Korean J Physiol Pharmacol.  2020 Nov;24(6):463-472. 10.4196/kjpp.2020.24.6.463.

Direct reprogramming of fibroblasts into diverse lineage cells by DNA demethylation followed by differentiating cultures

Affiliations
  • 1Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju 61186, Korea
  • 2Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan

Abstract

Direct reprogramming, also known as a trans-differentiation, is a technique to allow mature cells to be converted into other types of cells without inducing a pluripotent stage. It has been suggested as a major strategy to acquire the desired type of cells in cell-based therapies to repair damaged tissues. Studies related to switching the fate of cells through epigenetic modification have been progressing and they can bypass safety issues raised by the virus-based transfection methods. In this study, a protocol was established to directly convert fully differentiated fibroblasts into diverse mesenchymal-lineage cells, such as osteoblasts, adipocytes, chondrocytes, and ectodermal cells, including neurons, by means of DNA demethylation, immediately followed by culturing in various differentiating media. First, 24 h exposure of 5-azacytidine (5-aza-CN), a well-characterized DNA methyl transferase inhibitor, to NIH-3T3 murine fibroblast cells induced the expression of stem-cell markers, that is, increasing cell plasticity. Next, 5-aza-CN treated fibroblasts were cultured in osteogenic, adipogenic, chondrogenic, and neurogenic media with or without bone morphogenetic protein 2 for a designated period. Differentiation of each desired type of cell was verified by quantitative reverse transcriptase-polymerase chain reaction/western blot assays for appropriate marker expression and by various staining methods, such as alkaline phosphatase/alizarin red S/oil red O/alcian blue. These proposed procedures allowed easier acquisition of the desired cells without any transgenic modification, using direct reprogramming technology, and thus may help make it more available in the clinical fields of regenerative medicine.

Keyword

Cell lineage; Cellular reprogramming; DNA demethylation; Epigenesis, genetic 

Figure

  • Fig. 1 Epigenetic modification in 5-azacytidine (5-aza-CN)-treated NIH-3T3 cells. (A) After 24 h of 5 μM 5-aza-CN treatment, morphologic changes of cells were observed under an inverted microscope. (a) Untreated; (b) 5-aza-CN-treated; (c) incubated in fresh media without 5-aza-CN for an additional 24 h after treatment with 5-aza-CN. (B) After treatment at the indicated concentrations of 5-aza-CN for 24 h, the global DNA methylation patterns determined by using a methylated DNA quantification kit. (C) The expression of pluripotent stem cell markers such as SOX2, KLF4, NANOG and OCT4 genes was examined by conventional (left) and quantitative RT-PCR (RT-qPCR, right) with or without 5 μM 5-aza-CN for 24 h. (D) The protein expression of CD146, mesenchymal stem cell marker and OCT4 was analyzed by flow cytometry with or without 5 μM 5-aza-CN for 24 h, and quantified to a mean fluorescent intensity (MFI) value. Control (Ctrl) means untreated. These data represent three independent experiments. The values shown represent means ± standard errors. SOX2, sex determining region Y(SRY)-box 2; KLF4, Krüppel-like factor 4; OCT4, octamer-binding transcription factor 4. *p < 0.05.

  • Fig. 2 Osteogenic differentiation of 5-azacytidine (5-aza-CN)-treated NIH-3T3 cells. (A) The schematic diagram of this study is based on direct reprogramming principles. After 7 days culture in osteogenic differentiation medium (DMEM supplemented 2% FBS containing 170 μM AA, 5 mM β-GP and 200 ng/ml BMP2), expression of osteogenic marker genes such as alkaline phosphatase (ALP), osteopontin (OPN), osteocalcin (OCN), bone sialoprotein (BSP), Runt-related transcription factor 2 (RUNX2) and osterix (OSX) in 5 μM 5-aza-CN-treated NIH-3T3 cells was determined by RT-qPCR (B), and osteogenic differentiation of 5-aza-CN-treated NIH-3T3 was verified by ALP and alizarin red S (AR-S) staining on days 7 and 14, respectively (C). High magnification images of AR-S staining are shown in the lower panel. (D) Densities of ALP stain were quantified by Scion Image software. AR-S stain was extracted with 10% cetylpyridinium chloride and quantified by measuring the absorbance at 570 nm using an ELISA reader. These data represent three independent experiments. The values shown represent means ± standard errors. *p < 0.05.

  • Fig. 3 Adipogenic and chondrogenic differentiation of 5-azacytidine (5-aza-CN)-treated NIH-3T3 cells. (A) After 7 days culture in adipogenic differentiation medium containing DMEM supplemented 2% FBS with 0.5 mM IBMX, 1 μM DEX, 100 μM, 10 μg/ml insulin and 200 ng/ml BMP2, expression of adipogenic marker genes such as adipocyte protein 2 (AP2), CCAAT-enhancer-binding proteins (C/EBP), peroxisome proliferator-activated receptor gamma (PPARG) in 5 μM 5-aza-CN-treated NIH-3T3 cells was quantified by RT-qPCR. (B) The lipid droplet formations were observed by Oil-Red O staining on day 14 and high magnification images of Oil-Red O staining are shown in the lower panel. (C) After 7 days of chondrogenic differentiation medium culture, expression of chondrogenic marker genes such as aggrecan (ACAN), type II collagen (COL2), SRY-box 9 (SOX9) in 5 μM 5-aza-CN-treated NIH-3T3 cells was quantified by RT-qPCR. (D) The chondrogenic matrix formation was observed by Alcian blue staining on day 14 and high magnification images of Alcian blue staining are shown in the lower panel. These data represent three independent experiments. The values shown represent means ± standard errors. *p < 0.05.

  • Fig. 4 Neurogenic differentiation of 5-azacytidine (5-aza-CN)-treated NIH-3T3 cells. (A, B) After 5 and 14 days culture in neurogenic differentiation medium (NM) containing DMEM supplemented 2% FBS, 2% dimethyl sulfoxide and 100 μM butylated hydroxyanisole, expressions of neuronal specific intermediated filament genes such as neurofilament medium (NFM) and Nestin in 5 μM 5-aza-CN-treated NIH-3T3 cells were quantified by RT-qPCR and Western blot analysis, respectively. Arbitrary densities of proteins were quantified by Scion Image software, as shown in the graph below. (C) After 14 days of neurogenic differentiation medium culture, morphologic changes in 5 μM 5-aza-CN-treated NIH-3T3 cells were observed under an inverted microscope (arrows), and NFM protein expression was visualized by immunofluorescence staining. Ctrl means untreated. These data represent three independent experiments. The values shown represent means ± standard errors. *p < 0.05.


Reference

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. DOI: 10.1016/j.cell.2006.07.024. PMID: 16904174.
Article
2. Schröder AR, Shinn P, Chen H, Berry C, Ecker JR, Bushman F. 2002; HIV-1 integration in the human genome favors active genes and local hotspots. Cell. 110:521–529. DOI: 10.1016/S0092-8674(02)00864-4. PMID: 12202041.
Article
3. Wu X, Li Y, Crise B, Burgess SM. 2003; Transcription start regions in the human genome are favored targets for MLV integration. Science. 300:1749–1751. DOI: 10.1126/science.1083413. PMID: 12805549.
Article
4. Bushman F, Lewinski M, Ciuffi A, Barr S, Leipzig J, Hannenhalli S, Hoffmann C. 2005; Genome-wide analysis of retroviral DNA integration. Nat Rev Microbiol. 3:848–858. DOI: 10.1038/nrmicro1263. PMID: 16175173.
Article
5. Okita K, Nakagawa M, Hyenjong H, Ichisaka T, Yamanaka S. 2008; Generation of mouse induced pluripotent stem cells without viral vectors. Science. 322:949–953. DOI: 10.1126/science.1164270. PMID: 18845712.
Article
6. Cohen DE, Melton D. 2011; Turning straw into gold: directing cell fate for regenerative medicine. Nat Rev Genet. 12:243–252. DOI: 10.1038/nrg2938. PMID: 21386864.
Article
7. Graf T, Enver T. 2009; Forcing cells to change lineages. Nature. 462:587–594. DOI: 10.1038/nature08533. PMID: 19956253.
Article
8. Guo C, Morris SA. 2017; Engineering cell identity: establishing new gene regulatory and chromatin landscapes. Curr Opin Genet Dev. 46:50–57. DOI: 10.1016/j.gde.2017.06.011. PMID: 28667865.
Article
9. Davis RL, Weintraub H, Lassar AB. 1987; Expression of a single transfected cDNA converts fibroblasts to myoblasts. Cell. 51:987–1000. DOI: 10.1016/0092-8674(87)90585-X. PMID: 3690668.
Article
10. Maza I, Caspi I, Zviran A, Chomsky E, Rais Y, Viukov S, Geula S, Buenrostro JD, Weinberger L, Krupalnik V, Hanna S, Zerbib M, Dutton JR, Greenleaf WJ, Massarwa R, Novershtern N, Hanna JH. 2015; Transient acquisition of pluripotency during somatic cell transdifferentiation with iPSC reprogramming factors. Nat Biotechnol. 33:769–774. DOI: 10.1038/nbt.3270. PMID: 26098448. PMCID: PMC4500825.
Article
11. Xu J, Du Y, Deng H. 2015; Direct lineage reprogramming: strategies, mechanisms, and applications. Cell Stem Cell. 16:119–134. DOI: 10.1016/j.stem.2015.01.013. PMID: 25658369.
Article
12. Song K, Nam YJ, Luo X, Qi X, Tan W, Huang GN, Acharya A, Smith CL, Tallquist MD, Neilson EG, Hill JA, Bassel-Duby R, Olson EN. 2012; Heart repair by reprogramming non-myocytes with cardiac transcription factors. Nature. 485:599–604. DOI: 10.1038/nature11139. PMID: 22660318. PMCID: PMC3367390.
Article
13. Chanda S, Ang CE, Davila J, Pak C, Mall M, Lee QY, Ahlenius H, Jung SW, Südhof TC, Wernig M. 2014; Generation of induced neuronal cells by the single reprogramming factor ASCL1. Stem Cell Reports. 3:282–296. DOI: 10.1016/j.stemcr.2014.05.020. PMID: 25254342. PMCID: PMC4176533.
Article
14. Waddington CH. 1957. The strategy of the genes: a discussion of some aspects of theoretical biology. With an appendix by H. Kacser. George Allen & Unwin;London:
15. Goldberg AD, Allis CD, Bernstein E. 2007; Epigenetics: a landscape takes shape. Cell. 128:635–638. DOI: 10.1016/j.cell.2007.02.006. PMID: 17320500.
Article
16. Avgustinova A, Benitah SA. 2016; Epigenetic control of adult stem cell function. Nat Rev Mol Cell Biol. 17:643–658. DOI: 10.1038/nrm.2016.76. PMID: 27405257.
Article
17. Baubec T, Colombo DF, Wirbelauer C, Schmidt J, Burger L, Krebs AR, Akalin A, Schübeler D. 2015; Genomic profiling of DNA methyltransferases reveals a role for DNMT3B in genic methylation. Nature. 520:243–247. DOI: 10.1038/nature14176. PMID: 25607372.
Article
18. Pennarossa G, Maffei S, Campagnol M, Tarantini L, Gandolfi F, Brevini TA. 2013; Brief demethylation step allows the conversion of adult human skin fibroblasts into insulin-secreting cells. Proc Natl Acad Sci U S A. 110:8948–8953. DOI: 10.1073/pnas.1220637110. PMID: 23696663. PMCID: PMC3670366.
Article
19. Cho YD, Yoon WJ, Kim WJ, Woo KM, Baek JH, Lee G, Ku Y, van Wijnen AJ, Ryoo HM. 2014; Epigenetic modifications and canonical wingless/int-1 class (WNT) signaling enable trans-differentiation of nonosteogenic cells into osteoblasts. J Biol Chem. 289:20120–20128. DOI: 10.1074/jbc.M114.558064. PMID: 24867947. PMCID: PMC4106327.
Article
20. Stresemann C, Lyko F. 2008; Modes of action of the DNA methyltransferase inhibitors azacytidine and decitabine. Int J Cancer. 123:8–13. DOI: 10.1002/ijc.23607. PMID: 18425818.
Article
21. Heng JC, Orlov YL, Ng HH. 2010; Transcription factors for the modulation of pluripotency and reprogramming. Cold Spring Harb Symp Quant Biol. 75:237–244. DOI: 10.1101/sqb.2010.75.003. PMID: 21047904.
Article
22. Hua J, Gong J, Meng H, Xu B, Yao L, Qian M, He Z, Zou S, Zhou B, Song Z. 2014; Comparison of different methods for the isolation of mesenchymal stem cells from umbilical cord matrix: proliferation and multilineage differentiation as compared to mesenchymal stem cells from umbilical cord blood and bone marrow. Cell Biol Int. 38:198–210. DOI: 10.1002/cbin.10188. PMID: 24123669.
Article
23. An J, Yang H, Zhang Q, Liu C, Zhao J, Zhang L, Chen B. 2016; Natural products for treatment of osteoporosis: the effects and mechanisms on promoting osteoblast-mediated bone formation. Life Sci. 147:46–58. DOI: 10.1016/j.lfs.2016.01.024. PMID: 26796578.
Article
24. MacDougald OA, Lane MD. 1995; Transcriptional regulation of gene expression during adipocyte differentiation. Annu Rev Biochem. 64:345–373. DOI: 10.1146/annurev.bi.64.070195.002021. PMID: 7574486.
Article
25. Kurakazu I, Akasaki Y, Hayashida M, Tsushima H, Goto N, Sueishi T, Toya M, Kuwahara M, Okazaki K, Duffy T, Lotz MK, Nakashima Y. 2019; FOXO1 transcription factor regulates chondrogenic differentiation through transforming growth factor β1 signaling. J Biol Chem. 294:17555–17569. DOI: 10.1074/jbc.RA119.009409. PMID: 31601652. PMCID: PMC6873195.
Article
26. Li D, Zou XY, El-Ayachi I, Romero LO, Yu Z, Iglesias-Linares A, Cordero-Morales JF, Huang GT. 2019; Human dental pulp stem cells and gingival mesenchymal stem cells display action potential capacity in vitro after neuronogenic differentiation. Stem Cell Rev Rep. 15:67–81. DOI: 10.1007/s12015-018-9854-5. PMID: 30324358. PMCID: PMC6358481.
Article
27. Whitelaw E, Sutherland H, Kearns M, Morgan H, Weaving L, Garrick D. 2001; Epigenetic effects on transgene expression. Methods Mol Biol. 158:351–368. DOI: 10.1385/1-59259-220-1:351. PMID: 11236667.
Article
28. Spada F, Haemmer A, Kuch D, Rothbauer U, Schermelleh L, Kremmer E, Carell T, Längst G, Leonhardt H. 2007; DNMT1 but not its interaction with the replication machinery is required for maintenance of DNA methylation in human cells. J Cell Biol. 176:565–571. DOI: 10.1083/jcb.200610062. PMID: 17312023.
Article
29. Darmon M, Nicolas JF, Lamblin D. 1984; 5-Azacytidine is able to induce the conversion of teratocarcinoma-derived mesenchymal cells into epithelia cells. EMBO J. 3:961–967. DOI: 10.1002/j.1460-2075.1984.tb01914.x. PMID: 6203746. PMCID: PMC557458.
Article
30. Riemens RJM, van den Hove DLA, Esteller M, Delgado-Morales R. 2018; Directing neuronal cell fate in vitro: achievements and challenges. Prog Neurobiol. 168:42–68. DOI: 10.1016/j.pneurobio.2018.04.003. PMID: 29653249.
31. Raciti M, Granzotto M, Duc MD, Fimiani C, Cellot G, Cherubini E, Mallamaci A. 2013; Reprogramming fibroblasts to neural-precursor-like cells by structured overexpression of pallial patterning genes. Mol Cell Neurosci. 57:42–53. DOI: 10.1016/j.mcn.2013.10.004. PMID: 24128663.
Article
32. Cheng L, Hu W, Qiu B, Zhao J, Yu Y, Guan W, Wang M, Yang W, Pei G. 2015; Generation of neural progenitor cells by chemical cocktails and hypoxia. Cell Res. 25:645–646. DOI: 10.1038/cr.2015.55. PMID: 25941173. PMCID: PMC4423089.
Article
Full Text Links
  • KJPP
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr