DOI QR코드

DOI QR Code

Histone deacetylases inhibitor and RAD51 recombinase increase transcription activator-like effector nucleases-mediated homologous recombination on the bovine β-casein gene locus

  • Park, Da Som (Department of Animal Science, Chonnam National University) ;
  • Kim, Se Eun (Department of Animal Science, Chonnam National University) ;
  • Koo, Deog-Bon (Department of Biotechnology, College of Engineering, Daegu University) ;
  • Kang, Man-Jong (Department of Animal Science, Chonnam National University)
  • Received : 2019.08.13
  • Accepted : 2019.10.14
  • Published : 2020.06.01

Abstract

Objective: The efficiency of the knock-in process is very important to successful gene editing in domestic animals. Recently, it was reported that transient loosening of the nucleosomal folding of transcriptionally inactive chromatin might have the potential to enhance homologous recombination efficiency. The objective of this study was to determine whether histone deacetylases (HDAC) inhibitor and RAD51 recombinase (RAD51) expression were associated with increased knock-in efficiency on the β-casein (bCSN2) gene locus in mammary alveolar-large T antigen (MAC-T) cells using the transcription activator-like effector nucleases (TALEN) system. Methods: MAC-T cells were treated with HDAC inhibitors, valproic acid, trichostatin A, or sodium butyrate for 24 h, then transfected with a knock-in vector, RAD51 expression vector and TALEN to target the bCSN2 gene. After 3 days of transfection, the knock-in efficiency was confirmed by polymerase chain reaction and DNA sequencing of the target site. Results: The level of HDAC 2 protein in MAC-T cells was decreased by treatment with HDAC inhibitors. The knock-in efficiency in MAC-T cells treated with HDAC inhibitors was higher than in cells not treated with inhibitors. However, the length of the homologous arm of the knock-in vector made no difference in the knock-in efficiency. Furthermore, DNA sequencing confirmed that the precision of the knock-in was more efficient in MAC-T cells treated with sodium butyrate. Conclusion: These results indicate that chromatin modification by HDAC inhibition and RAD51 expression enhanced the homologous recombination efficiency on the bCSN2 gene locus in MAC-T cells.

Keywords

References

  1. Gaj T, Gersbach CA, Barbas CF 3rd. ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol 2013;31:397-405. https://doi.org/10.1016/j.tibtech.2013.04.004
  2. Sakuma T, Woltjen K. Nuclease-mediated genome editing: At the front-line of functional genomics technology. Dev Growth Differ 2014;56:2-13. https://doi.org/10.1111/dgd.12111
  3. Tan W, Proudfoot C, Lillico SG, Whitelaw CBA. Gene targeting, genome editing: from Dolly to editors. Transgenic Res 2016;25:273-87. https://doi.org/10.1007/s11248-016-9932-x
  4. Park TS. Current strategies of genomic modification in livestock and applications in poultry. J Anim Reprod Biotechnol 2019;34:65-9. https://doi.org/10.12750/JARB.34.2.65.
  5. Porteus MH, Carroll D. Gene targeting using zinc finger nucleases. Nat Biotechnol 2005;23:967-73. https://doi.org/10.1038/nbt1125
  6. Wei C, Liu J, Yu Z, Zhang B, Gao G, Jiao R. TALEN or Cas9 - rapid, efficient and specific choices for genome modifications. J Genet Genomics 2013;40:281-9. https://doi.org/10.1016/j.jgg.2013.03.013
  7. Liu X, Wang Y, Guo W, et al. Zinc-finger nickase-mediated insertion of the lysostaphin gene into the $\beta$-casein locus in cloned cows. Nat Commun 2013;4:2565. https://doi.org/10.1038/ncomms3565
  8. Liu X, Wang Y, Tian Y, et al. Generation of mastitis resistance in cows by targeting human lysozyme gene to $\beta$-casein locus using zinc-finger nucleases. Proc Biol Sci 2014;281:20133368. https://doi.org/10.1098/rspb.2013.3368
  9. Jeong YH, Kim YJ, Kim EY, et al. Knock-in fibroblasts and transgenic blastocysts for expression of human FGF2 in the bovine $\beta$-casein gene locus using CRISPR/Cas9 nuclease-mediated homologous recombination. Zygote 2016;24:442-56. https://doi.org/10.1017/S0967199415000374
  10. Yuan YG, Song SZ, Zhu MM, et al. Human lactoferrin efficiently targeted into caprine beta-lactoglobulin locus with transcription activator-like effector nucleases. Asian-Australas J Anim Sci 2017;30:1175-82. https://doi.org/10.5713/ajas.16.0697
  11. Lin S, Staahl BT, Alla RK, Doudna JA. Enhanced homology-directed human genome engineering by controlled timing of CRISPR/Cas9 delivery. eLife 2014;3:e04766. https://doi.org/10.7554/eLife.04766
  12. Chu VT, Weber T, Wefers B, et al. Increasing the efficiency of homology-directed repair for CRISPR-Cas9-induced precise gene editing in mammalian cells. Nat Biotechnol 2015;33:543-8. https://doi.org/10.1038/nbt.3198
  13. Song J, Yang D, Xu J, Zhu T, Chen YE, Zhang J. RS-1 enhances CRISPR/Cas9- and TALEN-mediated knock-in efficiency. Nat Commun 2016;7:10548. https://doi.org/10.1038/ncomms10548
  14. Verkuijl SAN, Rots MG. The influence of eukaryotic chromatin state on CRISPR-Cas9 editing efficiencies. Curr Opin Biotechnol 2019;55:68-73. https://doi.org/10.1016/j.copbio.2018.07.005
  15. Dae RM, Cutts JP, Brafman DA, Haynes KA. The impact of chromatin dynamics on Cas9-mediated genome editing in human cells. ACS Synth Biol 2017;6:428-38. https://doi.org/10.1021/acssynbio.5b00299
  16. Takayama K, Igai K, Hagihara Y, et al. Highly efficient biallelic genome editing of human ES/iPS cells using a CRISPR/Cas9 or TALEN system. Nucleic Acids Res 2017;45:5198-207. https://doi.org/10.1093/nar/gkx130
  17. Kim SE, Park DS, Koo D-B, Kang M-J. Knock-in efficiency depending on homologous arm structure on the knock-in vector in bovine fibroblasts. Reprod Dev Biol 2017;41:7-16. https://doi.org/10.12749/RDB.2017.41.1.7
  18. Mao Z, Bozzella M, Seluanov A, Gorbunova V. DNA repair by nonhomologous end joining and homologous recombination during cell cycle in human cells. Cell Cycle 2008;7:2902-6. https://doi.org/10.4161/cc.7.18.6679
  19. Hockemeyer D, Wang H, Kiani S, et al. Genetic engineering of human pluripotent cells using TALE nucleases. Nat Biotechnol 2011;29:731-4. https://doi.org/10.1038/nbt.1927
  20. Hisano Y, Sakuma T, Nakade S, et al. Precise in-frame integration of exogenous DNA mediated by CRISPR/Cas9 system in zebrafish. Sci Rep 2015;5:8841. https://doi.org/10.1038/srep08841
  21. Yoshimi K, Kunihiro Y, Kaneko T, Nagahora H, Voigt B, Mashimo T. ssODN-mediated knock-in with CRISPR-Cas for large genomic regions in zygotes. Nat Commun 2016;7:10431. https://doi.org/10.1038/ncomms10431
  22. Zhang JP, Li XL, Li GH, et al. Efficient precise knockin with a double cut HDR donor after CRISPR/Cas9-mediated double-stranded DNA cleavage. Genome Biol 2017;18:35. https://doi.org/10.1186/s13059-017-1164-8
  23. Thomas KR, Deng C, Capecchi MR. High-fidelity gene targeting in embryonic stem cells by using sequence replacement vectors. Mol Cell Biol 1992;12:2919-23. https://doi.org/10.1128/MCB.12.7.2919
  24. Urnov FD, Miller JC, Lee YL, et al. Highly efficient endogenous human gene correction using designed zinc finger nucleases. Nature 2005;435:646-51. https://doi.org/10.1038/nature03556
  25. Byrne SM, Ortiz L, Mali P, Aach J, Church GM. Multi-kilo base homozygous targeted gene replacement in human induced pluripotent stem cells. Nucleic Acids Res 2015;43:e21. https://doi.org/10.1093/nar/gku1246
  26. Maruyama T, Dougan SK, Truttmann MC, Bilate AM, Ingram JR, Ploegh HL. Increasing the efficiency of precise genome editing with CRISPR-Cas9 by inhibition of non homologous end joining. Nat Biotechnol 2015;33:538-42. https://doi.org/10.1038/nbt.3190
  27. Howden SE, Maufort JP, Duffin BM, Elefanty AG, Stanley EG, Thomson JA. Simultaneous reprogramming and gene correction of patient fibroblasts. Stem Cell Rep 2015;5:1109-18. https://doi.org/10.1016/j.stemcr.2015.10.009
  28. Raveux A, Vandormael-Pournin S, Cohen-Tannoudji M. Optimization of the production of knock-in alleles by CRISPR/Cas9 microinjection into the mouse zygote. Sci Rep 2017;7:42661. https://doi.org/10.1038/srep42661