Submit manuscript...
Journal of
eISSN: 2373-6453

Human Virology & Retrovirology

Correspondence:

Received: January 01, 1970 | Published: ,

Citation: DOI:

Download PDF

Abstract

Discovery of induced Pluripotent stem cells (iPSCs) has revolutionized the fields of stem cell biology and regenerative medicine. iPSCs can now be generated through reprogramming of somatic cells from various tissues and mammalian species by ectopic expression of defined factors (Oct3/4, Sox2, c-Myc, Klf4). However, these methods of gene transduction, often involving viral vectors for delivery and genomic integration, may not be safe for clinical application sowing to high rates of malignant transformation and tumor genesis. In this article we provide a comprehensive overview of various new strategies including but not limited to mRNA or protein based delivery and/or chemical induction of pluripotency that circumvent the use of viral vectors for gene delivery. These methodological advancements increase the ease and efficiency of reprogramming, avoiding genomic modification, thereby increasing the suitability for clinical and translational applications in humans.

Introduction

The seminal work by Takahashi and Yamanaka in 2006 laid the foundation for what we today known as the induced Pluripotent stem-cell (iPSC) field.1,2 Through an elegant set of experiments they were the first to reveal that mature somatic cells can be forced to reprogram into an embryonic-like state, with a capacity for unrestrained growth. This reprogramming and turning back the clock required expression of four essential genes, namely, Oct4, Sox2, c-Myc and Klf4. Several studies have drawn and demonstrated a parallel between iPSCs and embryonic stem cells (ESCs) in terms of their Pluripotent potential and hence therapeutic value.3-9 In fact, iPSCs may actually be preferable to ESCs in some therapeutic context as they do not carry a risk of immune rejection due to their patient specific nature, and are not affected by the same ethical concerns as ESCs.10 But the methods of gene delivery to induce pluripotency while generating iPSCs have often come under scrutiny owing to the concerns on genomic alterations that some methods involve that renders them unsuitable for clinical applications in humans.11-35 While Yamanaka et al.1 used the genome integrating viral vectors for gene delivery, the field has since seen an explosion of gene delivery methods including non-viral vectors, RNAs, proteins as well as reprogramming using small molecules and/or chemical treatment, all of which circumvent issues of genomic integration.

Genetic and integrating methods of reprogramming

Pioneering work in the iPSCs field suggested the requirement for stable expression of reprogramming factors like Oct4, Sox2, c-Myc, Klf4, Lin28 to induce a state of pluripotency.36-46 This required genomic integration of these genes into the host genome using lentivirus or retrovirus vectors. While after the iPSC generation, the stable and high expression level of integrated exogenous genes gets down-regulated, yet, the possibility of tumorigenesis due to expression of oncogenes and inactivation of tumor suppressor genes cannot be ruled out. To circumvent the clinical drawbacks of genetic induction of reprogramming, owing to genomic integration, several methods have been reported for generating iPSCs cells through the use of non-integrating plasmid and adenovirus vectors (Table 1). However, in such cases, poor reprogramming efficiency remains to be a challenge. Therefore, there is an imminent need for the establishment of new methods of reprogramming that can bypass genomic integration without compromising efficiency of reprogramming, thereby facilitating the use of iPSCs for therapeutic applications.

Reprogramming Method

Species

Reference

 

Plasmid

 

Mouse and Human

 

Okita K et al. 2008

Adenovirus vector

Mouse and Human

Stadtfeld M et al. 2008

Excisable poly cistronic lentiviral vector

Human

Chang CW et al. 2009; Sommer C et al. 2009

OriP/EBNA1-based episomal vector

Human

Yu J et al. 2009

PiggyBac transposition

Mouse and human

Woltjen K et al. 2009; Yusa K et al. 2009

Recombinant Proteins

Mouse and human

Zhou H et al. 2009
Kim D et al. 2009

Sendai virus

mRNA and micro RNA

Human

Mouse and human

Fusaki N et al. 2009; Lieu PT et al. 2013
Judson RL et al. 2009; Mallanna SK et al. 2010; Warren L et al. 2010; Anokye-Danso F et al. 2011; Miyoshi N et al. 2011; Yoshioka N et al. 2013; Liao B et al. 2011; Subramanyam D et al. 2011

Magnetic Nano particles
Small Molecules

Sleeping beauty transposon

Mouse and human
Mouse

Mouse and human

Lee CH et al. 2011
Pasha Z et al. 2011; Hou PP et al. 2013
Davis RP et al. 2013

Table 1 Non-integrating methods of reprogramming to generate iPSCs

Non-integrating methods of reprogramming

To circumvent the clinical drawbacks of genetic induction of reprogramming, owing to genomic integration, several avenues of research have proposed alternate strategies that work with varied efficiency (Figure 1). A comprehensive review and comparison of the various alternatives available is presented below.

Figure 1 The Evolution of Reprogramming Technologies.

Viral vectors for generating Transgene-free iPSCs

The pioneering work for the derivation of iPSCs by Yamanaka and colleagues undertook integrative viral vectors for the delivery of reprogramming factors largely because of their obvious merits,

  1. Delivery to a wide range of cell types
  2. Stable and high levels of expression.

One equally obvious drawback of this methodology is the genomic integration of these exogenous gene sequences into the host genome that may lead to

  1. Mutations in endogenous genes due to random integration
  2. Inactivation of tumor suppressor genes
  3. Hyper activation of oncogenes leading to cancer and tumorigenesis.

To circumvent these clinical drawbacks, several technical advancements have been suggested through research conducted in different parts of the world.47,48 Soldner et al. used dox-inducible lentiviral vectors that involved the use of cre-excisable lentiviruses that upon integration can subsequently be excised from the host genome.49 The technology was later improvised by Sommer et al. generated a novel version of this single polycistronic vector containing a reporter fluorochrome to allow direct visualization of vector excision in live iPSCs in real-time.50

While this approach equals retroviral transduction in terms of efficiency but has the drawback of the need for additional genetic manipulations for the excision of cre-lox cassette. Another concern is the possibility of introducing chromosomal translocations during cre-mediated recombination. Lastly, a small piece of the vector is left in the integration site even after excision, which again leaves the possibility of insertional mutagenesis. To further improvise, Papapetrou et al. developed a strategy that includes additional steps for mapping the integration site in the genome to allow for the selection of clones with a single appropriate integration site for the residual LTR, however there still remains a risk that insertional mutagenesis may arise from this genetically integrated exogenous DNA.51-53

To circumvent the integration step altogether, several groups have resorted to non-integrating viruses such as adenoviruses for reprogramming factor delivery. However, adenoviral methods are highly inefficient and limited to permissive cell types. Stadtfeld et al. reported mouse iPSCs from fibroblasts and liver cells at extremely low efficiency (0.0001% to 0.001%), which is significantly lower than that obtained from integrating viruses (0.01% to 0.1%).18,54 Their low efficiency has been largely attributed to the failure to sustain factor expression due to a gradual loss of viral vectors in dividing cells. Sendai virus (SeV), is another type of non-integrating virus vector which has been successfully used to reprogram human cells.55 SeV viruses are RNA viruses and hence do not integrate into host genome unlike other DNA viruses.56-58 Additionally, SeV viruses are replication competent and hence unlike the adenoviruses can sustain high-levels of factor expression, which can reach up to 10 times higher (efficiency ~1%) than those obtained by conventional retroviral vectors. An additional benefit of this system is the convenient removal of viruses by antibody mediated negative selection utilizing the cell surface marker, HN, expressed on SeV infected cells. 

Plasmid DNA vectors for generating Transgene-free iPSCs 

Several groups have successfully generated non-viral, transgene-free iPSCs using DNA-based expression vectors such as plasmid, mini circle vector, piggyBac transposon and episomal plasmid. But this transgene-free factor delivery comes at a price of low efficiency as transient transfection of DNA vectors is less efficient than viral transduction.59 To this end, several groups have reportedly used polycistronic expression cassettes to ensure that every cell gets the full complement of factors.17,60 Alternately, mini circle vectors expressing Oct4, Sox2, Nanog and Lin28 from a single cassette of OSNL plus a GFP reporter gene, each separated by self-cleaving peptide 2A sequences, have been used to generate transgene-free iPSCs from human adipose stem cells.61 These vectors used circular expression cassettes produced from regular plasmids by removing the bacterial backbone through intra molecular recombination. Compared to their parental plasmids, mini circles are smaller in size and less prone to silencing, both of which make them highly sought after for reprogramming somatic cells, however, they are far less efficient compared to retroviral vectors (reprogramming efficiency ~0.005%).62

Moreover, their usage requires cumbersome protocols with large number of starting cells and repeated transient transfections to compensate for the rapid loss of reprogramming factors in dividing cells. But compared to other methods of reprogramming mini circle approach has several advantages, including the use of a single vector without the need for subsequent drug selection, vector excision, or the inclusion of oncogenes such as SV40, making the derivation process free of foreign transgenes or chemicals. These unique features bagged it the FDA approval, making it a potentially significant approach from clinical and translational standpoint.

Some other groups designed a piggyBac transposon approach to deliver reprogramming factors to host cells.63-67 In this approach, a transient transposase expression leads to stable integration of the piggyBac transposon cassette into the host genome, thus ensuring a high level of expression of reprogramming factors. Interestingly, the piggyBac transposon cassette can be easily excised, taking advantage of the natural propensity of the system for seamless excision, by transiently expressing the transposase gene in iPSCs without any risk of mutations at the integration site.68 However, the excision step is inefficient and in certain cases reversion of iPSCs to wild type phenotypes has been observed after piggyBac transposon excision.

An alternate strategy for stable expression of reprogramming factors is made available by the use of non-integrating oriP/EBNA1-based episomal vectors.69,70 Derived from the Epstein-Barr virus, oriP vectors can be easily transfected into human somatic cells, without the need for viral packaging, and can be subsequently removed from cells in the absence of drug selection without further genetic manipulation. In such vectors the cis element oriP and the EBNA1 gene product act in Trans and together control the replication and partition of the episomes during cell cycle. This process provides these vectors with self-sufficiency to function in a variety of cell types, eliminating the need for repeated transfections. Additionally, the removal of episomal transgenes is not only automatic and efficient, it required repeat application and also error proof, and thus making oriP episomal vectors a method of choice. However, the stable transfection efficiency of the oriP vectors is several orders of magnitude less than that of the lentiviral vectors.

DNA-free methods for generating Transgene-free iPSCs 

To overcome the caveat of genome modification presented by DNA-based vectors, several groups have successfully attempted to deliver reprogramming factors as mRNAs or proteins. In such cases, instead of being produced by a transgene, recombinant proteins are directly delivered to cells by fusing them to cell-penetrating peptides such as poly-arginine.71-73 Although such proteins make it to the nucleus successfully, but require repeated application for a successful reprogramming. Moreover, all reports of protein-based reprogramming suffer from low efficiencies (~0.001%). Additionally, it is technically challenging to produce large quantities of biochemically active recombinant proteins.

Worth highlighting also is the discrepancy in the various published reports. Compared to Zhou et al.28 who show incapability in generating mouse iPSCs with only recombinant proteins, without the use of chemical supplements, Kim et al.29 demonstrated successful derivation of human iPSCs with direct delivery of reprogramming proteins in the absence of any chemical treatment. The plausible scientific explanation for these observed differences could be the mode of expression of reprogramming proteins, which involved mammalian cells in case of Kim et al. while expression in E.coli followed by protein refolding for Zhou et al.28 Taken together, direct protein based reprogramming approach still remains a cumbersome technique offering poor efficiencies compared to other known approaches, thereby lacking the competitive edge to be of value in clinical setting.

Schematic representation of the contrast in the efficiency and safety of reprogramming between various technologies reported till date. (Illustration adapted from Cardiovasc. Trans. Res. 2013, 6, 956-68) As an alternative, synthetic mRNAs have shown promise for fast and efficient reprogramming.74-78 Warren et al.74 proposed a non-integrating strategy for cellular reprogramming by delivery of synthetic mRNA, modified to overcome innate anti-viral responses.74,79,80 A five-factor cocktail (OKSML) was able to reprogram human fibroblasts with an efficiency of 2%, which is two orders of magnitude higher than virus-based reprogramming. Additionally, microRNAs (miRNAs) have shown great potential in regulating pluripotency.81-85 Intriguingly, five miRNAs (miR-302a-d and miR-367) have been shown to be sufficient in reprogramming human cells without any exogenous transcription factor.86,87 Later, another group reported Tg-free iPSCs in mouse and human systems by transfections of mature miRNAs (different combinations of miR-200c, 302s and 369s).88 While their ease of production and small size ensuring efficient delivery makes mRNAs and miRNAs an attractive option for the production of transgene-free iPSCs but poor efficiency still remains a stumbling block.

The quest for reprogramming somatic cells solely with the use of chemical compounds ended when Hou et al.89 reported the derivation of mouse iPSCs at a frequency as high as ~0.2% using a combination of seven small-molecule compounds.5,19,21-23,28,42,90-96 This discovery brought a paradigm shift to the technology of cellular reprogramming. Additionally, the use of small molecules alone brought with it several advantages like the molecules being cell permeable showing a promise of easy delivery, easily synthesized in large quantities and non-immunogenic. But while the ease of use and efficiency of these small molecules in mouse system has been well documented, their success in human cells is yet to be established.

Conclusion

Many roadblocks remain to be addressed before iPSCs reach the clinic.97-109 A major impediment to the use of iPSCs for therapeutic purposes remains the viral-based delivery of reprogramming factors. Additionally, for patient-specific autologous treatment, methods need to be improved able to generate iPSCs in sufficient quantity. Efficiency of reprogramming remains the biggest challenge, especially in the context of technologies that do not integrate transgenes into the host genome. Moreover, there still remains a risk of teratoma formation in case a subpopulation of iPSCs fails to terminally differentiate prior to transplantation.

Even upon differentiation it is difficult to control and predict events of spontaneous differentiation and dedifferentiation into other cell types and the clinical outcome of such a heterologous cell populations within a certain tissue. Current scientific and technical advancements are rapidly eliminating some of these fundamental issues in the iPSC field thereby reassuring the profound benefits of stem cell technology to regenerative medicine. But while a lot of ground has been covered, there might be a long way to go ahead before the full potential of iPSC technology may be harnessed.

Acknowledgments

We thank Mr. Daksh Khulbe for his constructive critiques and apologize to all colleagues whose work could not be cited due to space constraints. We are grateful to CSIR and JDRF for their generous support.

Conflicts of interest

None.

References

  1. 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.
  2. 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.
  3. Wernig M, Meissner A, Foreman R, et al. In vitro reprogramming of fibroblasts into a pluripotent ES‒cell‒like state. Nature. 2007;448(7151):318‒324.
  4. 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.
  5. Huangfu D, Maehr R, Guo W, et al. Induction of pluripotent stem cells by defined factors is greatly improved by small‒molecule compounds. Nature biotechnology. 2008;26(7):795‒797.
  6. Maherali N, Ahfeldt T, Rigamonti A, et al. A high‒efficiency system for the generation and study of human induced pluripotent stem cells. Cell Stem Cell. 2008;3(3):340‒345.
  7. Maherali N, Hochedlinger K. Guidelines and techniques for the generation of induced pluripotent stem cells. Cell Stem Cell. 2008;3(6):595‒605.
  8. Park IH, Zhao R, West JA, et al. Reprogramming of human somatic cells to pluripotency with defined factors. Nature. 2008;451(7175):141‒146.
  9. Zhao Y, Yin X, Qin H, et al. Two supporting factors greatly improve the efficiency of human iPSC generation. Cell Stem Cell. 2008;3(5):475‒479.
  10. Thomson JA, Itskovitz‒Eldor J, Shapiro SS, et al. Embryonic Stem Cell lines derived from human blastocysts. Science. 1998;282(5391):1145‒1147.
  11. Blelloch R, Venere M, Yen J, et al. Generation of induced pluripotent stem cells in the absence of drug selection. Cell Stem Cell. 2007;1(3):245‒247.
  12. Okita K, Ichisaka T, Yamanaka S. Generation of germline‒competent induced pluripotent stem cells. Nature. 2007;448(7151):313‒317.
  13. Hockemeyer D, Soldner F, Cook EG, et al. A drug‒inducible system for direct reprogramming of human somatic cells to pluripotency. Cell Stem Cell. 2008;3(3):346‒353.
  14. Huangfu D, Osafune K, Maehr R, et al. Induction of pluripotent stem cells from primary human fibroblasts with only Oct4 and Sox2. Nature Biotechnology. 2008;26(11):1269‒1275.
  15. Kim JB, Zaehres H, Wu G, et al. Pluripotent stem cells induced from adult neural stem cells by reprogramming with two factors. Nature. 2008;454(7204):646‒650.
  16. Nakagawa M, Koyanagi M, Zanabe K, et al. Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts. Nature Biotechnology. 2008;26(1):101‒106.
  17. Okita K, Nakagawa M, Hyenjong H, et al. Generation of mouse induced pluripotent stem cells without viral vectors. Science. 2008;322(5903):949‒953.
  18. Stadtfeld M, Nagaya M, Utikal J, et al. Induced pluripotent stem cells generated without viral integration. Science. 2008;322(5903):945‒949.
  19. Feng B, Ng JH, Heng JC, et al. Molecules that promote or enhance reprogramming of somatic cells to induced pluripotent stem cells. Cell Stem Cell. 2009;4(4):301‒312.
  20. Ichida JK, Blanchard J, Lam K, et al. A small‒molecule inhibitor of tgf‒Beta signaling replaces sox2 in reprogramming by inducing nanog. Cell Stem Cell. 2009;5(5):491‒503.
  21. Li W, Wei W, Zhu S, et al. Generation of rat and human induced pluripotent stem cells by combining genetic reprogramming and chemical inhibitors. Cell Stem Cell. 2009;4(1):16‒19.
  22. Lin T, Ambasudhan R, Yuan X, et al. A chemical platform for improved induction of human iPSCs. Nature Methods. 2009;6(11):805‒808.
  23. Lyssiotis CA, Foreman RK, Staerk J, et al. Reprogramming of murine fibroblasts to induced pluripotent stem cells with chemical complementation of Klf4. Proc Natl Acad Sci U S A. 2009;106(22):8912‒8917.
  24. Miura K, Okada Y, Aoi T, et al. Variation in the safety of induced pluripotent Stem Cell lines. Nature Biotechnology. 2009;27(8):743‒745.
  25. Ieda M, Fu JD, Delgado‒Olguin P, et al. Direct reprogramming of fibroblasts into functional cardiomyocytes by defined factors. Cell. 2010;142(3):375‒386.
  26. 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.
  27. Rodriguez‒Piza I, Richaud‒Patin Y, Vassena R, et al. Reprogramming of human fibroblasts to induced pluripotent stem cells under xeno‒free conditions. Stem Cells. 2010;28(1):36‒44.
  28. Zhu S, Li W, Zhou H, et al. Reprogramming of human primary somatic cells by OCT4 and chemical compounds. Cell Stem Cell. 2010;7(6):651‒655.
  29. Lee CH, Kim JH, Lee HJ, et al. The generation of iPS cells using non‒viral magnetic nanoparticle based transfection. Biomaterials. 2011;32(28):6683‒6691.
  30. Awe JP, Lee PC, Ramathal C, et al. Generation and characterization of transgene‒free human induced pluripotent stem cells and conversion to putative clinical‒grade status. Stem Cell Research & Therapy. 2013;4(4):87.
  31. Chen W, Tsai PH, Hung Y, et al. Nonviral cell labeling and differentiation agent for induced pluripotent stem cells based on mesoporous silica nanoparticles. ACS Nano. 2013;7(10):8423‒8440.
  32. Habib O, Habib G, Choi HW, et al. An improved method for the derivation of high quality iPSCs in the absence of c‒Myc. Experimental Cell Research. 2013;319(20):3190‒3200.
  33. Kishino Y, Seki T, Fujita J, et al. Derivation of transgene‒free human induced pluripotent stem cells from human peripheral T cells in defined culture conditions. PloS one. 2014;9(5):e97397.
  34. Lu HF, Chai C, Lim TC, et al. A defined xeno‒free and feeder‒free culture system for the derivation, expansion and direct differentiation of transgene‒free patient‒specific induced pluripotent stem cells. Biomaterials. 2014;35(9):2816‒2826.
  35. Raab S, Klingenstein M, Liebau S, et al. A Comparative View on Human Somatic Cell Sources for iPSC Generation. Stem Cells Int. 2014:768391.
  36. Hochedlinger K, Jaenisch R. Nuclear reprogramming and pluripotency. Nature. 2006;441(7097):1061‒1067.
  37. Eminli S, Utikal J, Arnold K, et al. Reprogramming of neural progenitor cells into induced pluripotent stem cells in the absence of exogenous Sox2 expression. Stem Cells. 2008;26(10):2467‒2474.
  38. Marson A, Foreman R, Chevalier B, et al. Wnt signaling promotes reprogramming of somatic cells to pluripotency. Cell Stem Cell. 2008;3(2):132‒135.
  39. Stadtfeld M, Maherali N, Breault NT, et al. Defining molecular cornerstones during fibroblast to iPS cell reprogramming in mouse. Cell Stem Cell. 2008;2(3):230‒240.
  40. Daley GQ, Lensch MW, Jaenisch R, et al. Broader implications of defining standards for the pluripotency of iPSCs. Cell Stem Cell. 2009;4(3):200‒201;author reply 202.
  41. Hanna J, Saha K, Pando B, et al. Direct cell reprogramming is a stochastic process amenable to acceleration. Nature. 2009;462(7273):595‒601.
  42. 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.
  43. Boue S, Paramonov I, Barrero MJ, et al. Analysis of human and mouse reprogramming of somatic cells to induced pluripotent stem cells. What is in the plate? PloS One. 2010;5(9):e12664.
  44. Malik N, Rao MS. A review of the methods for human iPSC derivation. Methods in Molecular Biology. 2013;997:23‒33.
  45. Graversen VK, Chavala SH. Induced Pluripotent Stem Cells: Generation, Characterization, and Differentiation‒Methods and Protocols. Methods mol biol. 2014
  46. Liu K, Song Y, Yu H, et al. Understanding the roadmaps to induced pluripotency. Cell Death & Disease. 2014;5:e1232.
  47. Carey BW, Markoulaki S, Hanna J, et al. Reprogramming of murine and human somatic cells using a single polycistronic vector. Proc Natl Acad Sci U S A. 2009;106(1):157‒162.
  48. Chang CW, Lai YS, Pawlik KM, et al. Polycistronic lentiviral vector for "hit and run" reprogramming of adult skin fibroblasts to induced pluripotent stem cells. Stem cells. 2009;27(5):1042‒1049.
  49. Soldner F, Hockemeyer D, Beard C, et al. Parkinson's disease patient‒derived induced pluripotent stem cells free of viral reprogramming factors. Cell. 2009;136(5):964‒977.
  50. Sommer CA, Stadtfeld M, Murphy GJ, et al. Induced pluripotent Stem Cell generation using a single lentiviral Stem Cell cassette. Stem cells. 2009;27(3):543‒549.
  51. Papapetrou EP, Sadelain M. Generation of transgene‒free human induced pluripotent stem cells with an excisable single polycistronic vector. Nat Protoc. 2011;6(9):1251‒1273.
  52. Papapetrou EP, Sadelain M. Derivation of genetically modified human pluripotent stem cells with integrated transgenes at unique mapped genomic sites. Nat Protoc. 2011;6(9):1274‒1289.
  53. Kuehle J, Turan S, Cantz T, et al. Modified lentiviral LTRs allow Flp recombinase‒mediated cassette exchange and in vivo tracing of "factor‒free" induced pluripotent stem cells. Mol ther. 2014;22(5):919‒928.
  54. Zhou W, Freed CR. Adenoviral gene delivery can reprogram human fibroblasts to induced pluripotent stem cells. Stem Cells. 2009;27(11):2667‒2674.
  55. Fusaki N, Ban H, Nishiyama A, et al. Efficient induction of transgene‒free human pluripotent stem cells using a vector based on Sendai virus, an RNA virus that does not integrate into the host genome. Proc Jpn Acad Ser B Phys Biol Sci. 2009;85(8):348‒362.
  56. Li HO, Zhu YF, Asakawa M, et al. A cytoplasmic RNA vector derived from nontransmissible Sendai virus with efficient gene transfer and expression. J Virol. 2000;74(14):6564‒6569.
  57. Lieu PT, Fontes A, Vemuri MC, et al. Generation of induced pluripotent stem cells with CytoTune, a non‒integrating Sendai virus. Methods Mol Biol. 2013;997:45‒56.
  58. Fujie Y, Fusaki N, Katayama T, et al. New type of Sendai virus vector provides transgene‒free iPS cells derived from chimpanzee blood. PloS One. 2014;9(12):e113052.
  59. Okita K, Hong H, Takahashi K, et al. Generation of mouse‒induced pluripotent stem cells with plasmid vectors. Nature Protocols. 2010;5(3):418‒428.
  60. Gonzalez F, Barragan Monasterio M, Tiscornia G, et al. Generation of mouse‒induced pluripotent stem cells by transient expression of a single nonviral polycistronic vector. Proc Natl Acad Sci U S A. 2009;106(22):8918‒8922.
  61. Jia F, Wilson KD, Sun N, et al. A nonviral minicircle vector for deriving human iPS cells. Nature Methods. 2010;7(3):197‒199.
  62. Chen ZY, He CY, Kay MA. Improved production and purification of minicircle DNA vector free of plasmid bacterial sequences and capable of persistent transgene expression in vivo. Hum Gene Ther. 2005;16(1):126‒131.
  63. Yusa K, Rad R, Takeda J, et al. Generation of transgene‒free induced pluripotent mouse stem cells by the piggyBac transposon. Nat Methods. 2009;6(5):363‒369.
  64. Woltjen K, Michael IP, Mohseni P, et al. piggyBac transposition reprograms fibroblasts to induced pluripotent stem cells. Nature. 2009;458(7239):766‒770.
  65. Kaji K, Norrby K, Paca A, et al. Virus‒free induction of pluripotency and subsequent excision of reprogramming factors. Nature. 2009;458(7239):771‒775.
  66. Wilson MH, Coates CJ, George AL. PiggyBac transposon‒mediated gene transfer in human cells. Mol ther. 2007;15(1):139‒145.
  67. Davis RP, Nemes C, Varga E, et al. Generation of induced pluripotent stem cells from human foetal fibroblasts using the Sleeping Beauty transposon gene delivery system. Differentiation; Research In Biological Diversity. 2013;86(1‒2):30‒37.
  68. Lacoste A, Berenshteyn F, Brivanlou AH. An efficient and reversible transposable system for gene delivery and lineage‒specific differentiation in human embryonic stem cells. Cell Stem Cell. 2009;5(3):332‒342.
  69. Yu J, Hu K, Smuga‒Otto K, et al. Human induced pluripotent stem cells free of vector and transgene sequences. Science. 2009;324(5298):797‒801.
  70. Fontes A, Macarthur CC, Lieu PT, et al. Generation of human‒induced pluripotent stem cells (hiPSCs) using episomal vectors on defined Essential 8 Medium conditions. Methods Mol Biol. 2013;997:57‒72.
  71. Zhou H, Wu S, Joo JY, et al. Generation of induced pluripotent stem cells using recombinant proteins. Cell Stem Cell. 2009;4(5):381‒384.
  72. Kim D, Kim CH, Moon JI, et al. Generation of human induced pluripotent stem cells by direct delivery of reprogramming proteins. Cell Stem. 2009;Cell 4(6):472‒476.
  73. Cho HJ, Lee CS, Kwon, et al. Induction of pluripotent stem cells from adult somatic cells by protein‒based reprogramming without genetic manipulation. Blood. 2010;116(3):386‒395.
  74. Warren L, Manos PD, Ahfeldt T, et al. Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Cell. 2010;7(5):618‒630.
  75. Li M, Sancho‒Martinez I, Izpisua Belmonte JC. Cell fate conversion by mRNA. Stem Cell Res Ther. 2011;2(1):5.
  76. Li Z, Yang CS, Nakashima K, et al. Small RNA‒mediated regulation of iPS cell generation. EMBO J. 2011;30(5):823‒834.
  77. Bernal JA. RNA‒based tools for nuclear reprogramming and lineage‒conversion: towards clinical applications. J Cardiovasc Trans Res. 2013;6(6):956‒968.
  78. Yoshioka N, Gros E, Li HR, et al. Efficient generation of human iPSCs by a synthetic self‒replicative RNA. Cell Stem Cell. 2013;13(2):246‒254.
  79. Warren L, Ni Y, Wang J, et al. Feeder‒free derivation of human induced pluripotent stem cells with messenger RNA. Sci Rep. 2012;2:657.
  80. Mandal PK, Rossi DJ. Reprogramming human fibroblasts to pluripotency using modified mRNA. Nat Protoc. 2013;8(3):568‒582.
  81. Judson RL, Babiarz JE, Venere M, et al. Embryonic stem cell‒specific microRNAs promote induced pluripotency. Nature Biotechnology. 2009;27(5):459‒461.
  82. Mallanna SK, Rizzino A. Emerging roles of microRNAs in the control of embryonic stem cells and the generation of induced pluripotent stem cells. Dev Biol. 2010;344(1):16‒25.
  83. Subramanyam D, Lamouille S, Judson RL, et al. Multiple targets of miR‒302 and miR‒372 promote reprogramming of human fibroblasts to induced pluripotent stem cells. Nat Biotechnol. 2011;29(5):443‒448.
  84. Wang T, Shi SB, Sha HY. MicroRNAs in regulation of pluripotency and somatic cell reprogramming: small molecule with big impact. RNA Biol. 2013;10(8):1255‒1261.
  85. Wang T, Warren ST, Jin P. Toward pluripotency by reprogramming: mechanisms and application. Protein Cell. 2013;4(11):820‒832.
  86. Anokye‒Danso F, Trivedi CM, Juhr D, et al. Highly efficient miRNA‒mediated reprogramming of mouse and human somatic cells to pluripotency. Cell Stem Cell. 2011;8(4):376‒388.
  87. Liao B, Bao X, Liu L, et al. MicroRNA cluster 302‒367 enhances somatic cell reprogramming by accelerating a mesenchymal‒to‒epithelial transition. J Biol Chem. 2011;286(19):17359‒17364.
  88. Miyoshi N, Ishii H, Nagano H, et al. Reprogramming of mouse and human cells to pluripotency using mature microRNAs. Cell Stem Cell. 2011;8(8):633‒638.
  89. Hou P, Li Y, Zhang X, et al. Pluripotent stem cells induced from mouse somatic cells by small‒molecule compounds. Science. 2013;341(6146):651‒654.
  90. Desponts C, Ding S. Using small molecules to improve generation of induced pluripotent stem cells from somatic cells. Methods in Molecular Biology. 2010;636:207‒218.
  91. Esteban MA, Wang T, Qin B, et al. Vitamin C enhances the generation of mouse and human induced pluripotent stem cells. Cell Stem Cell. 2010;6(1):71‒79.
  92. Pasha Z, Haider H, Ashraf M. Efficient non‒viral reprogramming of myoblasts to stemness with a single small molecule to generate cardiac progenitor cells. PloS one. 2011;6(8):e23667.
  93. Li Z, Rana TM. A kinase inhibitor screen identifies small‒molecule enhancers of reprogramming and iPS cell generation. Nat Commun. 2012;3:1085.
  94. Trokovic R, Weltner J, Manninen T, et al. Small molecule inhibitors promote efficient generation of induced pluripotent stem cells from human skeletal myoblasts. Stem Cells Dev. 2013;22(1):114‒123.
  95. Zhang R, Zhang LH, Xie X. iPSCs and small molecules: a reciprocal effort towards better approaches for drug discovery. Acta Pharmacol Sin. 2013;34(6):765‒776.
  96. Wei X, Chen Y, Xu Y, et al. Small molecule compound induces chromatin de‒condensation and facilitates induced pluripotent Stem Cell generation. J Mol Cell Biol. 2014;6(5):409‒420.
  97. Lu X, Zhao T. Clinical therapy using iPSCs: hopes and challenges. Genomics Proteomics Bioinformatics. 2013;11(5):294‒298.
  98. Rao M. iPSC‒Based cell therapy: an important step forward. Stem Cell Reports. 2013;1(4):281‒282.
  99. Rao M. iPSC crowdsourcing: a model for obtaining large panels of Stem Cell lines for screening. Cell Stem Cell. 2013;13(4):389‒391.
  100. Shtrichman R, Germanguz I, Itskovitz‒Eldor J. Induced pluripotent stem cells (iPSCs) derived from different cell sources and their potential for regenerative and personalized medicine. Curr Mol Med. 2013;13(5):792‒805.
  101. Wu M, Chen G, Hu B. Induced pluripotency for translational research. Genomics Proteomics Bioinformatics. 2013;11(5):288‒293.
  102. Abdelalim EM, Bonnefond A, Bennaceur‒Griscelli A, et al. Pluripotent stem cells as a potential tool for disease modelling and cell therapy in diabetes. Stem Cell rev. 2014;10(3):327‒337.
  103. Csobonyeiova M, Polak S, Koller J, et al. Induced pluripotent stem cells and their implication for regenerative medicine. Cell Tissue Bank. 2014.
  104. Inoue H, Nagata N, Kurokawa H, et al. iPS cells: a game changer for future medicine. EMBO J. 2014;33(5):409‒417.
  105. Kim C. iPSC technology‒ Powerful hand for disease modeling and therapeutic screen. BMB Rep. 2014.
  106. Ko HC, Gelb BD. Concise review: drug discovery in the age of the induced pluripotent stem cell. Stem Cells Transl Med. 2014;3(4):500‒509.
  107. Sivapatham R, Zeng X. Generation and Characterization of Patient‒Specific Induced Pluripotent Stem Cell for Disease Modeling. Methods Mol Biol. 2014.
  108. Walmsley GG, Hyun J, McArdle A, et al. Induced pluripotent stem cells in regenerative medicine and disease modeling. Curr Stem Cell Res Ther. 2014;9(2):73‒81.
  109. Santostefano KE, Hamazaki T, Biel NM, et al. A practical guide to induced pluripotent Stem Cell research using patient samples. Lab Invest. 2015;95(1):4‒13.
Creative Commons Attribution License

© . This is an open access article distributed under the terms of the, which permits unrestricted use, distribution, and build upon your work non-commercially.