GET THE APP

Induced Pluripotent Stem Cells for the Treatment of Hemophilia A
Journal of Cell Science & Therapy

Journal of Cell Science & Therapy
Open Access

ISSN: 2157-7013

+44 1300 500008

Editorial - (2014) Volume 5, Issue 6

Induced Pluripotent Stem Cells for the Treatment of Hemophilia A

Bilgimol C Joseph and Manjunath S Rao*
Micro Therapeutic Research Labs Pvt Ltd, Chennai, Tamilnadu, India
*Corresponding Author: Manjunath S Rao, No: 6, 'Rajam Bhavanam, Kamarajar Salai, East Tambaram, Selaiyur, Chennai - 600 059, India, Tel: +91 44 2239 0070 / 71 / 72, Fax: +91 44 2239 0068 Email:

Abstract

Factor VIII, one of the most complex proteins known, plays a major role in blood coagulation pathway. Defects in factor VIII protein result in hemophilia A, a severe bleeding disorder. Plasma derived factor VIII or recombinant factor VIII has been used extensively for treating hemophilia A patients. Number of attempts at gene therapy for hemophilia A has failed for various unknown/not much studied reasons including immune rejection. Here, the progress that has been made in establishing iPSC-based disease models and the potentials of iPSC technology for personalized medicine and cell therapy for hemophilia A are reviewed. The challenges of iPSC technology are also briefly discussed.

Introduction

Hemophilia A is one of the most common genetic coagulation disorders arising due to the deficiency of factor VIII protein. It is estimated that 1 in 5,000 males are affected by hemophilia A [1]. It is caused by several genetic mutations, which include deletions, insertions, inversions and point mutations in the factor VIII gene (Haemophilia A Mutation, Structure, Test and Resource Site; http://hadb.org.uk). According to the severity of bleeding and time taken for clotting, hemophilia A can be characterized as severe, moderate or mild [1]. Currently, there is no cure for hemophilia A. The only available treatment for this disease is the infusion of or administration of recombinant factor VIII. However, the treatment with recombinant factor VIII is limited due the formation of factor VIII inactivating antibodies, exorbitant cost and requirement of frequent injections.

After the introduction of gene therapy, it is found that gene therapy is a promising option for the treatment of hemophilia A. Natwani et al. has used the adeno-associated virus vector (AAV) to deliver factor IX cDNA to correct hemophilia B. This method of delivery could not be used for hemophilia A patients since the size of full length factor VIII cDNA is too large and AAV cannot accommodate the large size of factor VIII cDNA. Besides, gene therapy is ideally used to correct genetic defects rather than to deliver a functional gene.

Usage of iPSC in hemophilia A

Another promising option for the treatment of hemophilia A is the introduction of patient-derived induced pluripotent stem cells (iPSCs). The defective gene can be corrected in iPSCs by using programmable nucleases like zinc ginger nucleases (ZFNs) [2-5], transcription activator-like effector nucleases [6-8] and clusters of regularly interspaced palindromic repeats [9-16]. In case of these programmable nucleases they cleave the chromosomal DNA in a targeted manner and produce DNA double stranded breaks. The nick will be repaired by endogenous mechanisms known as homologous recombination or non-homologous end-joining. Finally, it will result in the correction mutagenesis such as deletions [17,18], duplications and inversion [19]. These gene-corrected iPSCs are then allowed to differentiate into appropriate somatic cells before delivery to patients to ensure the expression of the functional gene.

Challenges of the technology

iPSCs have their own merits and demerits. Though iPSCs are mentioned to be the cells that will rule the future medical industry to provide patient-specific stem cells [20-22], there are controversies over its application to human subjects [23,24]. Challenges are encountered with the recent advancements which can harness its true activity for biomedical research to successfully formulate effective therapeutic approach [25]. It is worth to estimate the potentials of iPSCs as they are quite prominent being the readily accessible such as skin or blood which are enough to generate the disease-specific models. Existing challenges include the kinetics of disease onset and progression and also the spatial localization of the disease to create disease models which are commented can be tackled with advanced strategies such as gene modification, biomaterials, reprogramming etc. [26]. Researchers have been already working to correlate hemophilia A using patient-specific iPSCs because of its unlimited self-renewal and differentiation capabilities [27-31].

Conclusion

Approaches to generate an effective mechanism involving iPSCs are noteworthy to clearly define the potentials to tackle Hemophilia A and with priority attention over this much needed technology will play an eminent role in therapeutic scenario.

References

  1. Graw J, Brackmann HH, Oldenburg J, Schneppenheim R, Spannagl M, et al. (2005) Haemophilia A: from mutation analysis to new therapies. Nat Rev Genet 6: 488-501.
  2. Porteus MH, Baltimore D (2003) Chimeric nucleases stimulate gene targeting in human cells. Science 300: 763.
  3. Bibikova M, Beumer K, Trautman JK, Carroll D (2003) Enhancing gene targeting with designed zinc finger nucleases. Science 300: 764.
  4. Urnov FD, Miller JC, Lee YL, Beausejour CM, Rock JM, et al. (2005) Highly efficient endogenous human gene correction using designed zinc-finger nucleases. Nature 435: 646-651.
  5. Kim HJ, Lee HJ, Kim H, Cho SW, Kim JS (2009) Targeted genome editing in human cells with zinc finger nucleases constructed via modular assembly. Genome Res 19: 1279-1288.
  6. Miller JC, Tan S, Qiao G, Barlow KA, Wang J, et al. (2011) A TALE nuclease architecture for efficient genome editing. Nat Biotechnol 29: 143-148.
  7. Kim Y, Kweon J, Kim A, Chon JK, Yoo JY, et al. (2013) A library of TAL effector nucleases spanning the human genome. Nat Biotechnol 31: 251-258.
  8. Kim YK, Wee G, Park J, Kim J, Baek D, et al. (2013) TALEN-based knockout library for human microRNAs. Nat Struct Mol Biol 20: 1458-1464.
  9. Cho SW, Lee J, Carroll D, Kim JS, Lee J (2013) Heritable gene knockout in Caenorhabditiselegans by direct injection of Cas9-sgRNA ribonucleoproteins. Genetics 195: 1177-1180.
  10. Sung YH, Kim JM, Kim HT, Lee J, Jeon J, et al. (2014) Highly efficient gene knockout in mice and zebrafish with RNA-guided endonucleases. Genome Res 24: 125-131.
  11. Cho SW, Kim S, Kim Y, Kweon J, Kim HS, et al. (2014) Analysis of off-target effects of CRISPR/Cas-derived RNA-guided endonucleases and nickases. Genome Res 24: 132-141.
  12. Cho SW, Kim S, Kim JM, Kim JS (2013) Targeted genome engineering in human cells with the Cas9 RNA-guided endonuclease. Nat Biotechnol 31: 230-232.
  13. Cong L, Ran FA, Cox D, Lin S, Barretto R, et al. (2013) Multiplex genome engineering using CRISPR/Cas systems. Science 339: 819-823.
  14. Hwang WY, Fu Y, Reyon D, Maeder ML, Tsai SQ, et al. (2013) Efficient genome editing in zebrafish using a CRISPR-Cas system. Nat Biotechnol 31: 227-229.
  15. Jiang W, Bikard D, Cox D, Zhang F, Marraffini LA (2013) RNA-guided editing of bacterial genomes using CRISPR-Cas systems. Nat Biotechnol 31: 233-239.
  16. Mali P, Yang L, Esvelt KM, Aach J, Guell M, et al. (2013) RNA-guided human genome engineering via Cas9. Science 339: 823-826.
  17. Lee HJ, Kim E, Kim JS (2010) Targeted chromosomal deletions in human cells using zinc finger nucleases. Genome Res 20: 81-89.
  18. Kim S, Lee HJ, Kim E, Kim JS (2010) Analysis of targeted chromosomal deletions induced by zinc finger nucleases. Cold Spring HarbProtoc 2010: pdb.
  19. Lee HJ, Kweon J, Kim E, Kim S, Kim JS (2012) Targeted chromosomal duplications and inversions in the human genome using zinc finger nucleases. Genome Res 22: 539-548.
  20. Sun N, Yazawa M, Liu J, Han L, Sanchez-Freire V, et al. (2012) Patient-specific induced pluripotent stem cells as a model for familial dilated cardiomyopathy. SciTransl Med4:130.
  21. CarvajalVergara X, Sevilla A, D’Souza SL, Ang YS, Schaniel C, et al. (2010) Patient-specific induced pluripotent stem-cell-derived models of LEOPARD syndrome. Nature465: 808-812.
  22. Egashira T, Yuasa S, Fukuda K (2013) Novel insights into disease modeling using induced pluripotent stem cells. Biol Pharm Bull 36: 182-188.
  23. Panopoulos AD, Ruiz S, Izpisua Belmonte JC (2011) iPSCs: induced back to controversy. Cell Stem Cell 8: 347-348.
  24. Jiang Z, Han Y, Cao X (2014) Induced pluripotent stem cell (iPSCs) and their application in immunotherapy. Cell Mol Immunol 11: 17-24.
  25. Sommer CA, Mostoslavsky G (2013) The evolving field of induced pluripotency: recent progress and future challenges. J Cell Physiol 228: 267-275.
  26. Saha K, Jaenisch R (2009) Technical challenges in using human induced pluripotent stem cells to model disease. Cell Stem Cell 5: 584-595.
  27. Liras A, Segovia C, Gabán AS (2012) Advanced therapies for the treatment of hemophilia: future perspectives. Orphanet J Rare Dis 7: 97.
  28. Park CY, Kim J, Kweon J, Son JS, Lee JS, et al. (2014) Targeted inversion and reversion of the blood coagulation factor 8 gene in human iPS cells using TALENs. ProcNatlAcadSci U S A 111: 9253-9258.
  29. Kashiwakura Y, Ohmori T, Mimuro J, Madoiwa S, Inoue M, et al. (2014) Production of functional coagulation factor VIII from iPSCs using a lentiviral vector. Haemophilia 20: e40-44.
  30. Antonio Sorrentino (2010) Induced pluripotent stem cells: the long-expected revolution in medical science and practice?.The Journal of Nucleic Acids Investigation 1.
  31. Liu GH, Sancho-Martinez I, Izpisua Belmonte JC (2012) Cut and paste: restoring cellular function by gene correction. Cell Res 22: 283-284.
Citation: Joseph BC, Rao MS (2014) Induced Pluripotent Stem Cells for the Treatment of Hemophilia A. J Cell Sci Ther 5:e119.

Copyright: © 2014 Joseph BC, et al. 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