Biopolym. Cell. 2019; 35(6): 427-436.
Molecular and Cell Biotechnologies
Generation of transgenic ducks by crispr/CAS9-mediated gene inser-tion combined with the sperm-mediated gene transfer (SMGT)
1, 3Konoval O., 5Korol P., 1Tabaka P., 4Kostenko S., 1Lizhi L., 4Chepiha A., 4Doroshenko M., 4Drahulian M., 1Xingchen B., 2Xuetao H., 2Liumeng L.
  1. Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences
    198 Shiqiao Road, Hangzhou, Zhejiang, China, 310021
  2. Zhuji Guowey Poultry Development Co, Ltd, Ltd.
    1, Gujing Road, Wangjiajing, Zhuji, Zhejiang, China, 311813
  3. Ukrainian Laboratory of Quality and Safety of Agricultural Products
    National University of Life and Environmental Sciences of Ukraine
    7, Mashynobudivnykiv Str., Chabany, Kyiv region, Ukraine, 08162
  4. National University of Life and Environmental Sciences of Ukraine
    15, Heroiv Oborony St., Kyiv, Ukraine,03041
  5. Institute of Animal Breeding and Genetics nd. a. M.V. Zubets of NAAS of Ukraine,
    1, Pohrebniaka Str., Chubyns’ke, Kyivs’ka obl., Ukraine, 083213

Abstract

Aim. To edit the duck genome by the methods of HDR-directed integration of the EGFP gene into the duck host genome in combination with SMGT using CRISPR / Cas9. Methods. The implementation of the HDR-mediated gene of green fluorescent protein (EGFP) was achieved by the combined action of four plasmids. The first pX330 plasmid contained the Cas9 gene. Two other plasmids contained sgRNA genes: pBR322-sgRNA1 and pBR322-sgRNA2. The fourth pBR322-HDR-EGFP plasmid was constructed to contain the DNA vector with left homologous sequence part(LHP), the EGFP gene coding sequences and the right homologous sequence part(RHP). The DNA sequence data for designing HDR-EGFP-insert and guide 20-nucleotide long parts of sgRNA 1 and sgRNA 2 were taken from the genome DNA sequense [sequence] data of «Anas platyrhynchos Spindlin 1 (SPIN1) gene. There were 24 ducks (13 males and 11 females) of the Shaoxing breed used for this experiment. The sperm transfection was performed using reagent Lipofectamine 2000. Results. As a result of these experiments 31 ducks were obtained, 19 of which carried the (F2) analysis revealed that in total 16 ducks (F1) (14 females and 2 males) transmitted the transgene DNA to their offsprings. The results showed that 27.6 % (56/203) of F2 descendants were positive for the transgene DNA construct. Conclusions. The results indicate that exogenous DNA was successfully inserted into the duck genome.
Keywords: CRISPR/Cas9, EGFP, SMGT, Transgenic poultry, Duck

References

[1] Davey MG, Tickle C. The chicken as a model for embryonic development. Cytogenet Genome Res. 2007;117(1-4):231-9.
[2] Collares T, Campos VF, De Leon PM, Cavalcanti PV, Amaral MG, Dellagostin OA, Deschamps JC, Seixas FK. Transgene transmission in chickens by sperm-mediated gene transfer after seminal plasma removal and exogenous DNA treated with dimethylsulfoxide or N,N-dimethylacetamide. J Biosci. 2011;36(4):613-20.
[3] Lillico SG, Sherman A, McGrew MJ, Robertson CD, Smith J, Haslam C, Barnard P, Radcliffe PA, Mitrophanous KA, Elliot EA, Sang HM. Oviduct-specific expression of two therapeutic proteins in transgenic hens. Proc Natl Acad Sci U S A. 2007;104(6):1771-6.
[4] Han JY. Germ cells and transgenesis in chickens. Comp Immunol Microbiol Infect Dis. 2009;32(2):61-80.
[5] Kwon MS, Koo BC, Kim D, Nam YH, Cui XS, Kim NH, Kim T. Generation of transgenic chickens expressing the human erythropoietin (hEPO) gene in an oviduct-specific manner: Production of transgenic chicken eggs containing human erythropoietin in egg whites. PLoS One. 2018;13(5):e0194721.
[6] Woodcock ME, Idoko-Akoh A, and McGrew MJ. Gene editing in birds takes flight. Mamm Genome, 2017; 28(7-8): 315–23.
[7] Mizuarai S, Ono K, Yamaguchi K, Nishijima K, Kamihira M, Iijima S. Production of transgenic quails with high frequency of germ-line transmission using VSV-G pseudotyped retroviral vector. Biochem Biophys Res Commun. 2001;286(3):456-63.
[8] Arthur J. Egg Innovations and Strategies for Improvements, Academic Pres (an imprint of Evsevier), 2017. 607 p.
[9] Kim YG, Cha J, Chandrasegaran S. Hybrid restriction enzymes: zinc finger fusions to Fok I cleavage domain. Proc Natl Acad Sci U S A. 1996;93(3):1156-60.
[10] Bedell VM, Wang Y, Campbell JM, Poshusta TL, Starker CG, Krug RG 2nd, Tan W, Penheiter SG, Ma AC, Leung AY, Fahrenkrug SC, Carlson DF, Voytas DF, Clark KJ, Essner JJ, Ekker SC. In vivo genome editing using a high-efficiency TALEN system. Nature. 2012;491(7422):114-8.
[11] Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, Hsu PD, Wu X, Jiang W, Marraffini LA, Zhang F. Multiplex genome engineering using CRISPR/Cas systems. Science. 2013;339(6121):819-23.
[12] Dimitrov L, Pedersen D, Ching KH, Yi H, Collarini EJ, Izquierdo S, van de Lavoir MC, Leighton PA. Germline Gene Editing in Chickens by Efficient CRISPR-Mediated Homologous Recombination in Primordial Germ Cells. PLoS One. 2016;11(4):e0154303.
[13] Salter DW, Smith EJ, Hughes SH, Wright SE, Fadly AM, Witter RL, Crittenden LB. Gene insertion into the chicken germ line by retroviruses. Poult Sci. 1986;65(8):1445-58.
[14] Thoraval P, Afanassieff M, Cosset FL, Lasserre F, Verdier G, Coudert F, Dambrine G. Germline transmission of exogenous genes in chickens using helper-free ecotropic avian leukosis virus-based vectors. Transgenic Res. 1995;4(6):369-77.
[15] Iba H. Gene transfer into chicken embryos by retrovirus vectors. Dev Growth Differ. 2000;42(3):213-8.
[16] Nakamura H, Funahashi J. Introduction of DNA into chick embryos by in ovo electroporation. Methods. 2001;24(1):43-8.
[17] Longmuir KJ, Haynes SM, Dickinson ME, Murphy JC, Willson RC, Waring AJ. Optimization of a peptide/non-cationic lipid gene delivery system for effective microinjection into chicken embryo in vivo. Mol Ther. 2001;4(1):66-74.
[18] Nakanishi A, Iritani A. Gene transfer in the chicken by sperm-mediated methods. Mol Reprod Dev. 1993;36(2):258-61.
[19] Rottmann OJ, Antes R, Höfer P, Maierhofer G. Liposome mediated gene transfer via spermatozoa into avian egg cells. J Anim Breed Genet. 1992; 109(1-6): 64–70.
[20] Yang CC, Chang HS, Lin C-J, Hsu CC, Cheung JI, Hwu L, Cheng Winston. Cock spermatozoa serve as the gene vector for generation of transgenic chicken (Gallus gallus). Asian-Australas J Anim Sci. 2004; 17: 885-91.
[21] Harel-Markowitz E, Gurevich M, Shore LS, Katz A, Stram Y, Shemesh M. Use of sperm plasmid DNA lipofection combined with REMI (restriction enzyme-mediated insertion) for production of transgenic chickens expressing eGFP (enhanced green fluorescent protein) or human follicle-stimulating hormone. Biol Reprod. 2009;80(5):1046-52.
[22] Samoilov A, Martirosian V, Baryshnikov A, Surayeva N. The usage linker of based chicken sperm-mediated gene transfer for development and study of methods of therapeutic proteins production. Ross Bioterapevt Z. 2011; 11(3): 53–6.
[23] El-Gendy EA, Ahmed MM, El-Tantawy SM. Development of transgenic chickens by sperm-mediated gene transfer. Afr J Biotechnol. 2013; 12: 2755-63.
[24] Samoylov AV, Kesyan AZ, Suraeva NM. Development of transgenic chicken with a gene of human granulocyte colony-stimulating factor using sperm-mediated gene transfer. Biology Bulletin. 2013; 40(5): 419–22.
[25] Li B, Sun G, Sun H, Xu Q, Gao B, Zhou G, Zhao W, Wu X, Bao W, Yu F, Wang K, Chen G. Efficient generation of transgenic chickens using the spermatogonial stem cells in vivo and ex vivo transfection. Sci China C Life Sci. 2008;51(8):734-42.
[26] Min S, Qing S, Hui Y, Zhi FD, Rong QY, Feng X, Chun BL Generation of antiviral transgenic chicken using spermatogonial stem cell transfected in vivo. Afr J Biotechnol. 2012; 11(6): 1509-15.
[27] Watanabe M, Sugimori Y. Studies on the artificial insemination in ducks. Zootecnica e Veterinaria, Anno XII. 1957; 3: 119-24.
[28] Onishi N, Kato Y, Futamura K. Studies on the artificial insemination in ducks. Bull Nat Inst Agric Sci, Series G. 1955; 11: 2-16.
[29] Mehravar M, Shirazi A, Nazari M, Banan M. Mosaicism in CRISPR/Cas9-mediated genome editing. Dev Biol. 2019;445(2):156-162.
[30] Zhang Z, Sun P, Yu F, Yan L, Yuan F, Zhang W, Wang T, Wan Z, Shao Q, Li Z. Transgenic quail production by microinjection of lentiviral vector into the early embryo blood vessels. PLoS One. 2012;7(12):e50817.
[31] Scott BB, Lois C. Generation of tissue-specific transgenic birds with lentiviral vectors. Proc Natl Acad Sci U S A. 2005;102(45):16443-7.
[32] Midic U, Hung PH, Vincent KA, Goheen B, Schupp PG, Chen DD, Bauer DE, VandeVoort CA, Latham KE. Quantitative assessment of timing, efficiency, specificity and genetic mosaicism of CRISPR/Cas9-mediated gene editing of hemoglobin beta gene in rhesus monkey embryos. Hum Mol Genet. 2017;26(14):2678-2689.
[33] Tu Z, Yang W, Yan S, Yin A, Gao J, Liu X, Zheng Y, Zheng J, Li Z, Yang S, Li S, Guo X, Li XJ. Promoting Cas9 degradation reduces mosaic mutations in non-human primate embryos. Sci Rep. 2017;7:42081.
[34] Niu Y, Shen B, Cui Y, Chen Y, Wang J, Wang L, Kang Y, Zhao X, Si W, Li W, Xiang AP, Zhou J, Guo X, Bi Y, Si C, Hu B, Dong G, Wang H, Zhou Z, Li T, Tan T, Pu X, Wang F, Ji S, Zhou Q, Huang X, Ji W, Sha J. Generation of gene-modified cynomolgus monkey via Cas9/RNA-mediated gene targeting in one-cell embryos. Cell. 2014;156(4):836-43.
[35] Chen Y, Zheng Y, Kang Y, Yang W, Niu Y, Guo X, Tu Z, Si C, Wang H, Xing R, Pu X, Yang SH, Li S, Ji W, Li XJ. Functional disruption of the dystrophin gene in rhesus monkey using CRISPR/Cas9. Hum Mol Genet. 2015;24(13):3764-74.