Journal List > J Rheum Dis > v.20(5) > 1064063

Jung and Ju: Application of Induced Pluripotent Stem Cells in Rheumatology

Abstract

Since induced pluripotent stem cell (iPSC) was first in-troduced by Yamanaka in 2006, it took only six years to win a Nobel Prize for his pioneering work. It is unusual to win a Nobel Prize for such recent research with a short history. Many scientists and clinicians are interested in iPSC for its potential application. Significant progression in this field has been made, while there remain many hurdles to overcome for application of iPSC technique in real clinics. In this review, the concept of reprogramming and the basic techniques of iPSC generation will be discussed for the reader's convenience, followed by discussion of recent progress, followed by the topics of “disease modeling” and “cell therapy” with iPSC in the second half of this article. Several examples of rheumatologic application of iPSC will be provided in the main text. If rheumatologists could understand the merits and potentials of iPSC, opportunities for innovative research and therapy can be expanded.

References

1. Somorjai IM, Lohmann JU, Holstein TW, Zhao Z. Stem cells: a view from the roots. Biotechnol J. 2012; 7:704–22.
crossref
2. Schugar RC, Robbins PD, Deasy BM. Small molecules in stem cell self-renewal and differentiation. Gene Ther. 2008; 15:126–35.
crossref
3. Grskovic M, Javaherian A, Strulovici B, Daley GQ. Induced pluripotent stem cells–opportunities for disease modelling and drug discovery. Nat Rev Drug Discov. 2011; 10:915–29.
4. Murry CE, Keller G. Differentiation of embryonic stem cells to clinically relevant populations: lessons from embryonic development. Cell. 2008; 132:661–80.
5. Ambasudhan R, Talantova M, Coleman R, Yuan X, Zhu S, Lipton SA, et al. Direct reprogramming of adult human fibroblasts to functional neurons under defined conditions. Cell Stem Cell. 2011; 9:113–8.
crossref
6. Zaehres H, Schöler HR. Induction of pluripotency: from mouse to human. Cell. 2007; 131:834–5.
crossref
7. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007; 131:861–72.
crossref
8. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006; 126:663–76.
crossref
9. Lowry WE, Richter L, Yachechko R, Pyle AD, Tchieu J, Sridharan R, et al. Generation of human induced pluripotent stem cells from dermal fibroblasts. Proc Natl Acad Sci U S A. 2008; 105:2883–8.
crossref
10. Stadtfeld M, Hochedlinger K. Induced pluripotency: history, mechanisms, and applications. Genes Dev. 2010; 24:2239–63.
crossref
11. Maherali N, Sridharan R, Xie W, Utikal J, Eminli S, Arnold K, et al. Directly reprogrammed fibroblasts show global epigenetic remodeling and widespread tissue contribution. Cell Stem Cell. 2007; 1:55–70.
12. Wilmut I, Schnieke AE, McWhir J, Kind AJ, Campbell KH. Viable offspring derived from fetal and adult mam-malian cells. Nature. 1997; 385:810–3.
crossref
13. Wakayama T, Hayashi Y, Ogura A. Participation of the female pronucleus derived from the second polar body in full embryonic development of mice. J Reprod Fertil. 1997; 110:263–6.
crossref
14. Freberg CT, Dahl JA, Timoskainen S, Collas P. Epigenetic reprogramming of OCT4 and NANOG regulatory regions by embryonal carcinoma cell extract. Mol Biol Cell. 2007; 18:1543–53.
15. Taranger CK, Noer A, S⊘rensen AL, Håkelien AM, Boquest AC, Collas P. Induction of dedifferentiation, ge-nomewide transcriptional programming, and epigenetic reprogramming by extracts of carcinoma and embryonic stem cells. Mol Biol Cell. 2005; 16:5719–35.
crossref
16. Tada M, Morizane A, Kimura H, Kawasaki H, Ainscough JF, Sasai Y, et al. Pluripotency of reprogrammed somatic genomes in embryonic stem hybrid cells. Dev Dyn. 2003; 227:504–10.
crossref
17. Yu J, Vodyanik MA, He P, Slukvin II, Thomson JA. Human embryonic stem cells reprogram myeloid precursors following cell-cell fusion. Stem Cells. 2006; 24:168–76.
crossref
18. Okita K, Ichisaka T, Yamanaka S. Generation of germ-line-competent induced pluripotent stem cells. Nature. 2007; 448:313–7.
crossref
19. Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007; 318:1917–20.
crossref
20. Lenardo MJ, Staudt L, Robbins P, Kuang A, Mulligan RC, Baltimore D. Repression of the IgH enhancer in ter-atocarcinoma cells associated with a novel octamer factor. Science. 1989; 243:544–6.
crossref
21. Okamoto K, Okazawa H, Okuda A, Sakai M, Muramatsu M, Hamada H. A novel octamer binding transcription factor is differentially expressed in mouse embryonic cells. Cell. 1990; 60:461–72.
crossref
22. Niwa H, Miyazaki J, Smith AG. Quantitative expression of Oct-3/4 defines differentiation, dedifferentiation or self-renewal of ES cells. Nat Genet. 2000; 24:372–6.
crossref
23. Wu da Y, Yao Z. Isolation and characterization of the murine Nanog gene promoter. Cell Res. 2005; 15:317–24.
crossref
24. Yuan H, Corbi N, Basilico C, Dailey L. Developmen-tal-specific activity of the FGF-4 enhancer requires the synergistic action of Sox2 and Oct-3. Genes Dev. 1995; 9:2635–45.
crossref
25. Tomioka M, Nishimoto M, Miyagi S, Katayanagi T, Fukui N, Niwa H, et al. Identification of Sox-2 regulatory region which is under the control of Oct-3/4-Sox-2 complex. Nucleic Acids Res. 2002; 30:3202–13.
crossref
26. Avilion AA, Nicolis SK, Pevny LH, Perez L, Vivian N, Lovell-Badge R. Multipotent cell lineages in early mouse development depend on SOX2 function. Genes Dev. 2003; 17:126–40.
crossref
27. Ivanova N, Dobrin R, Lu R, Kotenko I, Levorse J, DeCoste C, et al. Dissecting self-renewal in stem cells with RNA interference. Nature. 2006; 442:533–8.
crossref
28. Seoane J, Le HV, Massagué J. Myc suppression of the p21(Cip1) Cdk inhibitor influences the outcome of the p53 response to DNA damage. Nature. 2002; 419:729–34.
crossref
29. Lewitzky M, Yamanaka S. Reprogramming somatic cells towards pluripotency by defined factors. Curr Opin Biotechnol. 2007; 18:467–73.
crossref
30. Li Y, McClintick J, Zhong L, Edenberg HJ, Yoder MC, Chan RJ. Murine embryonic stem cell differentiation is promoted by SOCS-3 and inhibited by the zinc finger transcription factor Klf4. Blood. 2005; 105:635–7.
crossref
31. Nakagawa M, Koyanagi M, Tanabe K, Takahashi K, Ichisaka T, Aoi T, et al. Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts. Nat Biotechnol. 2008; 26:101–6.
crossref
32. Kim JB, Sebastiano V, Wu G, Araúzo-Bravo MJ, Sasse P, Gentile L, et al. Oct4-induced pluripotency in adult neural stem cells. Cell. 2009; 136:411–9.
crossref
33. Si-Tayeb K, Noto FK, Sepac A, Sedlic F, Bosnjak ZJ, Lough JW, et al. Generation of human induced pluripotent stem cells by simple transient transfection of plasmid DNA encoding reprogramming factors. BMC Dev Biol. 2010; 10:81.
crossref
34. Anokye-Danso F, Trivedi CM, Juhr D, Gupta M, Cui Z, Tian Y, et al. Highly efficient miRNA-mediated reprogramming of mouse and human somatic cells to pluripotency. Cell Stem Cell. 2011; 8:376–88.
crossref
35. Kim D, Kim CH, Moon JI, Chung YG, Chang MY, Han BS, et al. Generation of human induced pluripotent stem cells by direct delivery of reprogramming proteins. Cell Stem Cell. 2009; 4:472–6.
crossref
36. Park IH, Arora N, Huo H, Maherali N, Ahfeldt T, Shimamura A, et al. Disease-specific induced pluripotent stem cells. Cell. 2008; 134:877–86.
crossref
37. Tiscornia G, Vivas EL, Izpisúa Belmonte JC. Diseases in a dish: modeling human genetic disorders using induced pluripotent cells. Nat Med. 2011; 17:1570–6.
crossref
38. Moser HW. Adrenoleukodystrophy: phenotype, genetics, pathogenesis and therapy. Brain. 1997; 120:1485–508.
crossref
39. Urbach A, Bar-Nur O, Daley GQ, Benvenisty N. Differential modeling of fragile X syndrome by human embryonic stem cells and induced pluripotent stem cells. Cell Stem Cell. 2010; 6:407–11.
crossref
40. Saha K, Jaenisch R. Technical challenges in using human induced pluripotent stem cells to model disease. Cell Stem Cell. 2009; 5:584–95.
crossref
41. Lee G, Papapetrou EP, Kim H, Chambers SM, Tomishima MJ, Fasano CA, et al. Modelling pathogenesis and treatment of familial dysautonomia using patient-specific iPSCs. Nature. 2009; 461:402–6.
crossref
42. Soldner F, Hockemeyer D, Beard C, Gao Q, Bell GW, Cook EG, et al. Parkinson's disease patient-derived induced pluripotent stem cells free of viral reprogramming factors. Cell. 2009; 136:964–77.
crossref
43. Dimos JT, Rodolfa KT, Niakan KK, Weisenthal LM, Mitsumoto H, Chung W, et al. Induced pluripotent stem cells generated from patients with ALS can be differentiated into motor neurons. Science. 2008; 321:1218–21.
crossref
44. Itzhaki I, Maizels L, Huber I, Zwi-Dantsis L, Caspi O, Winterstern A, et al. Modelling the long QT syndrome with induced pluripotent stem cells. Nature. 2011; 471:225–9.
crossref
45. Ebert AD, Yu J, Rose FF Jr, Mattis VB, Lorson CL, Thomson JA, et al. Induced pluripotent stem cells from a spinal muscular atrophy patient. Nature. 2009; 457:277–80.
crossref
46. Carvajal-Vergara X, Sevilla A, D'Souza SL, Ang YS, Schaniel C, Lee DF, et al. Patient-specific induced pluripotent stem-cell-derived models of LEOPARD syndrome. Nature. 2010; 465:808–12.
crossref
47. Robinton DA, Daley GQ. The promise of induced pluripotent stem cells in research and therapy. Nature. 2012; 481:295–305.
crossref
48. Raya A, Rodríguez-Pizà I, Guenechea G, Vassena R, Navarro S, Barrero MJ, et al. Disease-corrected haemato-poietic progenitors from Fanconi anaemia induced pluripotent stem cells. Nature. 2009; 460:53–9.
crossref
49. Wernig M, Zhao JP, Pruszak J, Hedlund E, Fu D, Soldner F, et al. 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. 2008; 105:5856–61.
crossref
50. Hanna J, Wernig M, Markoulaki S, Sun CW, Meissner A, Cassady JP, et al. Treatment of sickle cell anemia mouse model with iPS cells generated from autologous skin. Science. 2007; 318:1920–3.
crossref
51. Huangfu D, Osafune K, Maehr R, Guo W, Eijkelenboom A, Chen S, et al. Induction of pluripotent stem cells from primary human fibroblasts with only Oct4 and Sox2. Nat Biotechnol. 2008; 26:1269–75.
crossref
52. Lyssiotis CA, Foreman RK, Staerk J, Garcia M, Mathur D, Markoulaki S, et al. Reprogramming of murine fibroblasts to induced pluripotent stem cells with chemical complementation of Klf4. Proc Natl Acad Sci U S A. 2009; 106:8912–7.
crossref
53. Okita K, Nakagawa M, Hyenjong H, Ichisaka T, Yamanaka S. Generation of mouse induced pluripotent stem cells without viral vectors. Science. 2008; 322:949–53.
crossref
54. Lujan E, Chanda S, Ahlenius H, Südhof TC, Wernig M. Direct conversion of mouse fibroblasts to self-renewing, tripotent neural precursor cells. Proc Natl Acad Sci U S A. 2012; 109:2527–32.
crossref
55. Han DW, Tapia N, Hermann A, Hemmer K, Höing S, Araúzo-Bravo MJ, et al. Direct reprogramming of fibroblasts into neural stem cells by defined factors. Cell Stem Cell. 2012; 10:465–72.
crossref
56. Szabo E, Rampalli S, Risueño RM, Schnerch A, Mitchell R, Fiebig-Comyn A, et al. Direct conversion of human fibroblasts to multilineage blood progenitors. Nature. 2010; 468:521–6.
crossref
57. Vierbuchen T, Ostermeier A, Pang ZP, Kokubu Y, Südhof TC, Wernig M. Direct conversion of fibroblasts to functional neurons by defined factors. Nature. 2010; 463:1035–41.
crossref
58. Huangfu D, Maehr R, Guo W, Eijkelenboom A, Snitow M, Chen AE, et al. Induction of pluripotent stem cells by defined factors is greatly improved by small-molecule compounds. Nat Biotechnol. 2008; 26:795–7.
crossref
59. Ichida JK, Blanchard J, Lam K, Son EY, Chung JE, Egli D, et al. A small-molecule inhibitor of tgf-Beta signaling replaces sox2 in reprogramming by inducing nanog. Cell Stem Cell. 2009; 5:491–503.
60. Zhu S, Li W, Zhou H, Wei W, Ambasudhan R, Lin T, et al. Reprogramming of human primary somatic cells by OCT4 and chemical compounds. Cell Stem Cell. 2010; 7:651–5.
crossref
61. Hockemeyer D, Soldner F, Beard C, Gao Q, Mitalipova M, DeKelver RC, et al. Efficient targeting of expressed and silent genes in human ESCs and iPSCs using zinc-fin-ger nucleases. Nat Biotechnol. 2009; 27:851–7.
crossref
62. Mali P, Ye Z, Hommond HH, Yu X, Lin J, Chen G, et al. Improved efficiency and pace of generating induced pluripotent stem cells from human adult and fetal fibroblasts. Stem Cells. 2008; 26:1998–2005.
crossref
63. Sun N, Panetta NJ, Gupta DM, Wilson KD, Lee A, Jia F, et al. Feeder-free derivation of induced pluripotent stem cells from adult human adipose stem cells. Proc Natl Acad Sci U S A. 2009; 106:15720–5.
crossref
64. Lin C, Yu C, Ding S. Toward directed reprogramming through exogenous factors. Curr Opin Genet Dev. 2013; 23:519–25.
crossref
65. Yoshioka N, Gros E, Li HR, Kumar S, Deacon DC, Maron C, et al. Efficient generation of human iPSCs by a synthetic self-replicative RNA. Cell Stem Cell. 2013; 13:246–54.
crossref
66. Lee MO, Moon SH, Jeong HC, Yi JY, Lee TH, Shim SH, et al. Inhibition of pluripotent stem cell-derived teratoma formation by small molecules. Proc Natl Acad Sci U S A. 2013; 110:E3281–90.
crossref

Figure 1.
Vector structure of 4-in-1 lentivirus. 4-in-1 lentiviral vector contains 4 Yamanaka factors of Oct4, Klf4, Sox2, and c-Myc.
jrd-20-286f1.tif
Figure 2.
Overall process of reprogramming. 293 T cells are used for production of 4-in-1 lentivirus. Lentivirus is transduced into dermal fibroblasts. After 1∼2 weeks, cells are transformed into stem cell like morphology. After 2∼3 weeks, stem cell colonies are formed and then picked up for cultivating stem cell lines.
jrd-20-286f2.tif
Figure 3.
Summarized images during the process of iPSC reprogramming. Fibroblast begins to be changed into the shapes of self-proliferating and colony-like feature. Background fibroblasts are not reprogrammed so they remain spindle shapes. Generated three colonies are positive for pluripotency markers.
jrd-20-286f3.tif
Figure 4.
Pluripotency markers are positive at generated clones. Immunofluorescence staining shows positive pluripotency markers such as Nanog, SSEA4, Sox2, Tra-1-81, Oct4, Tra-1-60. Embryonic stem cells and iPSC are alike in positivity of immunofluoresence study.
jrd-20-286f4.tif
Figure 5.
Various levels of studies with disease-specific iPSC. OA FLS and RA FLS are compared with each other at fibroblast level. They are also studied when they are reprogrammed into iPSC clones. They can be differentiated into target disease cells such as chondrocytes and osteocytes. If we could find any differences in OA&RA iPSC-differentiated chondrocytes, it would be a promising methodology for exploring disease mechanism.
jrd-20-286f5.tif
Figure 6.
An example of application of iPSC in disease modeling in future. iPSC from patients are differentiated into syno-viocytes, T cells, B cells and so on. After methotrexate is added in culture dishes, and then the effects of MTX on target cells could be studied.
jrd-20-286f6.tif
Table 1.
Application of iPSC technique in disease modelling
Diseases studied with iPSC Differentiated cells from iPSC in vitro Disease phenotype Drug screening or functional test in vitro
Amyotrophic lateral sclerosis Motor neurons and glial cells ND No
Spinal muscular dystrophy Neurons, astrocytes Yes Yes
Parkinson's disease Dopaminergic neurons No Yes
Down's syndrome Teratoma tissue Yes No
Sickle cell anemia None NA No
Type 1 diabetes Beta cell like cells ND No
Familial dysautonomia Central nervous system cells Yes Yes

iPSC: induced pluripotent stem cell, ND: not detected, NA: not accessible.

TOOLS
Similar articles