GET THE APP

Induced Pluripotent Stem Cells: To Model, To Treat
Journal of Cell Science & Therapy

Journal of Cell Science & Therapy
Open Access

ISSN: 2157-7013

+44 1300 500008

Editorial - (2011) Volume 2, Issue 4

Induced Pluripotent Stem Cells: To Model, To Treat

Hansen Wang*
Faculty of Medicine, University of Toronto, 1 King’s College Circle, Toronto, Ontario, Canada
*Corresponding Author: Hansen Wang, PhD, Faculty of Medicine, University of Toronto, 1 King’s College Circle, Toronto, Ontario, M5S 1A8, Canada Email:

Abstract

The development of the ability to convert somatic cells into induced pluripotent stem cells (iPSCs) through expression of a small combination of transcription factors is a major breakthrough in the field of stem cell research. It has raised the possibilities of providing personalized cells for the study and treatment of numerous human diseases. In recent years, iPSCs have been derived from patients of a large variety of disorders. Here, the progresses that has been made in establishing iPSC-based disease models and the potentials of iPSC technology for personalized medicine, drug discovery and cell therapy are reviewed. The challenges from iPSC technology are also briefly discussed.

Introduction

In 2006, the groundbreaking work by Takahashi and Yamanaka showed that retroviral expression of a set of four genes (Oct4, Sox2, Klf4 and c-Myc) converted somatic cells into a pluripotent state. Similar to the embryonic stem cells (ESCs), these cells exhibit the ability to self-renew endlessly and give rise to a multitude of cell types [1]. The remarkable progress in reprogramming technology over the past few years has facilitated the generation of iPSCs. iPSCs have been derived at increased efficiencies from several easily accessible human cell types, including blood cells, keratinocytes and dermal fibroblasts [2- 5]. The advances in iPSC technology has now opened new windows for developing human disease models, drug screening, as well as for providing a continuous autologous cell sources with potential for use in cell therapies [2,5-13].

Here, the increasing efforts in using iPSCs for human disease modeling and potentials of disease-specific iPSCs as platforms for disease mechanism discovery and drug screening are reviewed. In light of the prospects for iPSCs as a source of cell replacement in degenerative diseases, recent studies using iPSC-based therapy are also enumerated. Finally, the challenges for clinical applications of iPSC technology are also brought up to attentions.

iPSCs in Disease Modeling

One of the most promising features of somatic cell reprogramming is the possibility of using iPSCs to model human diseases in order to recapitulate their development, pathology, and pharmacological responsiveness. The ability to generate induced pluripotent cells from patients suffered from diseases of known and suspected aetiologies can allow people to obtain genetically matched cell types in unlimited quantity. In the past few years, great efforts have been put to generate iPSC lines from patients with a wide range of genetically inherited as well as sporadic diseases [2-4,14-16]. In most of these studies, iPSCs from patients have been successfully differentiated to the cell types relevant to the disorders. To date, there are many studies showing that patient-specific iPSCs do exhibit relevant disease features.

A hallmark of these studies was the generation of iPSCs from patients with spinal muscular atrophy (SMA), an inherited neurodegenerative disorder that affects motor neurons [17]. The disease was recapitulated by culturing motor neurons derived from SMA-iPSCs and demonstrating the degeneration of these motor neurons over time [17]. In another study, the enlargement of cardiomyocytes derived from iPSCs from patients with LEOPARD syndrome may reflect the hypertrophic cardiomyopathy associated with this disease [18]. In patients of Long QT syndrome (LQTS), QT intervals are increased on electrocardiography. The differentiated cardiomyocytes produced from iPSCs from these patients showed prolongation of action potential duration in electrophysiological assays [19,20]. LQTS-derived cells also showed marked arrhythmogenicity, characterized by early-after depolarizations and triggered arrhythmias [19]. In the studies of the disease phenotypes of iPSCs derived from Hutchinson-Gilford progeria patients, the premature senescence of the differentiated smooth muscle cells was found, indicating that vascular defects of patients can also be observed in vitro [21,22]. The iPSCs from patients of familial dysautonomia-derived exhibit decreased neurogenic differentiation and migration behaviours, compared with control iPSCs [7,23]. The tissue-specific alteration of gene splicing of the IKBKAP gene was also found in this study [23]. It thus provided the clues into the tissue specific pathology of familial dysautonomia. Similarly, iPSCs have been used to model many other neurological disorders [8,9,16,24-29], including schizophrenia [30-32], Alzheimer’s disease [33], amyotrophic lateral sclerosis [34], Rett syndrome [35-38], fragile X syndrome [39,40], X-linked adrenoleukodystrophy [41] and spinocerebellar ataxia [6]. These studies provide clear evidences that disease modelling using iPSC technology could be feasible.

iPSCs in Pharmaceutical and Cell Therapy

The iPSC technology not only allows us to study the pathological progression of the diseases, but also provides a new platform for testing the effects of drug treatment [3,12,13,42]. The identification of signal pathways or drugs that could affect the disease process will be the ultimate goal of iPSC technology. In the above mentioned study, the loss of neurons differentiated from iPSCs of SMA patients was ameliorated by treatment with small-molecule candidates that reverse disease features in other neuronal culture assays [17]. Likewise, the LQTS iPSC-derived cardiac-tissue model was used to evaluate the potency of existing and novel pharmacological agents that may either aggravate (potassium-channel blockers) or ameliorate (calciumchannel blockers, K(ATP)-channel openers and late sodium-channel blockers) the disease phenotype [19]. Key cellular and molecular elements of the schizophrenia phenotypes were ameliorated following treatment of schizophrenia iPSC derived neurons with the antipsychotic loxapine [31]. These studies illustrate the ability of iPSC technology to identify potential therapeutic agents. As such, it represents an inspiring paradigm for optimizing patient care (personalized medicine) and the development of drug screening.

Another exciting aspect of iPSC technology is the possibility of generating autologous cells for cell therapy. One of the excellent examples for advances in the development of iPSC-based therapies is related to Fanconi anemia (FA) [43]. The iPSCs derived from patients with FA (FA-iPSCs) can be generated and maintained only after correction of the FA-related gene. The study clearly showed that, after the introduction of the proper FA-related gene, FA-iPSCs could be differentiated into the hematopoietic lineage [43]. This study provided a model for the derivation of iPSCs from a patient, correction of the disease-associated mutation, and eventual provision of a continuous source for cell therapy. In another study, Kazuki et al. [44] corrected the defective gene in iPSCs derived from a patient with Duchenne muscular dystrophy (DMD-iPSCs) using human artificial chromosomes (HAC). The study suggested that combination of patient-specific iPSCs and HAC-containing defective genes represents a powerful tool for gene and cell therapies. With regards to transplantation of iPSCs for therapeutic regeneration, the most compelling study so far showed that haematopoietic cells derived from iPSCs can reduce the blood cell phenotype in a humanized mouse model of sickle cell anemia [45]. The Parkinson’s disease is characterized by the slow but steady decline in dopaminergic neurons. Wernig et al. [46] demonstrated that iPSCs could be induced to differentiate into dopamine neurons of midbrain character and were able to improve behavior in a rat model of Parkinson’s disease upon transplantation into the adult brain. Hargus et al. [11] developed the cell-based treatment approach by deriving iPSCs from Parkinson’s disease patients, differentiating these into dopaminergic neurons, and then transplanting the cells into the brains of rats with Parkinson’s disease. The iPSCs have also been utilized for treatment of stroke [47], hepatic failure [48] and myocardial infarction [49] in animal models.

Taken together, these studies provide strong support for the roles of iPSCs in both pharmaceutical and cell therapies of human diseases. These recent developments have brought the dreams of clinical applications of iPSCs much closer to reality.

Conclusion

Although studies in the field of iPSCs have been advanced dramatically, many issues still need to be addressed and improved in terms of the cell reprogramming, the differentiation potential of cells, and the future clinical application of iPSC technology [2,5,12,25,50]. The challenges especially involve the safety concerns regarding the generation of iPSCs and the introduction of iPSC-derived cells into patients. The iPSC technology has been proven to be a powerful approach for in vitro studies, it will hold enormous prospects for disease modeling, personalized medicine, drug screening and cellbased therapies, provided that all these fundamental challenges from iPSC technology are given due attention and priority.

Acknowledgements

This manuscript was prepared at the invitation of the Managing Editor of the Journal of Cell Science & Therapy.

References

  1. Takahashi K, Yamanaka S (2006) Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126: 663-676.
  2. Wu SM, Hochedlinger K (2011) Harnessing the potential of induced pluripotent stem cells for regenerative medicine. Nat Cell Biol 13: 497-505.
  3. Hussein SM, Nagy K, Nagy A (2011) Human induced pluripotent stem cells: the past, present, and future. Clin Pharmacol Ther 89: 741-745.
  4. Petit I, Salman Kesner N, Karry R, Robicsek O, Aberdam E, et al. (2012) Induced pluripotent stem cells from hair follicles as a cellular model for neurodevelopmental disorders. Stem Cell Res 8: 134-140.
  5. Barrilleaux B, Knoepfler PS (2011) Inducing iPSCs to escape the dish. Cell Stem Cell 9: 103-111.
  6. Koch P, Breuer P, Peitz M, Jungverdorben J, Kesavan J, et al. (2011) Excitationinduced ataxin-3 aggregation in neurons from patients with Machado-Joseph disease. Nature doi: 10.1038/nature10671.
  7. Lee G, Studer L (2011) Modelling familial dysautonomia in human induced pluripotent stem cells. Philos Trans R Soc Lond B Biol Sci 366: 2286-2296.
  8. Marchetto MC, Brennand KJ, Boyer LF, Gage FH (2011) Induced pluripotent stem cells (iPSCs) and neurological disease modeling: progress and promises. Hum Mol Genet 20: R109-R115.
  9. Marchetto MC, Carromeu C, Acab A, Yu D, Yeo GW, et al. (2011) A model for neural development and treatment of Rett syndrome using human induced pluripotent stem cells. Cell 143: 527-539.
  10. Pasca SP, Portmann T, Voineagu I, Yazawa M, Shcheglovitov A, et al. (2011) Using iPSC-derived neurons to uncover cellular phenotypes associated with Timothy syndrome. Nat Med 17: 1657-1662.
  11. Hargus G, Cooper O, Deleidi M, Levy A, Lee K, et al. (2010) Differentiated Parkinson patient-derived induced pluripotent stem cells grow in the adult rodent brain and reduce motor asymmetry in Parkinsonian rats. Proc Natl Acad Sci U S A 107: 15921-15926.
  12. Grskovic M, Javaherian A, Strulovici B, Daley GQ (2011) Induced pluripotent stem cells - opportunities for disease modelling and drug discovery. Nat Rev Drug Discov 10: 915-929.
  13. Inoue H, Yamanaka S (2011) The use of induced pluripotent stem cells in drug development. Clin Pharmacol Ther 89: 655-661.
  14. Sancho-Martinez I, Li M, Izpisua Belmonte JC (2011) Disease correction the iPSC way: advances in iPSC-based therapy. Clin Pharmacol Ther 89: 746-749.
  15. Josowitz R, Carvajal-Vergara X, Lemischka IR, Gelb BD (2011) Induced pluripotent stem cell-derived cardiomyocytes as models for genetic cardiovascular disorders. Curr Opin Cardiol 26: 223-229.
  16. Ming GL, Brüstle O, Muotri A, Studer L, Wernig M, et al. (2011) Cellular reprogramming: recent advances in modeling neurological diseases. J Neurosci 31: 16070-16075.
  17. Ebert AD, Yu J, Rose FF Jr, Mattis VB, Lorson CL, et al. (2009) Induced pluripotent stem cells from a spinal muscular atrophy patient. Nature 457: 277- 280.
  18. Carvajal-Vergara X, Sevilla A, D'Souza SL, Ang YS, Schaniel C, et al. (2010) Patient-specific induced pluripotent stem-cell-derived models of LEOPARD syndrome. Nature 465: 808-812.
  19. Itzhaki I, Maizels L, Huber I, Zwi-Dantsis L, Caspi O, et al. (2011) Modelling the long QT syndrome with induced pluripotent stem cells. Nature 471: 225-229.
  20. Moretti A, Bellin M, Welling A, Jung CB, Lam JT, et al. (2010) Patient-specific induced pluripotent stem-cell models for long-QT syndrome. N Engl J Med 363: 1397-1409.
  21. Liu GH, Barkho BZ, Ruiz S, Diep D, Qu J, et al. (2011) Recapitulation of premature ageing with iPSCs from Hutchinson-Gilford progeria syndrome. Nature 472: 221-225.
  22. Zhang J, Lian Q, Zhu G, Zhou F, Sui L, et al. (2011) A human iPSC model of Hutchinson Gilford Progeria reveals vascular smooth muscle and mesenchymal stem cell defects. Cell Stem Cell 8: 31-45.
  23. Lee G, Papapetrou EP, Kim H, Chambers SM, Tomishima MJ, et al. (2009) Modelling pathogenesis and treatment of familial dysautonomia using patientspecific iPSCs. Nature 461: 402-406.
  24. Cundiff PE, Anderson SA (2011) I Impact of induced pluripotent stem cells on the study of central nervous system disease. Curr Opin Genet Dev 21: 354-361.
  25. Dolmetsch R, Geschwind DH (2011) The human brain in a dish: the promise of iPSC-derived neurons. Cell 145: 831-834.
  26. Feng Z, Gao F (2011) Stem Cell Challenges in the Treatment of Neurodegenerative Disease. CNS Neurosci Ther doi: 10.1111/j.1755- 5949.2011.00239.
  27. Juopperi TA, Song H, Ming GL (2011) Modeling neurological diseases using patient-derived induced pluripotent stem cells. Future Neurol 6: 363-373.
  28. Lemonnier T, Blanchard S, Toli D, Roy E, Bigou S, et al. (2011) Modeling neuronal defects associated with a lysosomal disorder using patient-derived induced pluripotent stem cells. Hum Mol Genet 20: 3653-3666.
  29. Durnaoglu S, Genc S, Genc K (2011) Patient-specific pluripotent stem cells in neurological diseases. Stem Cells Int 2011:212487.
  30. Brennand KJ, Gage FH (2011) Concise review: the promise of human induced pluripotent stem cell-based studies of schizophrenia. Stem Cells 29: 1915- 1922.
  31. Brennand KJ, Simone A, Jou J, Gelboin-Burkhart C, Tran N, et al. (2011) Modelling schizophrenia using human induced pluripotent stem cells. Nature 473: 221-225.
  32. Pedrosa E, Sandler V, Shah A, Carroll R, Chang C, et al. (2011) Development of patient-specific neurons in schizophrenia using induced pluripotent stem cells. J Neurogenet 25: 88-103.
  33. Yagi T, Ito D, Okada Y, Akamatsu W, Nihei Y, et al. (2011) Modeling familial Alzheimer's disease with induced pluripotent stem cells. Hum Mol Genet 20: 4530-4539.
  34. Mitne-Neto M, Machado-Costa M, Marchetto MC, Bengtson MH, Joazeiro CA, et al. (2011) Downregulation of VAPB expression in motor neurons derived from induced pluripotent stem cells of ALS8 patients. Hum Mol Genet 20: 3642- 3652.
  35. Amenduni M, De Filippis R, Cheung AY, Disciglio V, Epistolato MC, et al. (2011) iPS cells to model CDKL5-related disorders. Eur J Hum Genet 19: 1246-1255.
  36. Ananiev G, Williams EC, Li H, Chang Q (2011) Isogenic pairs of wild type and mutant induced pluripotent stem cell (iPSC) lines from Rett syndrome patients as in vitro disease model. PLoS One 6: e25255.
  37. Cheung AY, Horvath LM, Grafodatskaya D, Pasceri P, Weksberg R, et al. (2011) Isolation of MECP2-null Rett Syndrome patient hiPS cells and isogenic controls through X-chromosome inactivation. Hum Mol Genet 20: 2103-2115.
  38. Kim KY, Hysolli E, Park IH (2011) Neuronal maturation defect in induced pluripotent stem cells from patients with Rett syndrome. Proc Natl Acad Sci U S A 108: 14169-14174.
  39. Sheridan SD, Theriault KM, Reis SA, Zhou F, Madison JM, et al. (2011) Epigenetic characterization of the FMR1 gene and aberrant neurodevelopment in human induced pluripotent stem cell models of fragile X syndrome. PLoS One 6: e26203.
  40. Urbach A, Bar-Nur O, Daley GQ, Benvenisty N (2010) Differential modeling of fragile X syndrome by human embryonic stem cells and induced pluripotent stem cells. Cell Stem Cell 6: 407-411.
  41. Jang J, Kang HC, Kim HS, Kim JY, Huh YJ, et al. (2011) Induced pluripotent stem cell models from X-linked adrenoleukodystrophy patients. Ann Neurol 70: 402-409.
  42. Nsair A, MacLellan WR (2011) Induced pluripotent stem cells for regenerative cardiovascular therapies and biomedical discovery. Adv Drug Deliv Rev 63: 324-330.
  43. Raya A, Rodríguez-Pizà I, Guenechea G, Vassena R, Navarro S, et al. (2009) Disease-corrected haematopoietic progenitors from Fanconi anaemia induced pluripotent stem cells. Nature 460: 53-59.
  44. Kazuki Y, Hiratsuka M, Takiguchi M, Osaki M, Kajitani N, et al. (2010) Complete genetic correction of ips cells from Duchenne muscular dystrophy. Mol Ther 18: 386-393.
  45. Hanna J, Wernig M, Markoulaki S, Sun CW, Meissner A, et al. (2007) Treatment of sickle cell anemia mouse model with iPS cells generated from autologous skin. Science 318: 1920-1923.
  46. Wernig M, Zhao JP, Pruszak J, Hedlund E, Fu D, et al. (2008) Neurons derived from reprogrammed fibroblasts functionally integrate into the fetal brain and improve symptoms of rats with Parkinson's disease. Proc Natl Acad Sci U S A 105: 5856-5861.
  47. Jensen MB, Yan H, Krishnaney-Davison R, Al Sawaf A, Zhang SC (2011) Survival and Differentiation of Transplanted Neural Stem Cells Derived from Human Induced Pluripotent Stem Cells in A Rat Stroke Model. J Stroke Cerebrovasc Dis [Epub ahead of print].
  48. Chiang CH, Chang CC, Huang HC, Chen YJ, Tsai PH, et al. (2011) Investigation of hepatoprotective activity of induced pluripotent stem cells in the mouse model of liver injury. J Biomed Biotechnol 2011: 219060.
  49. Nelson TJ, Martinez-Fernandez A, Yamada S, Perez-Terzic C, Ikeda Y, et al. (2009) Repair of acute myocardial infarction by human stemness factors induced pluripotent stem cells. Circulation 120: 408-416.
  50. Kiefer JC (2011) Primer and interviews: promises and realities of induced pluripotent stem cells. Dev Dyn 240: 2034-2041.
Citation: Wang H (2011) Induced Pluripotent Stem Cells: To Model, To Treat. J Cell Sci Ther 2:e103.

Copyright: © 2011 Wang H. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top