Browse > Article
http://dx.doi.org/10.5713/ajas.16.0749

Myotube differentiation in clustered regularly interspaced short palindromic repeat/Cas9-mediated MyoD knockout quail myoblast cells  

Kim, Si Won (Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University)
Lee, Jeong Hyo (Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University)
Park, Byung-Chul (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)
Publication Information
Asian-Australasian Journal of Animal Sciences / v.30, no.7, 2017 , pp. 1029-1036 More about this Journal
Abstract
Objective: In the livestock industry, the regulatory mechanisms of muscle proliferation and differentiation can be applied to improve traits such as growth and meat production. We investigated the regulatory pathway of MyoD and its role in muscle differentiation in quail myoblast cells. Methods: The MyoD gene was mutated by the clustered regularly interspaced short palindromic repeat (CRISPR)/Cas9 technology and single cell-derived MyoD mutant sublines were identified to investigate the global regulatory mechanism responsible for muscle differentiation. Results: The mutation efficiency was 73.3% in the mixed population, and from this population we were able to establish two QM7 MyoD knockout subline (MyoD KO QM7#4) through single cell pick-up and expansion. In the undifferentiated condition, paired box 7 expression in MyoD KO QM7#4 cells was not significantly different from regular QM7 (rQM7) cells. During differentiation, however, myotube formation was dramatically repressed in MyoD KO QM7#4 cells. Moreover, myogenic differentiation-specific transcripts and proteins were not expressed in MyoD KO QM7#4 cells even after an extended differentiation period. These results indicate that MyoD is critical for muscle differentiation. Furthermore, we analyzed the global regulatory interactions by RNA sequencing during muscle differentiation. Conclusion: With CRISPR/Cas9-mediated genomic editing, single cell-derived sublines with a specific knockout gene can be adapted to various aspects of basic research as well as in functional genomics studies.
Keywords
Myoblast; CRISPR-Cas9; Knockout; Muscle Differentiation; MyoD;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Weber D, Wiese C, Gessler M. Hey bHLH transcription factors. Curr Top Dev Biol 2014;110:285-315.
2 Benezra R, Davis RL, Lockshon D, Turner DL, Weintraub H. The protein Id: a negative regulator of helix-loop-helix DNA binding proteins. Cell 1990;61:49-59.   DOI
3 Ruzinova MB, Benezra R. Id proteins in development, cell cycle and cancer. Trends Cell Biol 2003;13:410-8.   DOI
4 Perk J, Iavarone A, Benezra R. The Id family of helix-loop-helix proteins in cancer. Nat Rev Cancer 2005;5:603-14.   DOI
5 Kaplan FS, Xu M, Seemann P, et al. Classic and atypical fibrodysplasia ossificans progressiva (FOP) phenotypes are caused by mutations in the bone morphogenetic protein (BMP) type I receptor ACVR1. Hum Mutat 2009;30:379-90.   DOI
6 Hillier LW, Miller W, Birney E et al. Sequence and comparative analysis of the chicken genome provide unique perspectives on vertebrate evolution. Nature 2004;432:695-716.   DOI
7 Rubin CJ, Zody MC, Eriksson J, et al. Whole-genome resequencing reveals loci under selection during chicken domestication. Nature 2010;464:587-91.   DOI
8 Al-Musawi SL, Lock F, Simbi BH, Bayol SAM, Stickland NC. Muscle specific differences in the regulation of myogenic differentiation in chickens genetically selected for divergent growth rates. Differentiation 2011;82:127-35.   DOI
9 Shin S, Song Y, Ahn J, et al. A novel mechanism of myostatin regulation by its alternative splicing variant during myogenesis in avian species. Am J Physiol Cell Physiol 2015;309:650-9.   DOI
10 Park TS, Han JY. Genetic modification of chicken germ cells. Ann NY Acad Sci 2012;1271:104-9.   DOI
11 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.   DOI
12 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.   DOI
13 Salter DW, Smith EJ, Hughes SH, et al. Gene insertion into the chicken germ line by retroviruses. Poult Sci 1986;65:1445-8.   DOI
14 Love J, Gribbin C, Mather C, Sang H. Transgenic birds by DNA microinjection. Biotechnology 1994;12:60-3.
15 Davis RL, Weintraub H, Lassar AB. Expression of a single transfected cDNA converts fibroblasts to myoblasts. Cell 1987;51:987-1000.   DOI
16 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-E72.   DOI
17 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.   DOI
18 Dimitrov L, Pedersen D, Ching KH, et al. Germline gene editing in chickens by efficient CRISPR-mediated homologous recombination in primordial germ cells. PLoS ONE 2016;11:e0154303.   DOI
19 Cong L, Ran FA, Cox D, et al. Multiplex genome engineering using CRISPR/Cas systems. Science 2013;339:819-23.   DOI
20 Mali P, Yang L, Esvelt KM, et al. RNA-guided human genome engineering via Cas9. Science 2013;339:823-6.   DOI
21 Rudnicki MA, Braun T, Hinuma S, Jaenisch R. Inactivation of MyoD in mice leads to up-regulation of the myogenic HLH gene Myf-5 and results in apparently normal muscle development. Cell 1992;71:383-90.   DOI
22 Rudnicki MA, Schnegelsberg PNJ, Stead RH, et al. MyoD or Myf-5 is required for the formation of skeletal muscle. Cell 1993;75:1351-9.   DOI
23 Berkes CA, Tapscott SJ. MyoD and the transcriptional control of myogenesis. Semin Cell Dev Biol 2005;16:585-95.   DOI
24 Buckingham M. Myogenic progenitor cells and skeletal myogenesis in vertebrates. Curr Opin Genet Dev 2006;16:525-32.   DOI
25 Arnold HH, Braun T. Targeted inactivation of myogenic factor genes reveals their role during mouse myogenesis: a review. Int J Dev Biol 1996;40:345-53.
26 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.   DOI
27 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.   DOI
28 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.   DOI