DOI QR코드

DOI QR Code

Myostatin gene knockout mediated by Cas9-D10A nickase in chicken DF1 cells without off-target effect

  • Lee, Jeong Hyo (Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University) ;
  • Kim, Si Won (Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University) ;
  • Park, Tae Sub (Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University)
  • Received : 2016.09.16
  • Accepted : 2016.10.06
  • Published : 2017.05.01

Abstract

Objective: Based on rapid advancement of genetic modification techniques, genomic editing is expected to become the most efficient tool for improvement of economic traits in livestock as well as poultry. In this study, we examined and verified the nickase of mutated CRISPR-associated protein 9 (Cas9) to modulate the specific target gene in chicken DF1 cells. Methods: Chicken myostatin which inhibits muscle cell growth and differentiation during myogenesis was targeted to be deleted and mutated by the Cas9-D10A nickase. After co-transfection of the nickase expression vector with green fluorescent gene (GFP) gene and targeted multiplex guide RNAs (gRNAs), the GFP-positive cells were sorted out by fluorescence-activated cell sorting procedure. Results: Through the genotyping analysis of the knockout cells, the mutant induction efficiency was 100% in the targeted site. Number of the deleted nucleotides ranged from 2 to 39 nucleotide deletion. There was no phenotypic difference between regular cells and knockout cells. However, myostatin protein was not apparently detected in the knockout cells by Western blotting. Additionally, six off-target sites were predicted and analyzed but any non-specific mutation in the off-target sites was not observed. Conclusion: The knockout technical platform with the nickase and multiplex gRNAs can be efficiently and stablely applied to functional genomics study in poultry and finally adapted to generate the knockout poultry for agribio industry.

Keywords

References

  1. Palmiter RD, Norstedt G, Gelinas RE, Hammer RE, Brinster RL. Metallothionein-human GH fusion genes stimulate growth of mice. Science 1983;222:809-14. https://doi.org/10.1126/science.6356363
  2. Park TS, Han JY. piggyBac transposition into primordial germ cells is an efficient tool for transgenesis in chickens. Proc Natl Acad Sci USA 2012;109:9337-41. https://doi.org/10.1073/pnas.1203823109
  3. Macdonald J, Taylor L, Sherman A, et al. Efficient genetic modification and germ-line transmission of primordial germ cells using piggyBac and Tol2 transposons. Proc Natl Acad Sci USA 2012;109:E1466-72. https://doi.org/10.1073/pnas.1118715109
  4. Park TS, Han JY. Genetic modification of chicken germ cells. Ann NY Acad Sci USA 2012:1271:104-9. https://doi.org/10.1111/j.1749-6632.2012.06744.x
  5. Thompson S, Clarke AR, Pow AM, Hooper ML, Melton DW. Germ line transmission and expression of a corrected HPRT gene produced by gene targeting in embryonic stem cells. Cell 1989;56:313-21. https://doi.org/10.1016/0092-8674(89)90905-7
  6. Koller BH, Hagemann LJ, Doetschman T, et al. Germ-line transmission of a planned alteration made in a hypoxanthine phosphoribosyltransferase gene by homologous recombination in embryonic stem cells. Proc Natl Acad Sci USA 1989;86:8927-31. https://doi.org/10.1073/pnas.86.22.8927
  7. Zijlstra M, Li E, Sajjadi F, Subramani S, Jaenisch R. Germ-line transmission of a disrupted beta 2-microglobulin gene produced by homologous recombination in embryonic stem cells. Nature 1989;342:435-8. https://doi.org/10.1038/342435a0
  8. Schwartzberg PL, Goff SP, Robertson EJ. Germ-line transmission of a c-abl mutation produced by targeted gene disruption in ES cells. Science 1989;246:799-803. https://doi.org/10.1126/science.2554496
  9. Park TS, Lee HJ, Kim KH, Kim JS, Han JY. Targeted gene knockout in chickens mediated by TALENs. Proc Natl Acad Sci USA 2014;111:12716-21. https://doi.org/10.1073/pnas.1410555111
  10. Cong L, Ran FA, Cox D, et al. Multiplex genome engineering using CRISPR/Cas systems. Science 2013;339:819-23. https://doi.org/10.1126/science.1231143
  11. Mali P, Yang L, Esvelt KM, et al. RNA-guided human genome engineering via Cas9. Science 2013;339:823-6. https://doi.org/10.1126/science.1232033
  12. Ran FA, Hsu PD, Lin CY, et al. Double nicking by RNA-guided CRISPR Cas9 for enhanced genome editing specificity. Cell 2013;154:1380-9. https://doi.org/10.1016/j.cell.2013.08.021
  13. Wang H, Yang H, Shivalila CS, et al. One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. Cell 2013;153:910-8. https://doi.org/10.1016/j.cell.2013.04.025
  14. Jao LE, Wente SR, Chen W. Efficient multiplex biallelic zebrafish genome editing using a CRISPR nuclease system. Proc Natl Acad Sci USA 2013;110:13904-9. https://doi.org/10.1073/pnas.1308335110
  15. Niu Y, Shen B, Cui Y, et al. Generation of gene-modified cynomolgus monkey via Cas9/RNA-mediated gene targeting in one-cell embryos. Cell 2014;156:836-43. https://doi.org/10.1016/j.cell.2014.01.027
  16. Hai T, Teng F, Guo R, Li W, Zhou Q. One-step generation of knockout pigs by zygote injection of CRISPR/Cas system. Cell Res 2014;24:372-5. https://doi.org/10.1038/cr.2014.11
  17. Stern CD. The chick; a great model system becomes even greater. Dev Cell 2005;8:9-17.
  18. Schusser B, Collarini EJ, Yi H, et al. Immunoglobulin knockout chickens via efficient homologous recombination in primordial germ cells. Proc Natl Acad Sci USA 2013;110:20170-5. https://doi.org/10.1073/pnas.1317106110
  19. Fu Y, Foden JA, Khayter C, et al. High-frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells. Nat Biotechnol 2013;31:822-6. https://doi.org/10.1038/nbt.2623
  20. Zhang XH, Tee LY, Wang XG, Huang QS, Yang SH. Off-target effects in CRISPR/Cas9-mediated genome engineering. Mol Ther Nucleic Acids 2015;4:e264. https://doi.org/10.1038/mtna.2015.37
  21. Cho SW, Kim S, Kim Y, et al. Analysis of off-target effects of CRISPR/Cas-derived RNA-guided endonucleases and nickases. Genome Res 2014;24:132-41. https://doi.org/10.1101/gr.162339.113
  22. McPherron AC, Lawler AM, Lee SJ. Regulation of skeletal muscle mass in mice by a new TGF-beta superfamily member. Nature 1997;387:83-90. https://doi.org/10.1038/387083a0
  23. Kambadur R, Sharma M, Smith TPL, Bass JJ. Mutations in myostatin (GDF8) in double-muscled Belgian Blue and Piedmontese cattle. Genome Res 1997;7:910-5. https://doi.org/10.1101/gr.7.9.910
  24. Schuelke M, Wagner KR, Stolz LE, et al. Myostatin mutation associated with gross muscle hypertrophy in a child. N Engl J Med 2004;350:2682-8. https://doi.org/10.1056/NEJMoa040933

Cited by

  1. CRISPR/Cascade 9-Mediated Genome Editing-Challenges and Opportunities vol.9, pp.1664-8021, 2018, https://doi.org/10.3389/fgene.2018.00240
  2. Forkhead box O3 promotes cell proliferation and inhibits myotube differentiation in chicken myoblast cells vol.60, pp.1, 2019, https://doi.org/10.1080/00071668.2018.1547362
  3. CRISPR/Cas9 gene editing in a chicken model: current approaches and applications vol.61, pp.2, 2017, https://doi.org/10.1007/s13353-020-00537-9
  4. Functional analyses of miRNA-146b-5p during myogenic proliferation and differentiation in chicken myoblasts vol.21, pp.1, 2017, https://doi.org/10.1186/s12860-020-00284-z
  5. Recent Advances in the Application of CRISPR/Cas9 Gene Editing System in Poultry Species vol.12, pp.None, 2017, https://doi.org/10.3389/fgene.2021.627714
  6. CRISPR Genome Editing Technology: A Powerful Tool Applied to Developing Agribusiness vol.69, pp.23, 2017, https://doi.org/10.1021/acs.jafc.1c01062