao D, Wu Z, Cen J, Chen X, Liu C, Hu Y, Lai D, Hu Z, Chen L, Zhang Y, Cheng X, Ma X, Pan G, Wang X, Hui L. Direct reprogramming of human fibroblasts to functional and expandable hepatocytes. Cell Stem Cell. 2014;14:370–84.

    Article  CAS  PubMed  Google Scholar 

  98. Du Y, Wang J, Jia J, Song N, Xiang C, Xu J, Hou Z, Su X, Liu B, Jiang T, Zhao D, Sun Y, Shu J, Guo Q, Yin M, Sun D, Lu S, Shi Y, Deng H. Human hepatocytes with drug metabolic function induced from fibroblasts by lineage reprogramming. Cell Stem Cell. 2014;14:394–403.

    Article  CAS  PubMed  Google Scholar 

  99. Zhang K, Liu GH, Yi F, Montserrat N, Hishida T, Esteban CR, Izpisua Belmonte JC. Direct conversion of human fibroblasts into retinal pigment epithelium-like cells by defined factors. Protein Cell. 2014;5:48–58.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. Liu ML, Zang T, Zou Y, Chang JC, Gibson JR, Huber KM, Zhang CL. Small molecules enable neurogenin 2 to efficiently convert human fibroblasts into cholinergic neurons. Nat Commun. 2013;4:2183.

    PubMed  PubMed Central  Google Scholar 

  101. Wada R, Muraoka N, Inagawa K, Yamakawa H, Miyamoto K, Sadahiro T, Umei T, Kaneda R, Suzuki T, Kamiya K, Tohyama S, Yuasa S, Kokaji K, Aeba R, Yozu R, Yamagishi H, Kitamura T, Fukuda K, Ieda M. Induction of human cardiomyocyte-like cells from fibroblasts by defined factors. Proc Natl Acad Sci U S A. 2013;110:12667–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Hendry CE, Vanslambrouck JM, Ineson J, Suhaimi N, Takasato M, Rae F, Little MH. Direct transcriptional reprogramming of adult cells to embryonic nephron progenitors. J Am Soc Nephrol. 2013;24:1424–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Nam YJ, Song K, Luo X, Daniel E, Lambeth K, West K, Hill JA, DiMaio JM, Baker LA, Bassel-Duby R, Olson EN. Reprogramming of human fibroblasts toward a cardiac fate. Proc Natl Acad Sci U S A. 2013;110:5588–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Islas JF, Liu Y, Weng KC, Robertson MJ, Zhang S, Prejusa A, Harger J, Tikhomirova D, Chopra M, Iyer D, Mercola M, Oshima RG, Willerson JT, Potaman VN, Schwartz RJ. Transcription factors ETS2 and MESP1 transdifferentiate human dermal fibroblasts into cardiac progenitors. Proc Natl Acad Sci U S A. 2012;109:13016–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Ring KL, Tong LM, Balestra ME, Javier R, Andrews-Zwilling Y, Li G, Walker D, Zhang WR, Kreitzer AC, Huang Y. Direct reprogramming of mouse and human fibroblasts into multipotent neural stem cells with a single factor. Cell Stem Cell. 2012;11:100–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Liu X, Li F, Stubblefield EA, Blanchard B, Richards TL, Larson GA, He Y, Huang Q, Tan AC, Zhang D, Benke TA, Sladek JR, Zahniser NR, Li CY. Direct reprogramming of human fibroblasts into dopaminergic neuron-like cells. Cell Res. 2012;22:321–32.

    Article  CAS  PubMed  Google Scholar 

  107. Son EY, Ichida JK, Wainger BJ, Toma JS, Rafuse VF, Woolf CJ, Eggan K. Conversion of mouse and human fibroblasts into functional spinal motor neurons. Cell Stem Cell. 2011;9:205–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Ambasudhan R, Talantova M, Coleman R, Yuan X, Zhu S, Lipton Stuart A, Ding S. Direct reprogramming of adult human fibroblasts to functional neurons under defined conditions. Cell Stem Cell. 2011;9:113–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Yoo AS, Sun AX, Li L, Shcheglovitov A, Portmann T, Li Y, Lee-Messer C, Dolmetsch RE, Tsien RW, Crabtree GR. MicroRNA-mediated conversion of human fibroblasts to neurons. Nature. 2011;476:228–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Tapscott SJ, Davis RL, Thayer MJ, Cheng PF, Weintraub H, Lassar AB. MyoD1: a nuclear phosphoprotein requiring a Myc homology region to convert fibroblasts to myoblasts. Science. 1988;242:405–11.

    Article  CAS  PubMed  Google Scholar 

  111. Qian L, Huang Y, Spencer CI, Foley A, Vedantham V, Liu L, Conway SJ, Fu J-D, Srivastava D. In vivo reprogramming of murine cardiac fibroblasts into induced cardiomyocytes. Nature. 2012;485:593–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Li W, Nakanishi M, Zumsteg A, Shear M, Wright C, Melton DA, Zhou Q. In vivo reprogramming of pancreatic acinar cells to three islet endocrine subtypes. Elife. 2014;3, e01846.

    PubMed  PubMed Central  Google Scholar 

  113. Zhou Q, Brown J, Kanarek A, Rajagopal J, Melton DA. In vivo reprogramming of adult pancreatic exocrine cells to beta-cells. Nature. 2008;455:627–32.

    Article  CAS  PubMed  Google Scholar 

  114. De la Rossa A, Bellone C, Golding B, Vitali I, Moss J, Toni N, Luscher C, Jabaudon D. In vivo reprogramming of circuit connectivity in postmitotic neocortical neurons. Nat Neurosci. 2013;16:193–200.

    Article  PubMed  CAS  Google Scholar 

  115. Guo Z, Zhang L, Wu Z, Chen Y, Wang F, Chen G. In vivo direct reprogramming of reactive glial cells into functional neurons after brain injury and in an Alzheimer's disease model. Cell Stem Cell. 2014;14:188–202.

    Article  CAS  PubMed  Google Scholar 

  116. Murry CE, Keller G. Differentiation of embryonic stem cells to clinically relevant populations: lessons from embryonic development. Cell. 2008;132:661–80.

    Article  CAS  PubMed  Google Scholar 

  117. Laflamme MA, Murry CE. Regenerating the heart. Nat Biotechnol. 2005;23:845–56.

    Article  CAS  PubMed  Google Scholar 

  118. Kehat I, Kenyagin-Karsenti D, Snir M, Segev H, Amit M, Gepstein A, Livne E, Binah O, Itskovitz-Eldor J, Gepstein L. Human embryonic stem cells can differentiate into myocytes with structural and functional properties of cardiomyocytes. J Clin Invest. 2001;108:407–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Zhang J, Wilson GF, Soerens AG, Koonce CH, Yu J, Palecek SP, Thomson JA, Kamp TJ. Functional cardiomyocytes derived from human induced pluripotent stem cells. Circ Res. 2009;104:e30–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Burridge Paul W, Keller G, Gold Joseph D, Wu Joseph C. Production of de novo cardiomyocytes: human pluripotent stem cell differentiation and direct reprogramming. Cell Stem Cell. 2012;10:16–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Lian X, Hsiao C, Wilson G, Zhu K, Hazeltine LB, Azarin SM, Raval KK, Zhang J, Kamp TJ, Palecek SP. Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling. Proc Natl Acad Sci U S A. 2012;109(27):E1848–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Beqqali A, Kloots J, Ward-van Oostwaard D, Mummery C, Passier R. Genome-wide transcriptional profiling of human embryonic stem cells differentiating to cardiomyocytes. Stem Cells. 2006;24:1956–67.

    Article  CAS  PubMed  Google Scholar 

  123. Burridge PW, Thompson S, Millrod MA, Weinberg S, Yuan X, Peters A, Mahairaki V, Koliatsos VE, Tung L, Zambidis ET. A universal system for highly efficient cardiac differentiation of human induced pluripotent stem cells that eliminates interline variability. PLoS ONE. 2011;6, e18293.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Uosaki H, Fukushima H, Takeuchi A, Matsuoka S, Nakatsuji N, Yamanaka S, Yamashita JK. Efficient and scalable purification of cardiomyocytes from human embryonic and induced pluripotent stem cells by VCAM1 surface expression. PLoS ONE. 2011;6, e23657.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Yang L, Soonpaa MH, Adler ED, Roepke TK, Kattman SJ, Kennedy M, Henckaerts E, Bonham K, Abbott GW, Linden RM, Field LJ, Keller GM. Human cardiovascular progenitor cells develop from a KDR+ embryonic-stem-cell-derived population. Nature. 2008;453:524–8.

    Article  CAS  PubMed  Google Scholar 

  126. Ma J, Guo L, Fiene SJ, Anson BD, Thomson JA, Kamp TJ, Kolaja KL, Swanson BJ, January CT. High purity human-induced pluripotent stem cell-derived cardiomyocytes: electrophysiological properties of action potentials and ionic currents. Am J Physiol Heart Circ Physiol. 2011;301:H2006–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Liu J, Sun N, Bruce MA, Wu JC, Butte MJ. Atomic force mechanobiology of pluripotent stem cell-derived cardiomyocytes. PLoS ONE. 2012;7, e37559.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Pillekamp F, Haustein M, Khalil M, Emmelheinz M, Nazzal R, Adelmann R, Nguemo F, Rubenchyk O, Pfannkuche K, Matzkies M, Reppel M, Bloch W, Brockmeier K, Hescheler J. Contractile properties of early human embryonic stem cell-derived cardiomyocytes: beta-adrenergic stimulation induces positive chronotropy and lusitropy but not inotropy. Stem Cells Dev. 2012;21(12):2111–21.

    Article  CAS  PubMed  Google Scholar 

  129. Klug MG, Soonpaa MH, Koh GY, Field LJ. Genetically selected cardiomyocytes from differentiating embronic stem cells form stable intracardiac grafts. J Clin Invest. 1996;98:216–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Cao F, Wagner RA, Wilson KD, Xie X, Fu JD, Drukker M, Lee A, Li RA, Gambhir SS, Weissman IL, Robbins RC, Wu JC. Transcriptional and functional profiling of human embryonic stem cell-derived cardiomyocytes. PLoS ONE. 2008;3, e3474.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  131. Binah O, Dolnikov K, Sadan O, Shilkrut M, Zeevi-Levin N, Amit M, Danon A, Itskovitz-Eldor J. Functional and developmental properties of human embryonic stem cells-derived cardiomyocytes. J Electrocardiol. 2007;40:S192–6.

    Article  PubMed  Google Scholar 

  132. He JQ, Ma Y, Lee Y, Thomson JA, Kamp TJ. Human embryonic stem cells develop into multiple types of cardiac myocytes: action potential characterization. Circ Res. 2003;93:32–9.

    Article  CAS  PubMed  Google Scholar 

  133. Satin J, Itzhaki I, Rapoport S, Schroder EA, Izu L, Arbel G, Beyar R, Balke CW, Schiller J, Gepstein L. Calcium handling in human embryonic stem cell-derived cardiomyocytes. Stem Cells. 2008;26:1961–72.

    Article  CAS  PubMed  Google Scholar 

  134. Guan X, Mack DL, Moreno CM, Strande JL, Mathieu J, Shi Y, Markert CD, Wang Z, Liu G, Lawlor MW, Moorefield EC, Jones TN, Fugate JA, Furth ME, Murry CE, Ruohola-Baker H, Zhang Y, Santana LF, Childers MK. Dystrophin-deficient cardiomyocytes derived from human urine: new biologic reagents for drug discovery. Stem Cell Res. 2014;12:467–80.

    Article  CAS  PubMed  Google Scholar 

  135. Zhang Y, McNeill E, Tian H, Soker S, Andersson KE, Yoo JJ, Atala A. Urine derived cells are a potential source for urological tissue reconstruction. J Urol. 2008;180:2226–33.

    Article  CAS  PubMed  Google Scholar 

  136. Bharadwaj S, Liu G, Shi Y, Wu R, Yang B, He T, Fan Y, Lu X, Zhou X, Liu H, Atala A, Rohozinski J, Zhang Y. Multipotential differentiation of human urine-derived stem cells: potential for therapeutic applications in urology. Stem Cells. 2013;31:1840–56.

    Article  CAS  PubMed  Google Scholar 

  137. Laflamme MA, Chen KY, Naumova AV, Muskheli V, Fugate JA, Dupras SK, Reinecke H, Xu C, Hassanipour M, Police S, O'Sullivan C, Collins L, Chen Y, Minami E, Gill EA, Ueno S, Yuan C, Gold J, Murry CE. Cardiomyocytes derived from human embryonic stem cells in pro-survival factors enhance function of infarcted rat hearts. Nat Biotechnol. 2007;25:1015–24.

    Article  CAS  PubMed  Google Scholar 

  138. Lee G, Papapetrou EP, Kim H, Chambers SM, Tomishima MJ, Fasano CA, Ganat YM, Menon J, Shimizu F, Viale A, Tabar V, Sadelain M, Studer L. Modelling pathogenesis and treatment of familial dysautonomia using patient-specific iPSCs. Nature. 2009;461:402–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Marchetto MC, Carromeu C, Acab A, Yu D, Yeo GW, Mu Y, Chen G, Gage FH, Muotri AR. A model for neural development and treatment of Rett syndrome using human induced pluripotent stem cells. Cell. 2010;143:527–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Brennand KJ, Simone A, Jou J, Gelboin-Burkhart C, Tran N, Sangar S, Li Y, Mu Y, Chen G, Yu D, McCarthy S, Sebat J, Gage FH. Modelling schizophrenia using human induced pluripotent stem cells. Nature. 2011;473:221–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Cooper O, Seo H, Andrabi S, Guardia-Laguarta C, Graziotto J, Sundberg M, McLean JR, Carrillo-Reid L, Xie Z, Osborn T, Hargus G, Deleidi M, Lawson T, Bogetofte H, Perez-Torres E, Clark L, Moskowitz C, Mazzulli J, Chen L, Volpicelli-Daley L, Romero N, Jiang H, Uitti RJ, Huang Z, Opala G, Scarffe LA, Dawson VL, Klein C, Feng J, Ross OA, Trojanowski JQ, Lee VM-Y, Marder K, Surmeier DJ, Wszolek ZK, Przedborski S, Krainc D, Dawson TM, Isacson O. Pharmacological rescue of mitochondrial deficits in iPSC-derived neural cells from patients with familial Parkinson’s disease. Sci Transl Med. 2012;4, 141ra190.

    Article  CAS  Google Scholar 

  142. Kaplitt MG, Feigin A, Tang C, Fitzsimons HL, Mattis P, Lawlor PA, Bland RJ, Young D, Strybing K, Eidelberg D, During MJ. Safety and tolerability of gene therapy with an adeno-associated virus (AAV) borne GAD gene for Parkinson's disease: an open label, phase I trial. Lancet. 2007;369:2097–105.

    Article  CAS  PubMed  Google Scholar 

  143. Ryan Scott D, Dolatabadi N, Chan Shing F, Zhang X, Akhtar Mohd W, Parker J, Soldner F, Sunico Carmen R, Nagar S, Talantova M, Lee B, Lopez K, Nutter A, Shan B, Molokanova E, Zhang Y, Han X, Nakamura T, Masliah E, Yates John R, Nakanishi N, Andreyev Aleksander Y, Okamoto S-i, Jaenisch R, Ambasudhan R, Lipton Stuart A. Isogenic human iPSC Parkinson s model shows nitrosative stress-induced dysfunction in MEF2-PGC1± transcription. Cell. 2013;155:1351–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Soldner F, Laganière J, Cheng Albert W, Hockemeyer D, Gao Q, Alagappan R, Khurana V, Golbe Lawrence I, Myers Richard H, Lindquist S, Zhang L, Guschin D, Fong Lauren K, Vu BJ, Meng X, Urnov Fyodor D, Rebar Edward J, Gregory Philip D, Zhang HS, Jaenisch R. Generation of isogenic pluripotent stem cells differing exclusively at two early onset Parkinson point mutations. Cell. 2011;146:318–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Yazawa M, Hsueh B, Jia X, Pasca AM, Bernstein JA, Hallmayer J, Dolmetsch RE. Using induced pluripotent stem cells to investigate cardiac phenotypes in Timothy syndrome. Nature. 2011;471:230–4.

    Google Scholar 

  146. Malan D, Friedrichs S, Fleischmann BK, Sasse P. Cardiomyocytes obtained from induced pluripotent stem cells with long-QT syndrome 3 recapitulate typical disease-specific features in vitro. Circ Res. 2011;109:841–7.

    Article  CAS  PubMed  Google Scholar 

  147. Sacco A, Mourkioti F, Tran R, Choi J, Llewellyn M, Kraft P, Shkreli M, Delp S, Pomerantz JH, Artandi SE, Blau HM. Short telomeres and stem cell exhaustion model Duchenne muscular dystrophy in mdx/mTR mice. Cell. 2010;143:1059–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Braunwald E. Biomarkers in heart failure. N Engl J Med. 2008;358:2148–59.

    Article  CAS  PubMed  Google Scholar 

  149. Coca SG, Yalavarthy R, Concato J, Parikh CR. Biomarkers for the diagnosis and risk stratification of acute kidney injury: a systematic review. Kidney Int. 2008;73:1008–16.

    Article  CAS  PubMed  Google Scholar 

  150. Berg JS, Khoury MJ, Evans JP. Deploying whole genome sequencing in clinical practice and public health: meeting the challenge one bin at a time. Genet Med. 2011;13:499–504.

    Article  PubMed  Google Scholar 

  151. Rizzo JM, Buck MJ. Key principles and clinical applications of “next-generation” DNA sequencing. Cancer Prev Res. 2012;5:887–900.

    Article  CAS  Google Scholar 

  152. Kim C, Wong J, Wen J, Wang S, Wang C, Spiering S, Kan NG, Forcales S, Puri PL, Leone TC, Marine JE, Calkins H, Kelly DP, Judge DP, Chen HS. Studying arrhythmogenic right ventricular dysplasia with patient-specific iPSCs. Nature. 2013;494:105–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Lan F, Lee AS, Liang P, Sanchez-Freire V, Nguyen PK, Wang L, Han L, Yen M, Wang Y, Sun N, Abilez OJ, Hu S, Ebert AD, Navarrete EG, Simmons CS, Wheeler M, Pruitt B, Lewis R, Yamaguchi Y, Ashley EA, Bers DM, Robbins RC, Longaker MT, Wu JC. Abnormal calcium handling properties underlie familial hypertrophic cardiomyopathy pathology in patient-specific induced pluripotent stem cells. Cell Stem Cell. 2013;12:101–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Wang Y, Liang P, Lan F, Wu H, Lisowski L, Gu M, Hu S, Kay MA, Urnov FD, Shinnawi R, Gold JD, Gepstein L, Wu JC. Genome editing of isogenic human induced pluripotent stem cells recapitulates long QT phenotype for drug testing. J Am Coll Cardiol. 2014;64:451–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Yang YM, Gupta SK, Kim KJ, Powers BE, Cerqueira A, Wainger BJ, Ngo HD, Rosowski KA, Schein PA, Ackeifi CA, Arvanites AC, Davidow LS, Woolf CJ, Rubin LL. A small molecule screen in stem-cell-derived motor neurons identifies a kinase inhibitor as a candidate therapeutic for ALS. Cell Stem Cell. 2013;12:713–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, Hsu PD, Wu X, Jiang W, Marraffini LA, Zhang F. Multiplex genome engineering using CRISPR/Cas systems. Science. 2013;339:819–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Gonzalez F, Zhu Z, Shi ZD, Lelli K, Verma N, Li QV, Huangfu D. An iCRISPR platform for rapid, multiplexable, and inducible genome editing in human pluripotent stem cells. Cell Stem Cell. 2014;15:215–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Li HL, Fujimoto N, Sasakawa N, Shirai S, Ohkame T, Sakuma T, Tanaka M, Amano N, Watanabe A, Sakurai H, Yamamoto T, Yamanaka S, Hotta A. Precise correction of the dystrophin gene in Duchenne muscular dystrophy patient induced pluripotent stem cells by TALEN and CRISPR-Cas9. Stem Cell Rep. 2014;4(1):143–54.

    Article  CAS  Google Scholar 

  159. Veres A, Gosis BS, Ding Q, Collins R, Ragavendran A, Brand H, Erdin S, Cowan CA, Talkowski ME, Musunuru K. Low incidence of off-target mutations in individual CRISPR-Cas9 and TALEN targeted human stem cell clones detected by whole-genome sequencing. Cell Stem Cell. 2014;15:27–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Filareto A, Parker S, Darabi R, Borges L, Iacovino M, Schaaf T, Mayerhofer T, Chamberlain JS, Ervasti JM, McIvor RS, Kyba M, Perlingeiro RC. An ex vivo gene therapy approach to treat muscular dystrophy using inducible pluripotent stem cells. Nat Commun. 2013;4:1549.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  161. Yamada S, Nelson TJ, Crespo-Diaz RJ, Perez-Terzic C, Liu XK, Miki T, Seino S, Behfar A, Terzic A. Embryonic stem cell therapy of heart failure in genetic cardiomyopathy. Stem Cells. 2008;26:2644–53.

    Article  PubMed  PubMed Central  Google Scholar 

  162. Moretti A, Bellin M, Welling A, Jung CB, Lam JT, Bott-Flugel L, Dorn T, Goedel A, Hohnke C, Hofmann F, Seyfarth M, Sinnecker D, Schomig A, Laugwitz KL. Patient-specific induced pluripotent stem-cell models for long-QT syndrome. N Engl J Med. 2010;363:1397–409.

    Article  CAS  PubMed  Google Scholar 

  163. Young LE, Platzer RF, Ervin DM, Izzo MJ. Hereditary spherocytosis. II. Observations on the role of the spleen. Blood. 1951 Nov;6(11):1099–1113.

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Martin K. Childers .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2016 Springer Science+Business Media New York

About this chapter

Cite this chapter

Guan, X., Mack, D., Childers, M.K. (2016). Patient-Derived Induced Pluripotent Stem Cells Provide a Regenerative Medicine Platform for Duchenne Muscular Dystrophy Heart Failure. In: Childers, M. (eds) Regenerative Medicine for Degenerative Muscle Diseases. Stem Cell Biology and Regenerative Medicine. Humana Press, New York, NY. https://doi.org/10.1007/978-1-4939-3228-3_6

Download citation

Publish with us

Policies and ethics