Hostname: page-component-848d4c4894-4hhp2 Total loading time: 0 Render date: 2024-05-31T13:47:12.747Z Has data issue: false hasContentIssue false

Disruption of exogenous eGFP gene using RNA-guided endonuclease in bovine transgenic somatic cells

Published online by Cambridge University Press:  26 November 2014

WooJae Choi
Affiliation:
Laboratory of Theriogenology and Biotechnology, Department of Veterinary Clinical Science, College of Veterinary Medicine and the Research Institute of Veterinary Science, Seoul National University, Republic of Korea.
SooYoung Yum
Affiliation:
Laboratory of Theriogenology and Biotechnology, Department of Veterinary Clinical Science, College of Veterinary Medicine and the Research Institute of Veterinary Science, Seoul National University, Republic of Korea.
SongJeon Lee
Affiliation:
Embryo Research Center in Seoul Milk Coop., Gyeonggi-do, Republic of Korea.
WonWu Lee
Affiliation:
Embryo Research Center in Seoul Milk Coop., Gyeonggi-do, Republic of Korea.
JiHyun Lee
Affiliation:
Laboratory of Theriogenology and Biotechnology, Department of Veterinary Clinical Science, College of Veterinary Medicine and the Research Institute of Veterinary Science, Seoul National University, Republic of Korea.
SeokJoong Kim
Affiliation:
Toolgen, INC., Seoul, Republic of Korea.
OkJae Koo
Affiliation:
Laboratory Animal Research Center, Samsung Biomedical Research Institute, Gyeonggi-do, Republic of Korea.
ByeongChun Lee
Affiliation:
Laboratory of Theriogenology and Biotechnology, Department of Veterinary Clinical Science, College of Veterinary Medicine and the Research Institute of Veterinary Science, Seoul National University, Republic of Korea.
Goo Jang*
Affiliation:
Laboratory of Theriogenology and Biotechnology, Department of Veterinary Clinical Science, College of Veterinary Medicine and the Research Institute of Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 151–742Korea. Emergence Center for Food-Medicine Personalized Therapy System, Advanced Institutes of Convergence Technology, Seoul National University, Gyeonggi-do, Republic of Korea.
*
All correspondence to: Goo Jang. Laboratory of Theriogenology and Biotechnology, Department of Veterinary Clinical Science, College of Veterinary Medicine and the Research Institute of Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 151–742Korea. Tel: +82 2 880 1280. Fax: +82 873 1269. e-mail: snujang@snu.ac.kr

Summary

Genome-editing technologies are considered to be an important tool for generating gene knockout cattle models. Here, we report highly efficient disruption of a chromosomally integrated eGFP gene in bovine somatic cells using RNA-guided endonucleases, a new class of programmable nucleases developed from a bacterial Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 system. In the present study, we obtained homogenously eGFP-expressing primary fibroblasts from cloned bovine transgenic embryonic tissues and employed them for further analysis. CRISPR/Cas9 plasmids specifically targeting the eGFP gene were transfected into the eGFP fibroblasts by electroporation. After 10 days of culture, more than 40% of the cells had lost eGFP expression in fluorescence activated cell sorting (FACS) analysis. Targeted sequences of the transfected cells were analyzed, and various small indel mutations (6–203 bp deletions) in the target sequence were found. The fibroblasts mutated with the CRISPR/Cas9 system were applied for somatic cell nuclear transfer, and the reconstructed embryos were successfully developed into the blastocyst stage. In conclusion, the CRISPR/Cas9 system was successfully utilized in bovine cells and cloned embryos. This will be a useful technique to develop livestock transgenesis for agricultural science.

Type
Research Article
Copyright
Copyright © Cambridge University Press 2014 

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Carlson, D.F., Tan, W., Lillico, S.G., Stverakova, D., Proudfoot, C., Christian, M., Voytas, D.F., Long, C.R., Whitelaw, C.B. & Fahrenkrug, S.C. (2012). Efficient TALEN-mediated gene knockout in livestock. Proc. Natl. Acad. Sci. USA 109, 17382–7.CrossRefGoogle ScholarPubMed
Cho, S.W., Kim, S., Kim, J.M. & Kim, J.S. (2013). Targeted genome engineering in human cells with the Cas9 RNA-guided endonuclease. Nat. Biotechnol. 31, 230–2.CrossRefGoogle ScholarPubMed
Cho, S.W., Kim, S., Kim, Y., Kweon, J., Kim, H.S., Bae, S. & Kim, J.S. (2014). Analysis of off-target effects of CRISPR/Cas-derived RNA-guided endonucleases and nickases. Genome Res. 24, 13241.CrossRefGoogle ScholarPubMed
Clark, A.J., Burl, S., Denning, C. & Dickinson, P. (2000). Gene targeting in livestock: a preview. Transgenic Res. 9, 263–75.Google Scholar
Dean, W., Santos, F., Stojkovic, M., Zakhartchenko, V., Walter, J., Wolf, E. & Reik, W. (2001). Conservation of methylation reprogramming in mammalian development: aberrant reprogramming in cloned embryos. Proc. Natl. Acad. Sci. USA 98, 13734–8.CrossRefGoogle ScholarPubMed
Ding, S., Wu, X., Li, G., Han, M., Zhuang, Y. & Xu, T. (2005). Efficient transposition of the piggyBac (PB) transposon in mammalian cells and mice. Cell 122, 473–83.Google Scholar
Galli, C., Duchi, R., Crotti, G., Turini, P., Ponderato, N., Colleoni, S., Lagutina, I. & Lazzari, G. (2003). Bovine embryo technologies. Theriogenology 59, 599616.CrossRefGoogle ScholarPubMed
Guan, C., Ye, C., Yang, X. & Gao, J. (2010). A review of current large-scale mouse knockout efforts. Genesis 48, 7385.CrossRefGoogle ScholarPubMed
Hasler, J.F. (2003). The current status and future of commercial embryo transfer in cattle. Anim. Reprod. Sci. 79, 245–64.CrossRefGoogle ScholarPubMed
Huang, X., Guo, H., Tammana, S., Jung, Y.C., Mellgren, E., Bassi, P., Cao, Q., Tu, Z.J., Kim, Y.C., Ekker, S.C., Wu, X., Wang, S.M. & Zhou, X. (2010). Gene transfer efficiency and genome-wide integration profiling of Sleeping Beauty, Tol2, and piggyBac transposons in human primary T cells. Mol. Ther. 18, 1803–13.Google Scholar
Jacob, H.J., Lazar, J., Dwinell, M.R., Moreno, C. & Geurts, A.M. (2010). Gene targeting in the rat: advances and opportunities. Trends Genet. 26, 510–8.Google Scholar
Jang, G., Hong, S.G. & Lee, B.C. (2011). Cloned calves derived from somatic cell nuclear transfer embryos cultured in chemically defined medium or modified synthetic oviduct fluid. J. Vet. Sci. 12, 83–9.CrossRefGoogle ScholarPubMed
Kim, S., Saadeldin, I.M., Choi, W.J., Lee, S.J., Lee, W.W., Kim, B.H., Han, H.J., Bang du, H., Lee, B.C. & Jang, G. (2011). Production of transgenic bovine cloned embryos using piggybac transposition. J. Vet. Med. Sci. 73, 1453–7.Google Scholar
Kim, Y., Kweon, J. & Kim, J.S. (2013). TALENs and ZFNs are associated with different mutation signatures. Nat. Methods 10, 185.Google Scholar
Lai, L., Kolber-Simonds, D., Park, K.W., Cheong, H.T., Greenstein, J.L., Im, G.S., Samuel, M., Bonk, A., Rieke, A., Day, B.N., Murphy, C.N., Carter, D.B., Hawley, R.J. & Prather, R.S. (2002). Production of alpha-1,3-galactosyltransferase knockout pigs by nuclear transfer cloning. Science 295, 1089–92.Google Scholar
Mapletoft, R.J. & Hasler, J.F. (2005). Assisted reproductive technologies in cattle: a review. Rev. Sci. Tech. 24, 393403.CrossRefGoogle ScholarPubMed
Niemann, H. & Lucas-Hahn, A. (2012). Somatic cell nuclear transfer cloning: practical applications and current legislation. Reprod. Domest. Anim. 47 (Suppl. 5), 210.Google Scholar
Rogers, C.S., Stoltz, D.A., Meyerholz, D.K., Ostedgaard, L.S., Rokhlina, T., Taft, P.J., Rogan, M.P., Pezzulo, A.A., Karp, P.H., Itani, O.A., Kabel, A.C., Wohlford-Lenane, C.L., Davis, G.J., Hanfland, R.A., Smith, T.L., Samuel, M., Wax, D., Murphy, C.N., Rieke, A., Whitworth, K., Uc, A., Starner, T.D., Brogden, K.A., Shilyansky, J., McCray, P.B. Jr., Zabner, J., Prather, R.S. & Welsh, M.J. (2008). Disruption of the CFTR gene produces a model of cystic fibrosis in newborn pigs. Science 321, 1837–41.Google Scholar
Sung, Y.H., Baek, I.J., Kim, D.H., Jeon, J., Lee, J., Lee, K., Jeong, D., Kim, J.S. & Lee, H.W. (2013). Knockout mice created by TALEN-mediated gene targeting. Nat. Biotechnol. 31, 23–4.Google Scholar
Sung, Y.H., Kim, J.M., Kim, H.T., Lee, J., Jeon, J., Jin, Y., Choi, J.H., Ban, Y.H., Ha, S.J., Kim, C.H., Lee, H.W. & Kim, J.S. (2014). Highly efficient gene knockout in mice and zebrafish with RNA-guided endonucleases. Genome Res. 24, 125–31.CrossRefGoogle ScholarPubMed
Tan, W., Carlson, D.F., Lancto, C.A., Garbe, J.R., Webster, D.A., Hackett, P.B. & Fahrenkrug, S.C. (2013). Efficient nonmeiotic allele introgression in livestock using custom endonucleases. Proc. Natl. Acad. Sci. USA 110, 16526–31.Google Scholar
Wheeler, M.B. (2003). Production of transgenic livestock: promise fulfilled. J. Anim. Sci. 81 (Suppl. 3), 32–7.Google Scholar
Yang, D., Yang, H., Li, W., Zhao, B., Ouyang, Z., Liu, Z., Zhao, Y., Fan, N., Song, J., Tian, J., Li, F., Zhang, J., Chang, L., Pei, D., Chen, Y.E. & Lai, L.(2011). Generation of PPARgamma mono-allelic knockout pigs via zinc-finger nucleases and nuclear transfer cloning. Cell Res. 21, 979–82.CrossRefGoogle ScholarPubMed
Yu, S., Luo, J., Song, Z., Ding, F., Dai, Y. & Li, N. (2011). Highly efficient modification of beta-lactoglobulin (BLG) gene via zinc-finger nucleases in cattle. Cell Res. 21, 1638–40.CrossRefGoogle ScholarPubMed
Supplementary material: File

Choi Supplementary Material

Figure S1

Download Choi Supplementary Material(File)
File 180.4 KB