J Vet Sci. 2024 Jan;25(1):e10. English.
Published online Oct 05, 2023.
© 2024 The Korean Society of Veterinary Science
Original Article

Outlook on genome editing application to cattle

Gyeong-Min Gim,1 and Goo Jang1,2,3,4
    • 1LARTBio Inco, Seoul 06221, Korea.
    • 2Laboratory of Theriogenology and Biotechnology, Department of Veterinary Clinical Science, College of Veterinary Medicine and the Research Institute of Veterinary Science, Seoul National University, Seoul 08826, Korea.
    • 3Comparative medicine Disease Research Center, Seoul National University, Seoul 08826, Korea.
    • 4Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya 60115, Indonesia.
Received May 15, 2023; Revised August 04, 2023; Accepted August 20, 2023.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (https://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

In livestock industry, there is growing interest in methods to increase the production efficiency of livestock to address food shortages, given the increasing global population. With the advancements in gene engineering technology, it is a valuable tool and has been intensively utilized in research specifically focused on human disease. In historically, this technology has been used with livestock to create human disease models or to produce recombinant proteins from their byproducts. However, in recent years, utilizing gene editing technology, cattle with identified genes related to productivity can be edited, thereby enhancing productivity in response to climate change or specific disease instead of producing recombinant proteins. Furthermore, with the advancement in the efficiency of gene editing, it has become possible to edit multiple genes simultaneously. This cattle breed improvement has been achieved by discovering the genes through the comprehensive analysis of the entire genome of cattle. The cattle industry has been able to address gene bottlenecks that were previously impossible through conventional breeding systems. This review concludes that gene editing is necessary to expand the cattle industry, improving productivity in the future. Additionally, the enhancement of cattle through gene editing is expected to contribute to addressing environmental challenges associated with the cattle industry. Further research and development in gene editing, coupled with genomic analysis technologies, will significantly contribute to solving issues that conventional breeding systems have not been able to address.

Keywords
Breeding; cattle; genetic engineering; genetically engineering cattle; somatic cell nuclear transfer

INTRODUCTION

Since the 1990s, genetically engineered cattle (GEC) have been produced using several techniques, including deletion and insertion, random integration into the host genome, and selective genome editing [1]. Initially, GEC had been used to produce recombinant pharmaceuticals and understanding human diseases. With increasing global population, we will face into food shortages in 2025. It is necessary to solve the food problem and need to improve productivity in cattle. Coupling with genetic engineering and analysis tools, such as next-generation sequencing (NGS) to figure out the gene related to productivity, can maximize productive efficiency more than the conventional breeding system.

Of the genetic engineering tools currently available, genome editing is remarkably powerful to edit bovine endogenous genes. The development of various genome editing tools, such as zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9, has made genome editing highly convenient and precise. Through the technologies, the cattle with improvement of disease resistance, and enhancement of the frequency of alleles or polymorphisms that are associated with favorable traits (e.g., lack of horns, heat tolerance, and efficient milk or meat production/composition) were produced [2, 3, 4, 5, 6]. In addition, gene editing can overcome the challenge of random integration into the host genome, which cause several risks. This review introduces the need of gene editing technology in cattle industry, explaining a general background of gene editing technology and production of cattle with superior traits using gene editing and gene analysis. In addition, our review showed DNA-based genome selection for improving cattle breeds, which involves analyzing the entire cattle genome to identify genetic markers associated with desirable traits, is more accurate and can improve the speed of genetic progress [7].

PAST TECHNOLOGIES FOR PRODUCING GEC

To date, most GEC have been produced through microinjection and somatic cell nuclear transfer (SCNT). Before the development of SCNT in the 1990s, most GEC were generated through microinjection, in which nucleic acids are microinjected into the cytoplasm of fertilized eggs (zygote). In the 1990s, GEC were produced by microinjecting plasmids or viruses carrying the gene of interest. Although the trials were successful, birth rates of GEC were very low, at about 12% [8, 9, 10]. Moreover, microinjection resulted in genetic mosaicism and was unsuitable for use on farm animals because of its high cost, long gestation period, small litter sizes, and low transgene integration rates (Fig. 1) [9].

Fig. 1
Representative techniques for the generation of genetically engineered cattle. SCNT is inefficient at generating healthy calves, while MI is time-consuming because of the emergence of mosaicism.
SCNT, somatic cell nuclear transfer; MI, microinjection.

Since the 2000s, SCNT has been the most powerful tool for GEC production. This method involves the selection of a donor cell with a desired mutation. Subsequently, the nucleus of the donor cell is replaced with that of a mature oocyte following which the embryo is transplanted (Fig. 1). SCNT led to the development of GEC that express recombinant proteins in milk [2, 3] and are disease resistant [4, 5], and also improved animal welfare [6]. However, SCNT-derived calves have numerous health complications because of abnormal reprogramming and epigenetic gene regulation [11, 12]. Although several efforts have been made [13, 14, 15, 16, 17, 18, 19] to address these limitations, some issues remain. Nonetheless, SCNT has several advantages, including easy selection of donor cells that have the desired gene or mutation, analysis when testing in cells, and selection of cells exhibiting high transgene expression. Thus, before the development of genome-editing tools (ZFNs, TALENs, and CRISPR/Cas9), SCNT was preferred over microinjection.

Before the development of genome-editing tools, GEC were developed through the random insertion of the gene of interest into the host genome (Table 1). However, random insertion has several potential drawbacks [13, 14]. First, the targeted host gene insertion locus can only be known after analyzing the transgenic animals using PCR and DNA sequencing. Second, gene insertion can alter the expression of endogenous genes. Third, several copies of the gene can be inserted into the host genome.

GEC PRODUCTION USING GENOME-EDITING TOOLS (ZFNs, TALENs, AND CRISPR/Cas9)

Before the development of targeted genome-editing methods, the genome was modified randomly, which was ineffective and expensive for the development of GEC. In addition, there were safety concerns about malfunctioning original genes or effects on gene regulation [13, 14]. However, the emergence of endonucleases as genome-editing tools helped significantly overcome these challenges. The endonucleases that are used for genome editing, including ZFNs, TALENs, and CRISPR/Cas9, recognize specific target gene sequences and induce double-strand breaks (DSBs), which trigger DNA repair via non-homologous end-joining (NHEJ) or homology-directed repair (HDR) [15], thereby markedly increasing the efficacy of targeted genetic modifications [16].

Because of its ability to accurately edit target loci, genome editing leads to quicker and more effective livestock improvements when compared with conventional breeding methods (Fig. 2). However, the long-term effects of genome editing are not fully understood and there are concerns that although it is less risky than conventional genetic engineering methods, it may endanger animal welfare. Barring some minor issues, current genome-editing tools allow quick investigation of superior traits, such as productivity, disease resistance, and resilience to climate change.

Fig. 2
Breeding methods: conventional breeding vs. genome editing. Although conventional breeding, which is based on observable traits has been used for centuries, it is time-consuming because it requires multiple animal generations. Modern breeding techniques that use genome editing can introduce new genetic traits that are impossible to transfer through conventional breeding. However, this requires a complete understanding of the long-term effects of genome editing and raises ethical and safety concerns.

ZFNs

ZFNs were discovered in the 1980s and the first specific one was reported 15 years later. A ZFN is comprised of 30 amino acids, with two anti-parallel beta-sheets opposing an alpha-helix [17]. The alpha-helix can bind three specific bases located in the major groove of DNA [18]. ZFNs have a site-specific zinc-finger DNA-binding domain and nonspecific FokI endonuclease cleavage domain. Two or more ZFN molecules are required to modify a specific gene. In general, ZFNs recognize 9–18 bp that can be specifically edited. Dimerized FokI induces DSBs, which trigger NHEJ, through which a specific gene can be blocked, or HDR, through which a desired DNA sequence can be inserted into a specific gene. Originally, the lengths of homologous arms were 6–7 kb but decreased to 0.5–1.5 kb with the emergence of ZFNs [19]. Several GEC produced by ZFN are listed in Table 2. Notable among these was the successful mutation of the beta-lactoglobulin (BLG) gene in cattle [20]. BLG protein is not present in human milk but is present in significant quantities in whey and is an allergen in cow milk. Importantly, the BLG gene modification was stably inherited by subsequent generations. Interestingly, milk obtained from the BLG-mutated cows was found to be digested by pepsin approximately 30 times faster than normal milk containing BLG [21].

Table 2
GEC lists produced by Zinc-finger nuclease

TALENs

TALENs are naturally produced by Xanthomonas, a genus of gram-negative bacteria that infect several plant species. TALENs are comprised of DNA-binding domains with 33–35 amino acid repeat domains and the nonspecific endonuclease, FokI. The repeat variable di-residue (RVD), the amino acid residues on the 12th and 13th positions of the DNA-binding domains, recognize single base pairs, with each RVD specifically binding a single genomic DNA nucleotide [22, 23]. The DNA-binding sequence of TALENs begins with thymidine and the target sequence length is 30–40 bp. The DNA-binding domains can be modified to target endogenous DNA sequences for cleavage by the nonspecific endonuclease, FokI, thereby triggering DNA repair via NHEJ and HDR [20, 24, 25, 26].

TALENs have several advantages over ZFNs: 1) the TALEN repeat is 3–4 times longer than the ZFN repeat and because TALENs recognize one nucleotide, they are more sophisticated than ZFNs, which recognize three nucleotides, 2) the ZFN modification requires a high-level design because DNA recognition may not be successful because of crosstalk between the fingers [27], 3) TALENs are easier to design than ZFNs because they are simpler and their production is quicker and more cost-effective, 4) TALENs have less off-target effects than ZFNs, and 5) compared with ZFNs, TALENs are more amenable to genome-editing because they can be injected into the cytoplasm of livestock embryos [28].

TALENs are a powerful tool for knock out genes in rats and zebrafish [77, 78, 79] but they have also been used to efficiently mutate genes of interest in cattle, sheep, and pigs [29, 30]. Table 3 summarizes the history of GEC production using TALENs, which was used to develop hornless cattle via the introduction of the Pc Celtic POLLED allele into dairy bulls [6]. Recently, crossing these bulls with horned cows resulted in the birth of six hornless calves and genome analysis confirmed that the calves possess the Pc Celtic POLLED allele without any unintended genomic alterations [31].

CRISPR/Cas9

CRISPR, which was discovered in the 1980s, provides adaptive immunity that protects bacteria and archaea from bacteriophage invasion [32, 33, 34, 35]. CRISPR mediates RNA-guided DNA cleavage. The CRISPR/Cas9 system is widely used to induce targeted DSBs. It contains a Cas9 endonuclease that causes DSBs after recognizing the protospacer adjacent motif downstream of a target sequence and a single-guide RNA that interacts with and directs Cas9 to the target DNA sequence [84]. The CRISPR/Cas9 system is much more efficient at targeting genes of interest than the older gene editing tools (ZFNs and TALENs). Moreover, unlike ZFNs and TALENs, which form DNA-protein complexes, CRISPR/Cas9 forms an RNA-protein complex (Table 4). Importantly, CRISPR/Cas9 can simultaneously induce DSBs in more than one target gene. CRISPR/Cas9 has also been used to generate gene-edited farm animals [36, 37]. In addition, a one-step method of generating gene-edited animals was developed by introducing Cas9 messenger RNA (mRNA) into early zebrafish [38, 39, 40], rats [40, 41], mice [42, 43, 44, 45], rabbits [46], pigs [37, 47, 48], sheep, and cattle embryos [29, 49]. The adoption of CRISPR/Cas9 as a gene-editing tool has improved the efficiency of gene editing in mammals and allowed the generation of highly sophisticated genetically engineered animals [42, 50, 51].

Table 4
Comparison of three classes of molecular scissors [84]

Although CRISPR/Cas9 has greatly increased the efficiency of generating genetically engineered animals, there are still limitations to the insertion of foreign genes. For instance, inserting foreign genes by HDR is challenging [52, 53, 54], especially when large cargoes that require extensive cell sorting or selection are involved [55]. Several techniques, including the utilization of long single-stranded DNA [56], homology-independent targeted insertion [57, 58], homology-mediated end-joining (HMEJ) [52, 59], microhomology-mediated end joining (MMEJ) [60], and targeted integration with linearized double strand DNA [61], have been used to enhance knock-in efficiency. However, further studies are required to improve knock-in efficiency in mammals. The history of CRISPR/Cas9-generated GEC is summarized in Table 5. One of the most recent studies on the generation of GEC using CRISPR/Cas9 involved the targeted editing of the MSTN gene, which doubled the muscle mass [62]. It has been confirmed that the cattle can transmit the MSTN mutation to the next generation, indicating stable germline transmission of the trait [63].

GENOMIC SELECTION

The recent increase in CRISPR/Cas9 use has led to the production of specific gene-targeted cattle [62, 64]. Even in cattle, germline transmission of the specific gene mutation was proven. The increasing popularity of specific gene targeting has led to increased research on the genomic selection of single nucleotide polymorphisms. In the case of DNA-based genomic selection, the improvement rate is relatively objective compared with that of the existing method, which is based on phenotypic selection and cannot always be a reliable indicator because of the influence of environmental factors (Fig. 2). In addition, the use of genomic selection has advanced cattle breeding by enabling breeders to identify and select animals with superior genetic traits at a younger age, which increases efficiency and productivity in the cattle industry.

DISCUSSION

Cattle are known to be a major source of animal protein in the world. With the increasing global population, enhancing cattle production efficiency is crucial. While conventional breeding systems have improved cattle breeds, gene editing technology can create new cattle breeds with higher productivity that conventional breed system cannot achieve. In addition, advances in genetic analysis technology have led to a shift from phenotype-based selective breeding to DNA-based selective breeding. The combination of genomic selection and genome editing can enhance superior traits more efficiently and with high accuracy (Fig. 3), especially in areas that have traditionally required prolonged periods for improvement. Further development of new breeding systems will contribute to solve the problems that conventional breeding systems have previously been unable to address.

Fig. 3
An improved breeding system for the future. (A) Cattle generated using an observable traits-based breeding system. (B) Based on DNA sequences, genetic marker analysis can detect superior traits that cannot be influenced by the environment. (C) When compared with conventional breeding techniques based on physical characteristics, genetic markers can quickly and accurately identify cattle with superior production traits and disease resistance. (D) Genome editing can produce genetically engineered cattle with new genetic traits that cannot be found in existing breeds.

Notes

Funding:This study was financially supported by NRF-2021R1A5A1 033157 for SRC program: 382 Comparative medicine Disease Research Center, the Research Institute of Veterinary Science, the BK21 Four for Future Veterinary Medicine Leading Education and Research Center, and a Seoul National University (SNU) grant (#550-20220044).

Conflict of Interest:The authors declare no conflicts of interest.

Author Contributions:

  • Conceptualization: Gim GM.

  • Data curation: Gim GM.

  • Funding acquisition: Jang G.

  • Project administration: Jang G.

  • Supervision: Jang G.

  • Writing - original draft: Gim GM.

  • Writing - review & editing: Jang G.

References

    1. Yum SY, Youn KY, Choi WJ, Jang G. Development of genome engineering technologies in cattle: from random to specific. J Anim Sci Biotechnol 2018;9:16.
    1. Salamone D, Barañao L, Santos C, Bussmann L, Artuso J, Werning C, et al. High level expression of bioactive recombinant human growth hormone in the milk of a cloned transgenic cow. J Biotechnol 2006;124(2):469–472.
    1. Johnson TE, Nelson GA. Caenorhabditis elegans: a model system for space biology studies. Exp Gerontol 1991;26(2-3):299–309.
    1. Wall RJ, Powell AM, Paape MJ, Kerr DE, Bannerman DD, Pursel VG, et al. Genetically enhanced cows resist intramammary Staphylococcus aureus infection. Nat Biotechnol 2005;23(4):445–451.
    1. Richt JA, Kasinathan P, Hamir AN, Castilla J, Sathiyaseelan T, Vargas F, et al. Production of cattle lacking prion protein. Nat Biotechnol 2007;25(1):132–138.
    1. Carlson DF, Lancto CA, Zang B, Kim ES, Walton M, Oldeschulte D, et al. Production of hornless dairy cattle from genome-edited cell lines. Nat Biotechnol 2016;34(5):479–481.
    1. de Almeida Camargo LS, Pereira JF. Genome-editing opportunities to enhance cattle productivity in the tropics. CABI Agric Biosci 2022;3(1):8.
    1. Krimpenfort P, Rademakers A, Eyestone W, van der Schans A, van den Broek S, Kooiman P, et al. Generation of transgenic dairy cattle using ‘in vitro’ embryo production. Biotechnology (N Y) 1991;9(9):844–847.
    1. Hyttinen JM, Peura T, Tolvanen M, Aalto J, Alhonen L, Sinervirta R, et al. Generation of transgenic dairy cattle from transgene-analyzed and sexed embryos produced in vitro. Biotechnology (N Y) 1994;12(6):606–608.
    1. Eyestone WH. Production and breeding of transgenic cattle using in vitro embryo production technology. Theriogenology 1999;51(2):509–517.
    1. Kubota C, Yamakuchi H, Todoroki J, Mizoshita K, Tabara N, Barber M, et al. Six cloned calves produced from adult fibroblast cells after long-term culture. Proc Natl Acad Sci U S A 2000;97(3):990–995.
    1. Zhao J, Hao Y, Ross JW, Spate LD, Walters EM, Samuel MS, et al. Histone deacetylase inhibitors improve in vitro and in vivo developmental competence of somatic cell nuclear transfer porcine embryos. Cell Reprogram 2010;12(1):75–83.
    1. Hacein-Bey-Abina S, Von Kalle C, Schmidt M, McCormack MP, Wulffraat N, Leboulch P, et al. LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science 2003;302(5644):415–419.
    1. Bushman F, Lewinski M, Ciuffi A, Barr S, Leipzig J, Hannenhalli S, et al. Genome-wide analysis of retroviral DNA integration. Nat Rev Microbiol 2005;3(11):848–858.
    1. Wyman C, Kanaar R. DNA double-strand break repair: all's well that ends well. Annu Rev Genet 2006;40:363–383.
    1. Rouet P, Smih F, Jasin M. Expression of a site-specific endonuclease stimulates homologous recombination in mammalian cells. Proc Natl Acad Sci U S A 1994;91(13):6064–6068.
    1. Pabo CO, Peisach E, Grant RA. Design and selection of novel Cys2His2 zinc finger proteins. Annu Rev Biochem 2001;70(1):313–340.
    1. Pavletich NP, Pabo CO. Zinc finger-DNA recognition: crystal structure of a Zif268-DNA complex at 2.1 A. Science 1991;252(5007):809–817.
    1. Orlando SJ, Santiago Y, DeKelver RC, Freyvert Y, Boydston EA, Moehle EA, et al. Zinc-finger nuclease-driven targeted integration into mammalian genomes using donors with limited chromosomal homology. Nucleic Acids Res 2010;38(15):e152
    1. Yu S, Luo J, Song Z, Ding F, Dai Y, Li N. Highly efficient modification of beta-lactoglobulin (BLG) gene via zinc-finger nucleases in cattle. Cell Res 2011;21(11):1638–1640.
    1. Sun Z, Wang M, Han S, Ma S, Zou Z, Ding F, et al. Production of hypoallergenic milk from DNA-free beta-lactoglobulin (BLG) gene knockout cow using zinc-finger nucleases mRNA. Sci Rep 2018;8(1):15430.
    1. Boch J, Scholze H, Schornack S, Landgraf A, Hahn S, Kay S, et al. Breaking the code of DNA binding specificity of TAL-type III effectors. Science 2009;326(5959):1509–1512.
    1. Moscou MJ, Bogdanove AJ. A simple cipher governs DNA recognition by TAL effectors. Science 2009;326(5959):1501.
    1. Miller JC, Tan S, Qiao G, Barlow KA, Wang J, Xia DF, et al. A TALE nuclease architecture for efficient genome editing. Nat Biotechnol 2011;29(2):143–148.
    1. Mahfouz MM, Li L, Shamimuzzaman M, Wibowo A, Fang X, Zhu JK. De novo-engineered transcription activator-like effector (TALE) hybrid nuclease with novel DNA binding specificity creates double-strand breaks. Proc Natl Acad Sci U S A 2011;108(6):2623–2628.
    1. Litos IK, Ioannou PC, Christopoulos TK, Tzetis M, Kanavakis E, Traeger-Synodinos J. Quadruple-allele dipstick test for simultaneous visual genotyping of A896G (Asp299Gly) and C1196T (Thr399Ile) polymorphisms in the toll-like receptor-4 gene. Clin Chim Acta 2011;412(21-22):1968–1972.
    1. Move over ZFNs. Nat Biotechnol 2011;29(8):681–684.
    1. Khan SH. Genome-editing technologies: concept, pros, and cons of various genome-editing techniques and bioethical concerns for clinical application. Mol Ther Nucleic Acids 2019;16:326–334.
    1. Proudfoot C, Carlson DF, Huddart R, Long CR, Pryor JH, King TJ, et al. Genome edited sheep and cattle. Transgenic Res 2015;24(1):147–153.
    1. Carlson DF, Tan W, Lillico SG, Stverakova D, Proudfoot C, Christian M, et al. Efficient TALEN-mediated gene knockout in livestock. Proc Natl Acad Sci U S A 2012;109(43):17382–17387.
    1. Young AE, Mansour TA, McNabb BR, Owen JR, Trott JF, Brown CT, et al. Genomic and phenotypic analyses of six offspring of a genome-edited hornless bull. Nat Biotechnol 2020;38(2):225–232.
    1. Wiedenheft B, Sternberg SH, Doudna JA. RNA-guided genetic silencing systems in bacteria and archaea. Nature 2012;482(7385):331–338.
    1. Mojica FJM, Díez-Villaseñor C, García-Martínez J, Soria E. Intervening sequences of regularly spaced prokaryotic repeats derive from foreign genetic elements. J Mol Evol 2005;60(2):174–182.
    1. Jansen R, Embden JD, Gaastra W, Schouls LM. Identification of genes that are associated with DNA repeats in prokaryotes. Mol Microbiol 2002;43(6):1565–1575.
    1. Doudna JA, Charpentier E. Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science 2014;346(6213):1258096
    1. Jinek M, East A, Cheng A, Lin S, Ma E, Doudna J. RNA-programmed genome editing in human cells. Elife 2013;2:e00471
    1. 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(3):372–375.
    1. Meng X, Noyes MB, Zhu LJ, Lawson ND, Wolfe SA. Targeted gene inactivation in zebrafish using engineered zinc-finger nucleases. Nat Biotechnol 2008;26(6):695–701.
    1. Doyon Y, McCammon JM, Miller JC, Faraji F, Ngo C, Katibah GE, et al. Heritable targeted gene disruption in zebrafish using designed zinc-finger nucleases. Nat Biotechnol 2008;26(6):702–708.
    1. Geurts AM, Cost GJ, Freyvert Y, Zeitler B, Miller JC, Choi VM, et al. Knockout rats via embryo microinjection of zinc-finger nucleases. Science 2009;325(5939):433.
    1. Cui X, Ji D, Fisher DA, Wu Y, Briner DM, Weinstein EJ. Targeted integration in rat and mouse embryos with zinc-finger nucleases. Nat Biotechnol 2011;29(1):64–67.
    1. Wang H, Yang H, Shivalila CS, Dawlaty MM, Cheng AW, Zhang F, et al. One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. Cell 2013;153(4):910–918.
    1. Meyer M, de Angelis MH, Wurst W, Kühn R. Gene targeting by homologous recombination in mouse zygotes mediated by zinc-finger nucleases. Proc Natl Acad Sci U S A 2010;107(34):15022–15026.
    1. Kurome M, Geistlinger L, Kessler B, Zakhartchenko V, Klymiuk N, Wuensch A, et al. Factors influencing the efficiency of generating genetically engineered pigs by nuclear transfer: multi-factorial analysis of a large data set. BMC Biotechnol 2013;13:43.
    1. Christian M, Cermak T, Doyle EL, Schmidt C, Zhang F, Hummel A, et al. Targeting DNA double-strand breaks with TAL effector nucleases. Genetics 2010;186(2):757–761.
    1. Flisikowska T, Thorey IS, Offner S, Ros F, Lifke V, Zeitler B, et al. Efficient immunoglobulin gene disruption and targeted replacement in rabbit using zinc finger nucleases. PLoS One 2011;6(6):e21045
    1. Yang H, Wang H, Shivalila CS, Cheng AW, Shi L, Jaenisch R. One-step generation of mice carrying reporter and conditional alleles by CRISPR/Cas-mediated genome engineering. Cell 2013;154(6):1370–1379.
    1. Whitworth KM, Lee K, Benne JA, Beaton BP, Spate LD, Murphy SL, et al. Use of the CRISPR/Cas9 system to produce genetically engineered pigs from in vitro-derived oocytes and embryos. Biol Reprod 2014;91(3):78.
    1. Lillico SG, Proudfoot C, Carlson DF, Stverakova D, Neil C, Blain C, et al. Live pigs produced from genome edited zygotes. Sci Rep 2013;3:2847.
    1. Sander JD, Joung JK. CRISPR-Cas systems for editing, regulating and targeting genomes. Nat Biotechnol 2014;32(4):347–355.
    1. Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, et al. Multiplex genome engineering using CRISPR/Cas systems. Science 2013;339(6121):819–823.
    1. Zhang JP, Li XL, Li GH, Chen W, Arakaki C, Botimer GD, et al. Efficient precise knockin with a double cut HDR donor after CRISPR/Cas9-mediated double-stranded DNA cleavage. Genome Biol 2017;18(1):35.
    1. Davis L, Maizels N. Homology-directed repair of DNA nicks via pathways distinct from canonical double-strand break repair. Proc Natl Acad Sci U S A 2014;111(10):E924–E932.
    1. Chu VT, Weber T, Wefers B, Wurst W, Sander S, Rajewsky K, et al. Increasing the efficiency of homology-directed repair for CRISPR-Cas9-induced precise gene editing in mammalian cells. Nat Biotechnol 2015;33(5):543–548.
    1. Byrne SM, Ortiz L, Mali P, Aach J, Church GM. Multi-kilobase homozygous targeted gene replacement in human induced pluripotent stem cells. Nucleic Acids Res 2015;43(3):e21
    1. Quadros RM, Miura H, Harms DW, Akatsuka H, Sato T, Aida T, et al. Easi-CRISPR: a robust method for one-step generation of mice carrying conditional and insertion alleles using long ssDNA donors and CRISPR ribonucleoproteins. Genome Biol 2017;18(1):92.
    1. Suzuki K, Izpisua Belmonte JC. In vivo genome editing via the HITI method as a tool for gene therapy. J Hum Genet 2018;63(2):157–164.
    1. Auer TO, Duroure K, De Cian A, Concordet JP, Del Bene F. Highly efficient CRISPR/Cas9-mediated knock-in in zebrafish by homology-independent DNA repair. Genome Res 2014;24(1):142–153.
    1. Yao X, Wang X, Hu X, Liu Z, Liu J, Zhou H, et al. Homology-mediated end joining-based targeted integration using CRISPR/Cas9. Cell Res 2017;27(6):801–814.
    1. Nakade S, Tsubota T, Sakane Y, Kume S, Sakamoto N, Obara M, et al. Microhomology-mediated end-joining-dependent integration of donor DNA in cells and animals using TALENs and CRISPR/Cas9. Nat Commun 2014;5:5560.
    1. Yao X, Zhang M, Wang X, Ying W, Hu X, Dai P, et al. Tild-CRISPR allows for efficient and precise gene knockin in mouse and human cells. Dev Cell 2018;45(4):526–536.e5.
    1. Gim GM, Kwon DH, Eom KH, Moon J, Park JH, Lee WW, et al. Production of MSTN-mutated cattle without exogenous gene integration using CRISPR-Cas9. Biotechnol J 2022;17(7):e2100198
    1. Gim GM, Uhm KH, Kwon DH, Kim MJ, Jung DJ, Kim DH, et al. Germline transmission of MSTN knockout cattle via CRISPR-Cas9. Theriogenology 2022;192:22–27.
    1. Owen JR, Hennig SL, McNabb BR, Mansour TA, Smith JM, Lin JC, et al. One-step generation of a targeted knock-in calf using the CRISPR-Cas9 system in bovine zygotes. BMC Genomics 2021;22(1):118.
    1. Chan AW, Homan EJ, Ballou LU, Burns JC, Bremel RD. Transgenic cattle produced by reverse-transcribed gene transfer in oocytes. Proc Natl Acad Sci U S A 1998;95(24):14028–14033.
    1. van Berkel PH, Welling MM, Geerts M, van Veen HA, Ravensbergen B, Salaheddine M, et al. Large scale production of recombinant human lactoferrin in the milk of transgenic cows. Nat Biotechnol 2002;20(5):484–487.
    1. Brophy B, Smolenski G, Wheeler T, Wells D, L’Huillier P, Laible G. Cloned transgenic cattle produce milk with higher levels of beta-casein and kappa-casein. Nat Biotechnol 2003;21(2):157–162.
    1. Hofmann A, Zakhartchenko V, Weppert M, Sebald H, Wenigerkind H, Brem G, et al. Generation of transgenic cattle by lentiviral gene transfer into oocytes. Biol Reprod 2004;71(2):405–409.
    1. Grosse-Hovest L, Müller S, Minoia R, Wolf E, Zakhartchenko V, Wenigerkind H, et al. Cloned transgenic farm animals produce a bispecific antibody for T cell-mediated tumor cell killing. Proc Natl Acad Sci U S A 2004;101(18):6858–6863.
    1. Yang P, Wang J, Gong G, Sun X, Zhang R, Du Z, et al. Cattle mammary bioreactor generated by a novel procedure of transgenic cloning for large-scale production of functional human lactoferrin. PLoS One 2008;3(10):e3453
    1. Echelard Y, Williams JL, Destrempes MM, Koster JA, Overton SA, Pollock DP, et al. Production of recombinant albumin by a herd of cloned transgenic cattle. Transgenic Res 2009;18(3):361–376.
    1. Kuroiwa Y, Kasinathan P, Sathiyaseelan T, Jiao JA, Matsushita H, Sathiyaseelan J, et al. Antigen-specific human polyclonal antibodies from hyperimmunized cattle. Nat Biotechnol 2009;27(2):173–181.
    1. Wongsrikeao P, Sutou S, Kunishi M, Dong YJ, Bai X, Otoi T. Combination of the somatic cell nuclear transfer method and RNAi technology for the production of a prion gene-knockdown calf using plasmid vectors harboring the U6 or tRNA promoter. Prion 2011;5(1):39–46.
    1. Jabed A, Wagner S, McCracken J, Wells DN, Laible G. Targeted microRNA expression in dairy cattle directs production of β-lactoglobulin-free, high-casein milk. Proc Natl Acad Sci U S A 2012;109(42):16811–16816.
    1. Garrels W, Talluri TR, Apfelbaum R, Carratala YP, Bosch P, Potzsch K, et al. One-step multiplex transgenesis via sleeping beauty transposition in cattle. Sci Rep 2016;6:21953.
    1. Su F, Wang Y, Liu G, Ru K, Liu X, Yu Y, et al. Generation of transgenic cattle expressing human β-defensin 3 as an approach to reducing susceptibility to Mycobacterium bovis infection. FEBS J 2016;283(5):776–790.
    1. Yum SY, Lee SJ, Kim HM, Choi WJ, Park JH, Lee WW, et al. Efficient generation of transgenic cattle using the DNA transposon and their analysis by next-generation sequencing. Sci Rep 2016;6:27185.
    1. Wang Y, Ding F, Wang T, Liu W, Lindquist S, Hernell O, et al. Purification and characterization of recombinant human bile salt-stimulated lipase expressed in milk of transgenic cloned cows. PLoS One 2017;12(5):e0176864
    1. Wang M, Sun Z, Yu T, Ding F, Li L, Wang X, et al. Large-scale production of recombinant human lactoferrin from high-expression, marker-free transgenic cloned cows. Sci Rep 2017;7(1):10733.
    1. Liu X, Wang Y, Tian Y, Yu Y, Gao M, Hu G, et al. Generation of mastitis resistance in cows by targeting human lysozyme gene to β-casein locus using zinc-finger nucleases. Proc Biol Sci 2014;281(1780):20133368
    1. Luo J, Song Z, Yu S, Cui D, Wang B, Ding F, et al. Efficient generation of myostatin (MSTN) biallelic mutations in cattle using zinc finger nucleases. PLoS One 2014;9(4):e95225
    1. Wu H, Wang Y, Zhang Y, Yang M, Lv J, Liu J, et al. TALE nickase-mediated SP110 knockin endows cattle with increased resistance to tuberculosis. Proc Natl Acad Sci U S A 2015;112(13):E1530–E1539.
    1. Wei J, Wagner S, Maclean P, Brophy B, Cole S, Smolenski G, et al. Cattle with a precise, zygote-mediated deletion safely eliminate the major milk allergen beta-lactoglobulin. Sci Rep 2018;8(1):7661.
    1. Petersen B, Niemann H. Molecular scissors and their application in genetically modified farm animals. Transgenic Res 2015;24(3):381–396.
    1. Ikeda M, Matsuyama S, Akagi S, Ohkoshi K, Nakamura S, Minabe S, et al. Correction of a disease mutation using CRISPR/Cas9-assisted genome editing in Japanese black cattle. Sci Rep 2017;7(1):17827.

Metrics
Share
Figures

1 / 3

Tables

1 / 5

Funding Information
PERMALINK