1. Introduction
In the 1990s, genome editing technology began to rise, and with the rapidupdate and iteration of technology, gene editing methods that accurately targeted modification genomes emerged [
1,
2], such as transcription activator like effector nuclease (TALEN), zinc-finger endonuclease (ZFN), clustered regularly interspaced short palidromic repeats (CRISPR) appeared in the late 1990s [
3]. Due to the advantages of CRISPR, such as low cost, simple operation, accurate editing, high efficiency, fast development rate, stable structure of Cas9 nuclease, easy design, system maturity, and carrier ability to simultaneously target multiple sites of genes [
4,
5,
6], CRISPR exceeds any other technology. It has been widely used in livestock and poultry gene function analysis, molecular breeding, genetic improvement, disease resistance breeding, molecular biology and molecular cytology and other genome editing fields [
7,
8,
9]. CRISPR-Cas9, a genetic tool, is a miracle in the 21st century [
10,
11,
12,
13].
Table 1 summarizes the advantages and disadvantages of the three gene editing techniques [
13,
14]. In addition, the epigenetic traits that affect livestock genetics and breedingare essentially regulated by genes. In order to breed better varieties and improve animal husbandry productivity, it is necessary to accurately edit the genome through specificity and multiple loci simultaneously. In this paper, the principle of CRISPR/Cas9 technology and its application in livestock and poultry breeding were reviewed, with a view to providing a reference for CRISPR/Cas9 technology to further study new breeding methods such as genome transcriptional regulation and epigenetics in large livestock.
2. CRISPR/Cas9 Gene Editing Technology
2.1. Overview and Principle of CRISPR/Cas9
CRISPR is an adaptive immune system discovered in the genomes of bacteria and archaea. It is composed of Cas9 protein, functional genes, short regularly clustered repeatsequences, and similarly long spacer sequences arranged from the 5 'to 3' [
15,
16],which can effectively defend against the invasion of exogenous viral DNA [
17] and protect one's own genetic information from destruction [
18]. The proteins encoded by CRISPR is called Cas (CRISPR association proteins), which is a nucleic acid endonucleasecomposed of a Nuclease domain (RuvC-like) and a Nuclease functional region (McrA-like HNH), and it mainly encodes functional proteins that bind to nucleic acids [
19]. According to the different mechanisms of action of Cas proteins, the CRISPR/Cas9 system is divided into two classes and six types [
20]. The class 1 includes type I, type III and type IV, and the class 2 includes type II, type V and type VI. Among them, the CRISPR/Cas9 system of type II is widely used as the most advanced gene editing technology in improving genetic breeding, reproductive performance, and nutrient intake levels of livestock due to its simplest structure, strongest specificity and highestefficiency [
21]. The mechanism of action of the Type II CRISPR/Cas9 system is based on the principle of base complementary pairing, that the Cas9 protein is guided by single guide RNA (sgRNA) to carry out targeted editing of the target DNA, causing double strand break (DSB), thereby stimulating the repair mechanism of DNA [
22].
The Cas9 protein needs to undergo cleavage through the combined action of specific CRISPR RNA (crRNA) and trans-activating CRISPR RNA (tracrRNA) by base pairing binding [
23]. Replacing crRNA and tracrRNA with sgRNA [
24] simplifies the operation of theCRISPR/Cas9 system. sgRNA is adjacent to protospacer adjacent motif (PAM) [
25], so it canensure that Cas9 protein accurately recognizes exogenous DNA without destroying its own Genetic material. However, not all sequences are efficient and specific for induction cutting. Currently, different PAM (5'-NGG-3', N as A/T/C/G) forms of CRISPR/Cas9 systems can be used depending on the sequence characteristics of editing sites [
26]. The CRISPR/Cas system immunoregulates invading elements through three stages: acquisition, transcription, and interference [
27]. It is divided into two different subsystems, the highly conserved "information processing" subsystem (acquisition) and the "implementation" subsystem (transcription and interference). The first stage is to obtain recognition and insert the ingested PAM into the dominant or adjacent area [
6]; In the second stage of transcription, the CRISPR sequence is transcribed together with PAM to form a long transcript called pre-CRISPR RNA, and mature small crRNAs are processed by a unique Cas protein and combinedwith tracrRNA to form complementary chains [
28]; In the third stage, interference, the crRNA-tracrRNA complex accurately induces the Cas9 protein targeted cleavage of exogenous DNA.
The CRISPR/Cas9 system generally repairs genetic material by nonhomologous end joining (NHEJ) and homologous recombination (HR) [
29]. NHEJ is mainlyused for the study of gene knockout such as insertion, deletion or chromosomerearrangement of small fragments, which is the first successful method for theCRISPR-Cas system has prevented the function of endogenous genes in domestic animals such as goats [
30], sheep [
31], cattle [
32] and pigs [
33]. When thehomologous template of heterologous mutated DNA serves as the repair template of DSB, HDR can insert exogenous DNA fragments into specific locations of the genome to achieve accurate editing of target sites [
34]. Following NHEJ,HDR achieved gene knockout in large animal for the first time. Although HDR can be accurately repaired, it also has certain drawbacks. Randomly introduced insertions or deletions may lead to gene knockout, new functions may be generated, and even activate DNA damage regulatory mechanisms to induce cellapoptosis [
35].
Based on the continuous research on the powerful Cas9 protein [
36], moreand more new CRISPR/Cas9 tools [
37] have emerged, such as Streptococcus pyogenesCs9 (SpCas9) is the main tool for studying animal gene editing [
38], the "BE" that makes C/T replace each other [
39,
40], and the "PrimeEditor" [
41] that can accurately target editing without the template of donor genetic material, etc. These new tools not only have a low probability of missing the target, but also there will be no new functional changes [
42].
2.2. Comparison between CRISPR/Cas9 and Other Gene Editing Techniques
At present, the mainstream gene editing technology is the CRISPR/Cas9 system mediated by a shorter RNA. Previously, research on gene editing techniques using Zinc-fingernuclease (ZFN) and transcript activator-like effector nucleases (TALEN) was more common. ZFN and TALEN both use the Fok I familyrestrictive endonuclease to form dimers to specifically recognize and non-specific cut the targeted DNA [
43], but ZFN recognizes triple bases, while TALEN can only recognize single nucleotides. Because of this feature of TALEN, it will not be affected by external factors when designing, and TALEN can target longer sequences [
44], which has a low probability of miss the advantages of short preparation cycle and low experimental cost are more favored than ZFN. Like CRISPR/Cas9, they also repair genetic material through HR and NHEJ mechanisms. Compared with traditional gene editing techniques, ZFN and TALEN achieve gene expression regulation through targeted insertion, base deletion and replacement, transcriptional inhibition or activation, etc., and improve gene targeting efficiency [
45]. However, the development of these two technologies islimited due to their complex operation and high cost.
However, compared with the above two technologies, CRISPR/Cas9 has incomparable advantages in livestock genetics and breeding.
Table 1 summarizes the main differences among the three genome editing technologies. Firstly, the construction of the CRISPR/Cas9 system is extremely simple, requiring no significant human or material resources to design, process, and screen proteins thatcan recognize DNA. Simply designing and synthesizing a gRNA that complements DNA can achieve twice the result with half the effort [
46], which is the main reason why this technology is widely used in the entire scientific research field; Secondly, the CRISPR/Cas9 system can simultaneously edit multiple independent target gene sites based on multiple gRNAs at different sites without the need for multiple domain connections [
47], with high efficiency being its significant advantage. Despite its high efficiency and specificity, the limitationsof the CRISPR-Cas9 system structure also result in off target effects. Therefore,researchers have calculatedand designed the most suitable editing sites to improve the specificity of sgRNA [
45], controlled the number of sgRNAs [
48], and continuously constructed new Cas9 protein mutants, such as EvoCas9 [
49], Sniper Cas9 [
50], xCas9 [
51], and AsCpf [
52], These gene editing tools with simpler structures and higher editing efficiency [
43] can be flexibly used for any nucleic acid sequence. Through continuous innovation and improvement, CRISPR/Cas9's editing ability will be stronger.
Overall, although several gene editing technologies have their own advantages, the CRISPR/Cas9 system has led a technological revolution in different fields due to its simple design and operation, low cost, ability to edit multiple genes simultaneously, and high mutation efficiency [
8,
53].
3. Application of CRISPR/Cas9 System in Livestock and Poultry
3.1. Application of CRISPR/Cas9 Technology in Pig Genetic Breeding
As an important livestock for meat, the scale of pig farming is gradually expanding, and intensive breeding has led to the emergence of different diseases. It is urgent to cultivate genetically modified pigs with disease resistance. Pigsarethe first large animals to be edited by CRISPR/Cas9 [
54], and CRISPR/Cas9 gene editing technology provides new ideas in genetic improvement and disease resistance breeding.
GUANGHAI et al. [
55] used the CRISPR/Cas9 system to edit the non-coding region of insulin-like growth factor 2 (IGF2) in Bama pigs, and found thatit could improve meat production in Bama pigs. Mitochondrial uncoupling protein 1 (UCP1) can regulate body temperature in a cold environment, thereby reducing the body fat production rate of newborn piglets and improving the leanmeat rate and birth rate [
56]. Using CRISPR/Cas9 technology to knock UCP1 into the chromosomes of pigs to construct healthy lean pigs with cold resistance and low fat, thereby reducing economic losses in agriculture and animal husbandry [
57].
Classical swine fever virus (CSFV) and Porcine reproductive and respiratory syndrome virus (PRRSV) are both envelope single-stranded sense RNA viruses [
58], and the replication process does not produce intermediate products. Transgenic pigs resistant to CSFV were constructed using the CRISPR/Cas9 system, and it was found that growing pigs, pregnant sows, and their fetuses are all resistant to PRRSV infection [
59]. CD163 knockout pigs were also constructed, and found that the replication of CSFV was restricted, and disease resistance could be stably transmitted to offspring. African swine fever virus (ASFV) has a higher mortality rate than ordinary swine fever, reaching 100%. HUBNERet al. utilized CRISPR/Cas9 targeting ASFV phosphoprotein p30 to reduce the number of African swine fever viruses [
60]. Xu et al. [
61] constructed CD163and porcine aminopeptidase (pAPN) Double-gene Knockout (DKO) pigs, which inactivated the virus receptor CD163 and pAPN proteins. This not only prevented both PRRSV and TGEV virus infections, but also did not affect reproductive performance and meat quality [
62]. The construction of these disease-resistantgene editing pigs can reduce economic losses caused by diseases and promote a healthier and faster development of the pig industry.
3.2. Application of CRISPR/Cas9 Technology in Chicken Genetic Breeding
Due to the unique structure of chicken fertilized eggs, the development ofCRISPR/Cas9 technology in poultry breeding lags behind that of mammals [
63].Therefore, CRISPR/Cas9 gene editing technology needs to accelerate the pace in genetic aspects such aspoultry reproductive development and disease resistance breeding [
64].
Primordial germ cells (PGCs) in chickens can carry genetic information to the next generation, but both the donor PGC and the recipient PGC develop simultaneously in the recipient testis, leading to rejection of the donor PGC and reduced sperm count. Researchers used gene editing technology to construct sterile hosts to eliminate the role of endogenous receptor PGC. Ballantyne et al. [
65] obtained DAZL gene edited chickens using CRISPR/Cas9 technology, theiCaspase9 gene was fixedly point inserted into the chicken PGC and drugs were added to inhibit the growth of PGC in the gonads, and the effectiveness of infertile hosts was verified by carrying exogenous PGC with feather color. Later, researchers used the CRISPR/Cas9 system to knockout the key gene DMRT1 that determines gonadal differentiation and gender development in male chicken PGC [
66]. The edited chicken PGC was injected into the aforementionedsterile host chicken, and the obtained embryos and chicks' gonads eventually developed into ovaries, proving that the DMRT1 gene is particularly important in testicular development [
67]. Zhang et al. [
68] used CRISPR/Cas9 technology to knockout the important gene Stra8 (Stimulated by recurrent acid gene8) that affects reproductive development in chicken embryonic stem cells,and found that ECS cannot be transformed into male germ cells. MSTN (Myostatin) negativelyregulates the proliferation and differentiation of skeletal muscle cells [
69]. Xu et al. [
70] injected the MSTN-knockout sgRNAs adenovirus vector into theleg muscles of chicks, and the expression of MSTN was significantly downregulated, but gene editing offspring could not be obtained. It can be seen that the study of combining PGC with CRISPR/Cas9 for poultry gene editing is more reasonable.
Poultry is highly susceptible to viruses, such as the MDV (Mark's diseasevirus), the highly pathogenic ALV-J (Avian leukosis virus J) [
71], and the AIV (Avian influenza virus) [
72], with fast transmission speed and high mortalityrate. Koslova et al. successfully cultivated chNHE1-KO homozygous mutant chickens resistant to ALV infection using CRISPR/Cas9 technology [
73]. Use CRISPR/Cas9 to modify the residues of the chANP32A gene to reduce AIV replication [
74]. Hellrich et al. [
75] used the CRISPR/Cas9 system to knockout Trp38 encoded by the chNHE1 (Chicken Na+/H+ exchange type 1) of the ALV-J receptor in chicken PGC, obtaining anti ALV-J chickens. Liu et al. [
72] successfully constructed a CRISPR/Cas9 vector that blocks the expression of ALV-J in vivo using coinfected virus MDV for the first time. In summary, CRISPR/Cas9 gene editing technology has achieved significant results in poultry disease prevention and breeding.
3.3. Application of CRISPR/Cas9 Technology in Cattle Genetic Breeding
Cattle are premium species that produce meat, milk, and derivative leather.At present, research on gene editing in cattle mainly includes disease resistant breeding, increasing meat production, and eliminating allergens. By using Cas9 technology to precisely insert beneficial genes into animals, thus promoting thehealthy and efficient development of the industry, reducing economic losses, and ensuring human food safety.
Bovine tuberculosis is a zoonosis induced by Mycobacterium bovis (M.bovis) infection. Gao et al. [
76] for the first time used Cas9 nickase (Cas9 n) to prepare naturally resistant associated macrophage protein-1 (NRAMP1) gene edited cows, enhancing their resistance to Mycobacterium infection and reducing the risk of human infection caused by contact with diseased cows and their by-products. Subsequently, Yuan et al. [
77] used CRISPR/Cas9 system to integrate NRAMP1 gene into FSCN1-ACTB (F-A) site and bovine homologous mouse Rosa26 site, enhancing the resistance of cows to tuberculosis. SZILLAT et al. [
78] constructed a CD46 gene knockout cell line dependent on the invasion of Bovine Viral Diarrhea Virus (BVDV) using CRISPR/Cas9 technology, laying the foundation for elucidating the mechanism of CD46's role in the replication cycle of plague viruses.
Prevention and treatment of mastitis is a major problem in dairy industry.Bovine paratuberculosis caused by Mycobacterium bovis paratuberculosis (MAP)has a huge impact on dairy industry. Using CRISPR/Cas9 technology to knockout the Interleukin-10 Receptor Alpha (IL-10RA)in bovine mammary epithelial cells that can immunoregulate MAP can inhibit or promote the expression of certain pro-inflammatory cytokines, providing a cellular model for further research on the anti-inflammatory mechanisms of IL-10RA [
79].
Cattle horns are prone to accidental injuries during play and struggle, which can lead to miscarriages in severe cases. Therefore, the use of gene editing technology to cultivate hornless cattle is of great significance. Gu Mingjuan et al. [
80] used CRISPR/Cas9 technology to insert the 202bp repetitive Pc (polling of cellular origin) site of chromosome 1, a gene that controls hornless traits, into the genome of fibroblasts from horned Mongolian cattle, providing a basic material for cultivating hornless cattle.
The OCT4 gene plays an important role in maintaining the pluripotency ofearly embryonic stem cells in mammals. SIMMET et al. [
81] used CRISPR/Cas9 technology to knockout the OCT4 gene in bovine embryos and found that its blastocyst development process was similar to that of human blastocysts lacking OCT4. Bradford et al. [
82] targeted the injection of the OCT4 gene intobovine fertilized eggs and found that on day 5, embryonic development stagnated and blastocyst formation was not possible. Based on this, CAMARGO et al. [
83] further utilized optimized electroporation technology to transfect the CRISPR/Cas9 system targeting the sgRNA-Cas9 protein complex of OCT4 into bovine fertilized eggs, greatly reducing operational difficulties and making it easier to obtain transgenic animals. These results not only provide a model animal for treating early human embryonic developmental defects, but also lay a scientific foundation for promoting cattle reproduction.
4. Application of CRISPR/Cas9 Technology in Goat and Sheep Genetic Breeding
Due to the superiority of cashmere, meat, milk, and other by-products, sheep or goat are also one of the most important livestock in agricultural production and play an important role in animal husbandry. In the genetic breeding processof goats and sheep, gene editing is used to improve growth status, increase cashmere yield and fertility, and obtain individuals with multiple excellent traits. Scientists use gene editing technology to modify their traits according to different needs, in order to accelerate the breeding process of sheep and goat.
4.1. Promote Hair Follicle Growth and Development
Cashmere goats are the main local economic livestock in Inner Mongolia, and cashmere is an important natural raw material for the textile industry. As an important economic value structure for cashmere goats, it is also an important source of income for farmers and herdsmen. Using CRISPR/Cas9 gene editing technology to study the functions of genes related to hair follicle development is of great significance for improving the cashmere production performanceof cashmere producing goat varieties.
Studies have shown that CRISPR/cas9 inhibition of FGF5 expression can improve cashmere growth in sheep [
84,
85] or goats [
86], increasing hair follicledensity, length, and improving quality. Fibroblast growth factor 5 (FGF5) is the main inhibitor that controls fiber length and growth. Wang et al. [
86] targeted the knockout of the FGF5 gene in Shaanbei white cashmere goats using CRISPR/Cas9 technology, and found that the number and density of secondary hair follicles in edited cashmere goats significantly increased, and cashmere lengthsignificantly extended; They further performed HE staining on the skin tissues of aborted and normal goats, and showed that the number of secondary hair follicles in the skin of FGF5 mutant goats was more than that of wild-type goats, the diameters of primary and secondary hair follicles were longer than wild-type goats, and the SHF/PHF ratio was significantly higher than that of normalgoats. Zhang et al. [
87] also confirmed that knocking out FGF5 resulted in a significant increase in fiber length and growth rate. Li et al. [
88] used CRISPR/Cas9 to insert nonsense codons into the FGF5 gene, which improved cashmereyield. Hao et al. [
89] revealed the inhibition of hair follicle growth and development in goats by producing SCNT mediated extracellular receptor abnormalities (EDAR) knockout through CRISPR/Cas9. In addition, Li [
90] added exogenous thymosin β 4 (Tβ4) targeted integration into cashmere goats and discoveryof Tβ4 can induce hair follicle development,accelerate the differentiation of hair follicle stem cells, and play an important role in improving hair production. Hao [
89] combined the CRISPR-Cas9 systemand somatic cell nuclear transfer technology to construct EDAR gene targeted cashmere goats, which exhibited baldness. Hu [
85] utilized the CRISPR/Cas9 system to target Vascular Endothelial Growth Factor (VEGF) in Inner Mongoliawhite cashmere goats with FGF5 and CCR5 genes, which played a crucial rolein cashmere breeding by simultaneously knocking in hair follicle development promoting genes and knocking out hair follicle development inhibiting genes. Wool color is also a key production trait for cashmere. In order to change thecolor of cashmere, Zhang et al. [
91] applied CRISPR/Cas9 technology to targetand knock out the ASIP gene. Compared with wild-type individuals with whitehair, the ASIP edited sheep wool color produced diversity, indicating that ASIPaffects fur color formation and serves as a marker gene. CRISPR/Cas9 currently has a prominent ability to disrupt genes that inhibit hair follicle development into an ideal phenotype, providing a scientific basis for further cultivating high-quality and high-yield cashmere goat breeds, and also providing insights for the treatment of androgenic alopecia.
4.2. Improve Muscle Growth and Development
Researchers [
92,
93,
94] used Cas9/gRNA to block genes that inhibit muscleproduction in meat sheep [
95], improving meat production and quality to meetpeople's livelihood needs for mutton. Myostatin (MSTN) can regulate skeletal muscle growth and promote muscle proliferation. Therefore, inhibiting the expression of MSTN can induce the appearance of "dual muscle" traits in developedmuscle areas such as the buttocks, shoulders, and legs, providing application value for improving meat quality and increasing lean meat percentage in livestock. Albertio and Wolf [
96] boldly hypothesize the use of DNA nucleases to apply this phenotype to the growth of cattle, sheep, and pigs, promoting meat production performance in livestock and poultry.
He et al. [
97] injected targeted MSTN into the cytoplasm of prokaryotic embryos to obtain gene edited goats with dual muscle traits. Wang et al. [
92] demonstrated that the muscle development level of MSTN-edited sheep was significantly faster than that of wild-type sheep. Niu et al. [
98] found that sheep modified with BCO2 biallelic genes exhibited yellow fat, indicating that the BCO2 gene may determine the formation of fat color. Wan et al. [
99] used CRISPR/Cas9 system to knock out the conserved site of the goatCLPG1 gene for the first time in order to study the "beautiful buttocks" of sheep. In addition, Zhang et al. [
93] demonstrated that the loss of MSTN function in sheep skeletalmuscle satellite cells (sSMSCs) promotes the differentiation and growth of sSMSCs. Zhou et al. [
100] introduced single nucleotide point mutations in the Recombinant SuppressorsOf Cytokine Signaling 2 (SOCS2) using the CRISPR/cas9system, resulting in an increase in sheep weight and milk production; in 2022, they knocked out the MSTN bialleles with a high proportion of mutations and found that meat quality was almost unaffected [
101]. On the basis of MSTN mutant sheep, not only were changes in transcriptome gene expression analyzedin MSTN knockout goats [
102], but also confirmed how knockout alleles are inherited to offspring [
92]. Mei obtained one double allele and one single allele MSTN gene mutated goat through microinjection of fertilized eggs. Both edited sheep fetuses had "double muscle" and significantly increased weight and length. Further sectioning revealed a significant increase in muscle cross-section and muscle fiber density [
103]. CRISPR/Cas9 significantly highlights the function of genes related to muscle growthand development in sheep, and has potential roles in the cultivation of new breeds of meat sheep.
4.3. Improving Reproductive Capacity
Improving reproductive performance is a key goal of breeding. The Fec familygenes are associated with the fecundity of sheep. Zhou et al. [
104] used CRISPR/Cas9 technology to inject single-strand FecB DNA oligonucleotides into thezygote of sheep, obtaining FecB homozygous mutant sheep, proving that FecB homozygous ewes not only have higher lambing rates but also improve reproductive rates. The CRISPR/Cas9 system simultaneously edits HYAL2 and PrP genes to improve lambing rate [
8]. Niu et al. [
28] introduced point mutations in goat growth differentiation factor 9 (GDF9) and found that it also affects ovulation rate and litter size. Zhang et al. [
105] used the CRISPR/Cas9 systemto induce functional deletion mutations in the bone morphogenetic protein receptor type IB (BMPR-IB) gene in sheep, leading to increased ovulation rate and litter size. Based on ssODNs, Zhou et al. [
104] further introduced point mutations to generate BMPR-1B gene edited sheep. Tian et al. [
106] microinjected the AANAT gene into frozen and unfrozen sheep embryos, and the results showed that there was no significant difference in the reproductive ability of transgenic offspring between the two environments, but AANAT transgenic individuals had good reproductive ability. It can be seen that CRISPR/Cas9 technology is an effective editing tool for developing ideal traits of farm animals, promoting the development of animal husbandry.
4.4. Improving Milk Composition
At present, researchers have enriched the composition of sheep milk by genetically modifying essential nutrients and knocking out non essential proteins in sheep milk. Goat milk is rich in fat and protein, and its composition is similar to human milk, but β-Lactoglobulin (BLG) is prone to sensitization. Zhou et al. [
107] prepared using CRISPR/Cas9 technology knock out of β-lactoglobulin in goats reduces the protein concentration of BLG, laying the foundation for improving the composition of goat milk. Wei et al. [
108] constructed BLG gene knock out cows from fertilized eggs to broaden the audience for milk and its by-products. Another study used the CRISPR/Cas9 system to knock out factors that affect milk traits, such as Stearoyl CoA Desaturase 1 (SCD1) [
109] and Acetyl CoA acyltransferase 2 (ACAA2) [
110]. In breast cells, Ma et al. [
111] used CRISPR/Cas9 technology to inject Arylkylamine N-acetyltransferase (AANAT) and Acetylserotonin O-methyltransferase (ASMT), which mediate melatonin expression, into the cytoplasm of sheep fertilized eggs, and constructed an AANAT/ASMT breast bioreactor, resulting in edited sheep with high-yield of melatonin milk. CRISPR/Cas9 technology plays an irreplaceable role in the development of non artificial natural low-fat orspecific functional dairy products, promoting the development of the dairy industry rich in specific nutrients or pharmaceutical ingredients.
4.5. Establishment of Animal Disease Resistance Breeding and Human Disease Models
The economic losses caused by livestock and poultry diseases not only limit the development of animal husbandry, but also pose a threat to human safety in the event of an outbreak of zoonotic diseases. The CRISPR/Cas9 systemhas been used in disease resistance breeding for several diseases, providing a good therapeutic model for studying the pathogenesis and improving animal health. In addition, the body size and anatomical structure of sheep are similar to humans, and CRISPR/Cas9 has been used in sheep to provide an effective modelfor studying human diseases.
Fan et al. [
112] targeted knock out PRP in goat fibroblast donor cells to produce SCNT mediated anti-PRP goats, and using CRISPR/Cas9 to generate NUP155 gene knockout donor cells in goat fibroblasts, a NUP155 gene knockout goat model for studying heart disease was obtained through SCNT program. Menchaca et al. [
113] knocked out hyaluronidase 2 (HYAL2) in lambs, leading to lung adenocarcinoma syndrome, and confirmed the possibility of the CRISPR/Cas9 system producing antiviral animals, they also discovered for the first time that the deletion of the sheep otoferlin (OOF) gene is an effective model for treating deafness [
114]. Li [
115] inserted Tβ4 gene into the CCR5 site in goat, laying the foundation for establishing a goat knock inmodel. Fan et al. [
116] used CRISPR/Cas9 combined with SCNT technology to cultivate IFNA gene knock out sheep, providing a large-scale animal model for fetalresistance to Zika virus (ZIKV) infection. Subsequently, they pioneered the human cystic fibrosis (CF) sheep model with CRISPR/Cas9 mediated cystic fibrosis transmembrane transmission regulator (CFTR) deficiency using the same technology [
59]. The liver and gallbladder disease phenotype of newborn CFTR−/− sheep was consistent with that of humans. Williams et al. [
117] applied CRISPR/Cas9 to point mutate the non-specific alkaline phosphatase (TNSALP) gene alkaline phosphatase (ALPL) , resulting in edited lambs exhibiting human hypophosphatasia (HPP), providing an effective animal model for studying this rare metabolic bone disease. Vilarino et al. [
118] constructed Pancreas/duodenumhomeobox protein 1 (PDX1) fetuses, laying the foundation for gene edited sheep as host organs for xenotransplantation. In addition, the interspecies blastocystcomplementary technology combining embryonic gene editing and pluripotent stem cells (PSCs) has achieved xenotransplantation of human organs in large animals as hosts. The production of these disease-related gene knock out sheep demonstrates the possibility and effectiveness of the CRISPR/Cas9 system in breeding antiviral livestock, as well as its enormous potential for the treatment of human diseases.
4.6. Current Problems of Gene Editing Sheep
Although CRISPR/Cas9 technology has broad prospects in the developmentof animal husbandry, the low targeting efficiency, poor chimerism, and off-target effects of the CRISPR/Cas9 system [
119] seriously restrict the sheep breeding process. Scientists are constantly optimizing the CRISPR/Cas9 system to be more efficient, accurate, and safe. The CRISPR/Cas9 system has developmentaldefects and low survival rates when editing cells in vitro to obtain gene editedhomozygotes. Direct injection into the fertilized egg results in the formation ofchimeras by simultaneously editing multiple cells due to division. Therefore, electroporation of Cas9 protein and sgRNA into embryos to construct gene edited sheep not only obtained homozygous positive individuals but also improved editing efficiency. Improving the targeting efficiency of gene edited sheep is related to various factors, such as increasing the embryo injection concentration of the CRISPR/Cas9 system or improving the conception rate [
120].
Off-target effects cause unpredictable consequences such as base mutations,deletions, rearrangements, and immune responses [
121]. Therefore, reducing theoff-target rate is of utmost importance in improving the CRISPR/Cas9 system. At present, strategies such as optimizing sgRNA length, modifying Cas9 nuclease specificity, and using other Cas variants are mainly used to reduce off target effects [
122]. Wienert et al. [
123] developed a method called DISCOVER-Seq for identifying Cas off-target. Due to its ability to recruit DNA repair factors in cells and individuals, this method is applicable to various sgRNAs and Cas proteins. Donohue et al. [
124] proposed a technique called CRISPR hybrid RNA DNA (chRDNA) to reduce off-target rates while maintaining targeted editing ability and improving Cas9 specificity. Although the efficiency of using viral vectors to deliver CRISPR systems is high [
125], the Cas9 protein is relatively large and needs to be packaged separately before co-injection. MOUT et al. [
126] developed an engineered DNA-Free Virus Like Particles (eVLPs) that can be efficiently edited in various cells and organs, minimizing off-target rates and the risk of DNA integration. Perez et al. [
127] developed an algorithm called CRISPR Specific Correction (CSC) system for correcting specific gRNAs targeting mismatches in non coding and repetitive regions. The SpCas9 mutant and other gene editing techniques have overcome the drawbacks of off target efficiency in the CRISPR/Cas9 system [
128], but they have also changed the species gene pool and ecosystem to a certain extent. Therefore, it is necessary to upgrade and improve the development of more accurate and easily controllableCRISPR/Cas9 systems [
129].
5. Conclusion and Prospect
Since the emergence of the CRISPR/Cas9 system, it has made a qualitative leap in the progress of modern breeding in agriculture and animal husbandrywith its characteristics of precision, efficiency, simplicity, and economy. As a popular method for studying target genes, the development of CRISPR has solved some new challenges in genetic breeding, production performance, animal health, environmental protection, and human health in modern animal husbandry. With the development of technology, molecular breeding and gene editing breeding that achieve gene recombination through targeted gene transfer have replaced traditional breeding methods such as artificial selection and multi generationhybridization. CRISPR/Cas9 technology can accurately integrate, delete, and replace genes across species, insert excellent trait genes according to needs, break reproductive isolation, and achieve mutual acquisition of disease resistance genes from different species [
130], reducing the time and cost of long-term breeding, accelerating the breeding process, providing new research ideas for cultivating high-quality and high-yield disease resistant livestock varieties, and creating new production models for gene edited animals. In addition, constructing multiple sgRNA vectors while editing multiple genes of the same individual genomeis currently the main research direction of resistance breeding [
61]. Despite the many benefits of gene editing breeding, there are still some issues and risks:how to ensure the safety of gene edited species and their by-products? How tofind effective genes that affect the disease resistance of livestock? Have national regulations introduced policies for the use and promotion of genetically modified animals? Can humans accept the consumption of genetically modified agricultural products? Once these issues are resolved, the application of CRISPR gene editing technology in animal husbandry breeding will be even more fruitful.
At present, the regulatory standards for gene edited animals in most countries are not clear. Although regulatory agencies attempt to restrict the production of genome edited livestock and poultry, scientists believe that as long as standardized biosafety testing is passed, it can be applied to commerce. Government departments have established policies such as monitoring the breeding and consumption mechanisms of gene edited agricultural and livestock products, gradually allowing genetically modified animals to enter the market [
131]. With thecontinuous improvement of supervision mechanisms and scientific technology, the public's recognition of genetically modified livestock and poultry products will also continue to increase. The emergence of CRISPR/Cas9 whole genome scanning technology not only saves time and cost, but also avoids the risk of disease transmission, making it an effective tool for accurately screening resistance genes. However, for disease resistant traits that are not suitable for cell death and virus replication screening, it is currently insufficient to explore the function of their target genes [
61]. Although the existing CRISPR/Cas9 technologycan achieve the level of simultaneous editing of multiple genes, it cannot achieve over 20 or even hundreds of genes. It can be seen that in the future, continuous exploration and innovation are needed to optimize the CRISPR screeninglibrary and screening conditions, and build a more solid and efficient multi gene editing technology, in order to achieve simultaneous regulation of multiple important economic traits of livestock and poultry micro effect genes [
132], thereby obtaining the most ideal genetic improvement species.
In summary, CRISPR/Cas9 technology has achieved fruitful results in genetic breeding and improvement of livestock, promoting the sustainable development of animal husbandry. With the continuous improvement of gene editing technology and the continuous exploration of the structure and function of Cas9, further enhancing the safety and superiority of gene editing, CRISPR technology will undoubtedly have a more profound impact. On the basis of innovation and protection, we will promote the progress of gene editing technology. We believe that gene edited animal products will eventually become market-oriented, and the development of human society will achieve a more inclusive and sustainable future.
Author Contributions
methodology, Lu Zeyu and Zhang Lingtian; validation, Lu Zeyu, Zhang Lingtian, Mu Qing and Liu Junyang; formal analysis, Lu Zeyu and Teri Gele; investigation, Lu Zeyu, Chen Yu and Wang Haoyuan; writing—original draft preparation, Lu Zeyu; writing—review and editing, Lu Zeyu; supervision, Zhao Yanhong, Zhang Yanjun, Su Rui, Wang Ruijun, Wang Zhiying, Lv Qi, Liu Zhihong, Liu Jiasen and Li Yunhua; project administration, Zhao Yanhong; funding acquisition, Zhao Yanhong. All authors have read and agreed to the published version of the manuscript.
Acknowledgments
This research was funded by the National Natural Science Foundation of China (32160772), Science and Technology Project of Inner Mongolia Autonomous Region (2023YFHH0076), Science and technology Major Project of Inner Mongolia (2021ZD0012), and Supported by Program for Innovative Research Team in Universities of Inner Mongolia Autonomous Region (NMGIRT2322).
Conflicts of Interest
The authors declare no conflict of interest.
References
- Mirza, Z.; Karim, S. Advancements in CRISPR/Cas9 technology—Focusing on cancer therapeutics and beyond. Seminars in Cell & Developmental Biology 2019, 96, 13-21. [CrossRef]
- Mehravar, M.; Shirazi, A.; Nazari, M.; Banan, M. Mosaicism in CRISPR/Cas9-mediated genome editing. Developmental Biology 2019, 445, 156-162. [CrossRef]
- Abdelrahman, M.; Al-Sadi, A.M.; Pour-Aboughadareh, A.; Burritt, D.J.; Tran, L.-S.P. Genome editing using CRISPR/Cas9–targeted mutagenesis: An opportunity for yield improvements of crop plants grown under environmental stresses. Plant Physiology and Biochemistry 2018, 131, 31-36. [CrossRef]
- Tyagi, S.; Kumar, R.; Das, A.; Won, S.Y.; Shukla, P. CRISPR-Cas9 system: A genome-editing tool with endless possibilities. Journal of biotechnology 2020, 319, 36-53. [CrossRef]
- Gupta, D.; Bhattacharjee, O.; Mandal, D.; Sen, M.K.; Dey, D.; Dasgupta, A.; Kazi, T.A.; Gupta, R.; Sinharoy, S.; Acharya, K.; et al. CRISPR-Cas9 system: A new-fangled dawn in gene editing. Life sciences 2019, 232, 116636. [CrossRef]
- 张佳珊; 谭韬. CRISPR-Cas9系统编辑DNA诱导基因敲除的发展及优缺点 %J 中国免疫学杂志. 2019, 35, 767-770.
- 宋绍征; 陆睿; 张婷; 何正义; 吴赵曼秋; 成勇; 周鸣鸣. CRISPR /Cas9基因编辑技术在山羊和绵羊中的应用研究进展 %J 生物技术通报. 2020, 36, 62-68. [CrossRef]
- Kalds, P.; Zhou, S.; Cai, B.; Liu, J.; Wang, Y.; Petersen, B.; Sonstegard, T.; Wang, X.; Chen, Y. Sheep and Goat Genome Engineering: From Random Transgenesis to the CRISPR Era. Frontiers in genetics 2019, 10, 750. [CrossRef]
- 徐嘉威; 贺花; 沈雪梅; 刘鲲鹏; 雷初朝; 陈宏; 黄永震. 基因编辑技术在家畜育种中的研究进展 %J 基因组学与应用生物学. 2018, 37, 1423-1430. [CrossRef]
- Baliou, S.; Adamaki, M.; Kyriakopoulos, A.; Spandidos, D.; Panagiotidis, M.; Christodoulou, I.; Zoumpourlis, V. CRISPR therapeutic tools for complex genetic disorders and cancer (Review). International Journal of Oncology 2018. [CrossRef]
- Martinez-Lage, M.; Puig-Serra, P.; Menendez, P.; Torres-Ruiz, R.; Rodriguez-Perales, S. CRISPR/Cas9 for Cancer Therapy: Hopes and Challenges. Biomedicines 2018, 6, 105. [CrossRef]
- Kruminis-Kaszkiel, E.; Juranek, J.; Maksymowicz, W.; Wojtkiewicz, J. CRISPR/Cas9 Technology as an Emerging Tool for Targeting Amyotrophic Lateral Sclerosis (ALS). International Journal of Molecular Sciences 2018, 19, 906. [CrossRef]
- 王欢; 邹惠影; 朱化彬; 赵善江. CRISPR/Cas9基因编辑技术在家畜育种新材料创制中的研究进展 %J 畜牧兽医学报. 2021, 52, 851-861.
- Lamas-Toranzo, I.; Guerrero-Sánchez, J.; Miralles-Bover, H.; Alegre-Cid, G.; Pericuesta, E.; Bermejo-Álvarez, P. CRISPR is knocking on barn door. Reproduction in domestic animals = Zuchthygiene 2017, 52 Suppl 4, 39-47. [CrossRef]
- Horvath, P.; Barrangou, R. CRISPR/Cas, the Immune System of Bacteria and Archaea. Science 2010, 327, 167-170. [CrossRef]
- Han, X.; Liu, Z.; Jo, M.C.; Zhang, K.; Li, Y.; Zeng, Z.; Li, N.; Zu, Y.; Qin, L. CRISPR-Cas9 delivery to hard-to-transfect cells via membrane deformation. Science Advances 2015, 1, e1500454. [CrossRef]
- 黄玉. 山羊CRISPR/Cas9基因编辑系统优化的研究. 硕士, 2018.
- Vink, J.N.A.; Baijens, J.H.L.; Brouns, S.J.J. PAM-repeat associations and spacer selection preferences in single and co-occurring CRISPR-Cas systems. Genome biology 2021, 22, 281. [CrossRef]
- Perisse, I.V.; Fan, Z.; Singina, G.N.; White, K.L.; Polejaeva, I.A. Improvements in Gene Editing Technology Boost Its Applications in Livestock. Frontiers in genetics 2021, 11, 614688. [CrossRef]
- 李岚. 利用CRISPR文库鉴定绒山羊毛乳头细胞增殖的必需基因研究. 硕士, 2021.
- Khadempar, S.; Familghadakchi, S.; Motlagh, R.A.; Farahani, N.; Dashtiahangar, M.; Rezaei, H.; Gheibi Hayat, S.M. CRISPR–Cas9 in genome editing: Its function and medical applications. Journal of Cellular Physiology 2019, 234, 5751-5761. [CrossRef]
- Pelletier, S.; Gingras, S.; Green, Douglas R. Mouse Genome Engineering via CRISPR-Cas9 for Study of Immune Function. Immunity 2015, 42, 18-27. [CrossRef]
- Cong, L.; Zhang, F. Genome engineering using CRISPR-Cas9 system. Methods in molecular biology (Clifton, N.J.) 2015, 1239, 197-217. [CrossRef]
- Xiao, A.; Cheng, Z.; Kong, L.; Zhu, Z.; Lin, S.; Gao, G.; Zhang, B. CasOT: a genome-wide Cas9/gRNA off-target searching tool. Bioinformatics 2014, 30, 1180-1182. [CrossRef]
- Song, M.; Koo, T. Recent advances in CRISPR technologies for genome editing. Archives of Pharmacal Research 2021, 44, 537-552. [CrossRef]
- Xue, C.; Sashital, D.G. Mechanisms of Type I-E and I-F CRISPR-Cas Systems in Enterobacteriaceae. EcoSal Plus 2019, 8. [CrossRef]
- Faure, G.; Shmakov, S.A.; Makarova, K.S.; Wolf, Y.I.; Crawley, A.B.; Barrangou, R.; Koonin, E.V. Comparative genomics and evolution of trans-activating RNAs in Class 2 CRISPR-Cas systems. RNA Biology 2019, 16, 435-448. [CrossRef]
- Niu, Y.; Zhao, X.; Zhou, J.; Li, Y.; Huang, Y.; Cai, B.; Liu, Y.; Ding, Q.; Zhou, S.; Zhao, J.; et al. Efficient generation of goats with defined point mutation (I397V) in GDF9 through CRISPR/Cas9. Reproduction, Fertility and Development 2018, 30, 307. [CrossRef]
- Eaton, S.L.; Proudfoot, C.; Lillico, S.G.; Skehel, P.; Kline, R.A.; Hamer, K.; Rzechorzek, N.M.; Clutton, E.; Gregson, R.; King, T.; et al. CRISPR/Cas9 mediated generation of an ovine model for infantile neuronal ceroid lipofuscinosis (CLN1 disease). Scientific reports 2019, 9, 9891. [CrossRef]
- Perota, A.; Lagutina, I.; Duchi, R.; Zanfrini, E.; Lazzari, G.; Judor, J.P.; Conchon, S.; Bach, J.M.; Bottio, T.; Gerosa, G.; et al. Generation of cattle knockout for galactose-α1,3-galactose and N-glycolylneuraminic acid antigens. Xenotransplantation 2019, 26, e12524. [CrossRef]
- Wang, K.; Tang, X.; Liu, Y.; Xie, Z.; Zou, X.; Li, M.; Yuan, H.; Ouyang, H.; Jiao, H.; Pang, D. Efficient Generation of Orthologous Point Mutations in Pigs via CRISPR-assisted ssODN-mediated Homology-directed Repair. Molecular Therapy - Nucleic Acids 2016, 5, e396. [CrossRef]
- Bravo, J.P.K.; Liu, M.-S.; Hibshman, G.N.; Dangerfield, T.L.; Jung, K.; McCool, R.S.; Johnson, K.A.; Taylor, D.W. Structural basis for mismatch surveillance by CRISPR–Cas9. Nature 2022, 603, 343-347. [CrossRef]
- Wang, T.; Yu, H.; Hughes, N.W.; Liu, B.; Kendirli, A.; Klein, K.; Chen, W.W.; Lander, E.S.; Sabatini, D.M. Gene Essentiality Profiling Reveals Gene Networks and Synthetic Lethal Interactions with Oncogenic Ras. Cell 2017, 168, 890-903.e815. [CrossRef]
- Collias, D.; Beisel, C.L. CRISPR technologies and the search for the PAM-free nuclease. Nature Communications 2021, 12, 555. [CrossRef]
- 冯爽; 王春伟; 苏小虎. 动物基因组编辑中提升CRISPR/Cas9介导的同源重组效率研究进展 %J 中国生物工程杂志. 2022, 42, 83-92. [CrossRef]
- Fu, Y.-W.; Dai, X.-Y.; Wang, W.-T.; Yang, Z.-X.; Zhao, J.-J.; Zhang, J.-P.; Wen, W.; Zhang, F.; Oberg, K.C.; Zhang, L.; et al. Dynamics and competition of CRISPR–Cas9 ribonucleoproteins and AAV donor-mediated NHEJ, MMEJ and HDR editing. Nucleic Acids Research 2021, 49, 969-985. [CrossRef]
- Komor, A.C.; Kim, Y.B.; Packer, M.S.; Zuris, J.A.; Liu, D.R. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 2016, 533, 420-424. [CrossRef]
- Gaudelli, N.M.; Komor, A.C.; Rees, H.A.; Packer, M.S.; Badran, A.H.; Bryson, D.I.; Liu, D.R. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature 2017, 551, 464-471. [CrossRef]
- Chen, P.J.; Hussmann, J.A.; Yan, J.; Knipping, F.; Ravisankar, P.; Chen, P.F.; Chen, C.; Nelson, J.W.; Newby, G.A.; Sahin, M.; et al. Enhanced prime editing systems by manipulating cellular determinants of editing outcomes. Cell 2021, 184, 5635-5652.e5629. [CrossRef]
- Anzalone, A.V.; Randolph, P.B.; Davis, J.R.; Sousa, A.A.; Koblan, L.W.; Levy, J.M.; Chen, P.J.; Wilson, C.; Newby, G.A.; Raguram, A.; et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 2019, 576, 149-157. [CrossRef]
- 呼啸. 利用CRISPR/Cas9技术制备FGF5位点定点整合VEGF基因绒山羊. 硕士, 2018.
- 郝斐. 利用CRISPR-Cas9系统与体细胞核移植技术制备EDAR基因打靶绒山羊的研究. 博士, 2018.
- 李尚; 石岗; 冯韶华; 张正敏; 周一彤; 芦春莲; 曹洪战. CRISPR/Cas9系统在猪基因编辑中的研究进展 %J 中国畜牧杂志. 2019, 55, 27-32. [CrossRef]
- Li, C.; Brant, E.; Budak, H.; Zhang, B. CRISPR/Cas: a Nobel Prize award-winning precise genome editing technology for gene therapy and crop improvement. Journal of Zhejiang University-SCIENCE B 2021, 22, 253-284. [CrossRef]
- Ng, I.S.; Keskin, B.B.; Tan, S.I. A Critical Review of Genome Editing and Synthetic Biology Applications in Metabolic Engineering of Microalgae and Cyanobacteria. Biotechnology Journal 2020, 15, 1900228. [CrossRef]
- Pickar-Oliver, A.; Gersbach, C.A. The next generation of CRISPR–Cas technologies and applications. Nature Reviews Molecular Cell Biology 2019, 20, 490-507. [CrossRef]
- Komor, A.C.; Badran, A.H.; Liu, D.R. CRISPR-Based Technologies for the Manipulation of Eukaryotic Genomes. Cell 2017, 168, 20-36. [CrossRef]
- 郭全娟; 韩秋菊; 张建. CRISPR/Cas9技术的脱靶效应及优化策略 %J 生物化学与生物物理进展. 2018, 45, 798-807. [CrossRef]
- Casini, A.; Olivieri, M.; Petris, G.; Montagna, C.; Reginato, G.; Maule, G.; Lorenzin, F.; Prandi, D.; Romanel, A.; Demichelis, F.; et al. A highly specific SpCas9 variant is identified by in vivo screening in yeast. Nature biotechnology 2018, 36, 265-271. [CrossRef]
- Lee, J.K.; Jeong, E.; Lee, J.; Jung, M.; Shin, E.; Kim, Y.-h.; Lee, K.; Jung, I.; Kim, D.; Kim, S.; et al. Directed evolution of CRISPR-Cas9 to increase its specificity. Nature Communications 2018, 9, 3048. [CrossRef]
- Hu, J.H.; Miller, S.M.; Geurts, M.H.; Tang, W.; Chen, L.; Sun, N.; Zeina, C.M.; Gao, X.; Rees, H.A.; Lin, Z.; et al. Evolved Cas9 variants with broad PAM compatibility and high DNA specificity. Nature 2018, 556, 57-63. [CrossRef]
- Bin Moon, S.; Lee, J.M.; Kang, J.G.; Lee, N.-E.; Ha, D.-I.; Kim, D.Y.; Kim, S.H.; Yoo, K.; Kim, D.; Ko, J.-H.; et al. Highly efficient genome editing by CRISPR-Cpf1 using CRISPR RNA with a uridinylate-rich 3′-overhang. Nature Communications 2018, 9, 3651. [CrossRef]
- 刘皎. MSTN基因敲除绒山羊生产性能及分子生物学评价. 硕士, 2020.
- 房元杰; 张晓爱; 魏文康; 刘春朋. CRISPR-Cas9技术原理及其在猪的应用研究新进展 %J 现代畜牧兽医. 2021, 92-96.
- Xiang, G.; Ren, J.; Hai, T.; Fu, R.; Yu, D.; Wang, J.; Li, W.; Wang, H.; Zhou, Q. Editing porcine IGF2 regulatory element improved meat production in Chinese Bama pigs. Cellular and Molecular Life Sciences 2018, 75, 4619-4628. [CrossRef]
- 徐景; 杨光; 江美祺; 丁向彬; 郭益文; 胡德宝; 李新; 郭宏; 张林林. CRISPR/Cas9技术在畜禽育种中的研究进展 %J 中国畜牧兽医. 2022, 49, 1374-1383. [CrossRef]
- Jabbar, A.; Zulfiqar, F.; Mahnoor, M.; Mushtaq, N.; Zaman, M.H.; Din, A.S.U.; Khan, M.A.; Ahmad, H.I. Advances and Perspectives in the Application of CRISPR-Cas9 in Livestock. Molecular Biotechnology 2021, 63, 757-767. [CrossRef]
- 方满新. 猪繁殖与呼吸综合征病毒复制及感染影响因素的研究新进展 %J 中国预防兽医学报. 2021, 43, 679-685.
- Fan, Z.; Perisse, I.V.; Cotton, C.U.; Regouski, M.; Meng, Q.; Domb, C.; Van Wettere, A.J.; Wang, Z.; Harris, A.; White, K.L.; et al. A sheep model of cystic fibrosis generated by CRISPR/Cas9 disruption of the CFTR gene. JCI insight 2018, 3, e123529. [CrossRef]
- Hübner, A.; Petersen, B.; Keil, G.M.; Niemann, H.; Mettenleiter, T.C.; Fuchs, W. Efficient inhibition of African swine fever virus replication by CRISPR/Cas9 targeting of the viral p30 gene (CP204L). Scientific reports 2018, 8, 1449. [CrossRef]
- Xu, K.; Zhou, Y.; Mu, Y.; Liu, Z.; Hou, S.; Xiong, Y.; Fang, L.; Ge, C.; Wei, Y.; Zhang, X.; et al. CD163 and pAPN double-knockout pigs are resistant to PRRSV and TGEV and exhibit decreased susceptibility to PDCoV while maintaining normal production performance. eLife 2020, 9, e57132. [CrossRef]
- 李晓娇; 何燕华; 朱新宇; 邹娴; 罗成龙. CRISPR/Cas9技术在猪、鸡中的应用研究进展 %J 中国畜牧兽医. 2022, 49, 4665-4673. [CrossRef]
- Tatiana, L.; Anna, K.; Grigory, P.; Yuri, S.; Olga, B. Development of optimal technological approaches for obtaining PGCs in Pushkin breed chickens for further transformation by the CRISPR / Cas9 system %J The FASEB Journal. 2021, 35.
- Lee, J.; Kim, D.-H.; Lee, K. Current Approaches and Applications in Avian Genome Editing. International Journal of Molecular Sciences 2020, 21, 3937. [CrossRef]
- Ballantyne, M.; Woodcock, M.; Doddamani, D.; Hu, T.; Taylor, L.; Hawken, R.J.; McGrew, M.J. Direct allele introgression into pure chicken breeds using Sire Dam Surrogate (SDS) mating. Nature Communications 2021, 12, 659. [CrossRef]
- 林晓; 李硕; 金子笛; 耿拓宇; 龚道清; 刘龙. DMRT1和FOXL2基因在动物性别决定中的功能研究进展 %J 中国家禽. 2021, 43, 98-105. [CrossRef]
- Ioannidis, J.; Taylor, G.; Zhao, D.; Liu, L.; Idoko-Akoh, A.; Gong, D.; Lovell-Badge, R.; Guioli, S.; McGrew, M.J.; Clinton, M. Primary sex determination in birds depends on DMRT1 dosage, but gonadal sex does not determine adult secondary sex characteristics. Proceedings of the National Academy of Sciences 2021, 118, e2020909118. [CrossRef]
- Zhang, Y.; Wang, Y.; Zuo, Q.; Li, D.; Zhang, W.; Wang, F.; Ji, Y.; Jin, J.; Lu, Z.; Wang, M.; et al. CRISPR/Cas9 mediated chicken Stra8 gene knockout and inhibition of male germ cell differentiation. PLOS ONE 2017, 12, e0172207. [CrossRef]
- 陈楚雯; 李洁; 赵瑞鹏; 李志雄. CRISPR/Cas9技术在鸡遗传育种中的研究进展 %J 中国家禽. 2023, 45, 96-103. [CrossRef]
- Xu, K.; Han, C.X.; Zhou, H.; Ding, J.M.; Xu, Z.; Yang, L.Y.; He, C.; Akinyemi, F.; Zheng, Y.M.; Qin, C.; et al. Effective MSTN Gene Knockout by AdV-Delivered CRISPR/Cas9 in Postnatal Chick Leg Muscle. International Journal of Molecular Sciences 2020, 21, 2584. [CrossRef]
- 王金萍; 路宏朝; 张涛; 王令. CRISPR/Cas9技术在鸡抗病毒感染中应用的研究进展 %J 黑龙江畜牧兽医. 2022, 28-34+41. [CrossRef]
- Liu, Y.; Xu, Z.; Zhang, Y.; Yu, M.; Wang, S.; Gao, Y.; Liu, C.; Zhang, Y.; Gao, L.; Qi, X.; et al. Marek's disease virus as a CRISPR/Cas9 delivery system to defend against avian leukosis virus infection in chickens. Veterinary microbiology 2020, 242, 108589. [CrossRef]
- Koslová, A.; Trefil, P.; Mucksová, J.; Reinišová, M.; Plachý, J.; Kalina, J.; Kučerová, D.; Geryk, J.; Krchlíková, V.; Lejčková, B.; et al. Precise CRISPR/Cas9 editing of the NHE1 gene renders chickens resistant to the J subgroup of avian leukosis virus. Proceedings of the National Academy of Sciences 2020, 117, 2108-2112. [CrossRef]
- Park, Y.H.; Woo, S.J.; Chungu, K.; Lee, S.B.; Shim, J.H.; Lee, H.J.; Kim, I.; Rengaraj, D.; Song, C.S.; Suh, J.Y.; et al. Asp149 and Asp152 in chicken and human ANP32A play an essential role in the interaction with influenza viral polymerase. The FASEB Journal 2021, 35. [CrossRef]
- Hellmich, R.; Sid, H.; Lengyel, K.; Flisikowski, K.; Schlickenrieder, A.; Bartsch, D.; Thoma, T.; Bertzbach, L.D.; Kaufer, B.B.; Nair, V.; et al. Acquiring Resistance Against a Retroviral Infection via CRISPR/Cas9 Targeted Genome Editing in a Commercial Chicken Line. Frontiers in Genome Editing 2020, 2, 3. [CrossRef]
- Gao, Y.; Wu, H.; Wang, Y.; Liu, X.; Chen, L.; Li, Q.; Cui, C.; Liu, X.; Zhang, J.; Zhang, Y. Single Cas9 nickase induced generation of NRAMP1 knockin cattle with reduced off-target effects. Genome biology 2017, 18, 13. [CrossRef]
- Yuan, M.; Zhang, J.; Gao, Y.; Yuan, Z.; Zhu, Z.; Wei, Y.; Wu, T.; Han, J.; Zhang, Y. HMEJ-based safe-harbor genome editing enables efficient generation of cattle with increased resistance to tuberculosis. Journal of Biological Chemistry 2021, 296, 100497. [CrossRef]
- Szillat, K.P.; Koethe, S.; Wernike, K.; Höper, D.; Beer, M. A CRISPR/Cas9 Generated Bovine CD46-knockout Cell Line-A Tool to Elucidate the Adaptability of Bovine Viral Diarrhea Viruses (BVDV). Viruses 2020, 12. [CrossRef]
- 王彤; 高元鹏; 韩静; 张景程; 李喜和; 何婷依; 曹贵方. CRISPR/Cas9基因编辑技术在家畜中的应用研究进展 %J 动物医学进展. 2021, 42, 78-84. [CrossRef]
- 谷明娟; 高丽; 周新宇; 吴迪; 魏著英; 李光鹏; 白春玲. 蒙古牛无角POLLED位点的定点编辑 %J 农业生物技术学报. 2020, 28, 242-250.
- Simmet, K.; Zakhartchenko, V.; Philippou-Massier, J.; Blum, H.; Klymiuk, N.; Wolf, E. OCT4/POU5F1 is required for NANOG expression in bovine blastocysts. Proceedings of the National Academy of Sciences 2018, 115, 2770-2775. [CrossRef]
- Daigneault, B.W.; Rajput, S.; Smith, G.W.; Ross, P.J. Embryonic POU5F1 is Required for Expanded Bovine Blastocyst Formation. Scientific reports 2018, 8, 7753. [CrossRef]
- Camargo, L.S.A.; Owen, J.R.; Van Eenennaam, A.L.; Ross, P.J. Efficient One-Step Knockout by Electroporation of Ribonucleoproteins Into Zona-Intact Bovine Embryos. Frontiers in genetics 2020, 11, 570069. [CrossRef]
- Zhang, R.; Li, Y.; Jia, K.; Xu, X.; Li, Y.; Zhao, Y.; Zhang, X.; Zhang, J.; Liu, G.; Deng, S.; et al. Crosstalk between androgen and Wnt/β-catenin leads to changes of wool density in FGF5-knockout sheep. Cell Death & Disease 2020, 11, 407. [CrossRef]
- Hu, R.; Fan, Z.Y.; Wang, B.Y.; Deng, S.L.; Zhang, X.S.; Zhang, J.L.; Han, H.B.; Lian, Z.X. RAPID COMMUNICATION: Generation of FGF5 knockout sheep via the CRISPR/Cas9 system12. Journal of Animal Science 2017, 95, 2019-2024. [CrossRef]
- Wang, X.; Cai, B.; Zhou, J.; Zhu, H.; Niu, Y.; Ma, B.; Yu, H.; Lei, A.; Yan, H.; Shen, Q.; et al. Disruption of FGF5 in Cashmere Goats Using CRISPR/Cas9 Results in More Secondary Hair Follicles and Longer Fibers. PLOS ONE 2016, 11, e0164640. [CrossRef]
- Zhang, R.; Wu, H.; Lian, Z. Bioinformatics analysis of evolutionary characteristics and biochemical structure of FGF5 Gene in sheep. Gene 2019, 702, 123-132. [CrossRef]
- Li, G.; Zhou, S.; Li, C.; Cai, B.; Yu, H.; Ma, B.; Huang, Y.; Ding, Y.; Liu, Y.; Ding, Q.; et al. Base pair editing of goat embryos: nonsense codon introgression into <i>FGF5</i> to improve cashmere yield; Genetics: 2018/06/15/ 2018.
- Hao, F.; Yan, W.; Li, X.; Wang, H.; Wang, Y.; Hu, X.; Liu, X.; Liang, H.; Liu, D. Generation of Cashmere Goats Carrying an <i>EDAR</i> Gene Mutant Using CRISPR-Cas9-Mediated Genome Editing. International Journal of Biological Sciences 2018, 14, 427-436. [CrossRef]
- 李晓聪. CRISPR/Cas9介导绒山羊Tβ4基因定点敲入的研究. 硕士, 2017.
- Zhang, X.; Li, W.; Liu, C.; Peng, X.; Lin, J.; He, S.; Li, X.; Han, B.; Zhang, N.; Wu, Y.; et al. Alteration of sheep coat color pattern by disruption of ASIP gene via CRISPR Cas9. Scientific reports 2017, 7, 8149. [CrossRef]
- Wang, X.; Niu, Y.; Zhou, J.; Zhu, H.; Ma, B.; Yu, H.; Yan, H.; Hua, J.; Huang, X.; Qu, L.; et al. CRISPR/Cas9-mediated MSTN disruption and heritable mutagenesis in goats causes increased body mass. Animal genetics 2018, 49, 43-51. [CrossRef]
- Zhang, Y.; Wang, Y.; Yulin, B.; Tang, B.; Wang, M.; Zhang, C.; Zhang, W.; Jin, J.; Li, T.; Zhao, R.; et al. CRISPR/Cas9-mediated sheep <i>MSTN</i> gene knockout and promote sSMSCs differentiation. Journal of cellular biochemistry 2019, 120, 1794-1806. [CrossRef]
- Zhang, J.; Cui, M.L.; Nie, Y.W.; Dai, B.; Li, F.R.; Liu, D.J.; Liang, H.; Cang, M. CRISPR/Cas9-mediated specific integration of fat-1 at the goat MSTN locus. The FEBS journal 2018, 285, 2828-2839. [CrossRef]
- Wang, H.T.; Li, T.T.; Huang, X.; Ma, R.L.; Liu, Q.Y. Application of genetic modification technologies in molecular design breeding of sheep. Yi chuan = Hereditas 2021, 43, 580-600. [CrossRef]
- Alberio, R.; Wolf, E. 25th ANNIVERSARY OF CLONING BY SOMATIC-CELL NUCLEAR TRANSFER: Nuclear transfer and the development of genetically modified/gene edited livestock. Reproduction (Cambridge, England) 2021, 162, F59-f68. [CrossRef]
- He, Z.; Zhang, T.; Jiang, L.; Zhou, M.; Wu, D.; Mei, J.; Cheng, Y. Use of CRISPR/Cas9 technology efficiently targetted goat myostatin through zygotes microinjection resulting in double-muscled phenotype in goats. Bioscience Reports 2018, 38, BSR20180742. [CrossRef]
- Niu, Y.; Jin, M.; Li, Y.; Li, P.; Zhou, J.; Wang, X.; Petersen, B.; Huang, X.; Kou, Q.; Chen, Y. Biallelic <i>β-carotene oxygenase 2</i> knockout results in yellow fat in sheep via <span style="font-variant:small-caps;">CRISPR</span> /Cas9. Animal genetics 2017, 48, 242-244. [CrossRef]
- Wan, Y.; Guo, R.; Deng, M.; Liu, Z.; Pang, J.; Zhang, G.; Wang, Z.; Wang, F. Efficient generation of <i>CLPG1</i> -edited rabbits using the CRISPR/Cas9 system. Reproduction in Domestic Animals 2019, 54, 538-544. [CrossRef]
- Zhou, S.; Cai, B.; He, C.; Wang, Y.; Ding, Q.; Liu, J.; Liu, Y.; Ding, Y.; Zhao, X.; Li, G.; et al. Programmable Base Editing of the Sheep Genome Revealed No Genome-Wide Off-Target Mutations. Frontiers in genetics 2019, 10, 215. [CrossRef]
- Zhou, S.; Kalds, P.; Luo, Q.; Sun, K.; Zhao, X.; Gao, Y.; Cai, B.; Huang, S.; Kou, Q.; Petersen, B.; et al. Optimized Cas9:sgRNA delivery efficiently generates biallelic MSTN knockout sheep without affecting meat quality. BMC Genomics 2022, 23, 348. [CrossRef]
- Wang, L.; Cai, B.; Zhou, S.; Zhu, H.; Qu, L.; Wang, X.; Chen, Y. RNA-seq reveals transcriptome changes in goats following myostatin gene knockout. PLOS ONE 2017, 12, e0187966. [CrossRef]
- 梅珺琰. CRISPR/Cas9技术对山羊MSTN基因的靶向敲除研究. 硕士, 2017.
- Zhou, S.; Yu, H.; Zhao, X.; Cai, B.; Ding, Q.; Huang, Y.; Li, Y.; Li, Y.; Niu, Y.; Lei, A.; et al. Generation of gene-edited sheep with a defined Booroola fecundity gene (FecBB) mutation in bone morphogenetic protein receptor type 1B (BMPR1B) via clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated (Cas) 9. Reproduction, Fertility and Development 2018, 30, 1616. [CrossRef]
- Zhang, X.; Li, W.; Wu, Y.; Peng, X.; Lou, B.; Wang, L.; Liu, M. Disruption of the sheep BMPR-IB gene by CRISPR/Cas9 in in vitro-produced embryos. Theriogenology 2017, 91, 163-172.e162. [CrossRef]
- Tian, X.; Lv, D.; Ma, T.; Deng, S.; Yang, M.; Song, Y.; Zhang, X.; Zhang, J.; Fu, J.; Lian, Z.; et al. <i>AANAT</i> transgenic sheep generated via OPS vitrified-microinjected pronuclear embryos and reproduction efficiency of the transgenic offspring. PeerJ 2018, 6, e5420. [CrossRef]
- Zhou, W.; Wan, Y.; Guo, R.; Deng, M.; Deng, K.; Wang, Z.; Zhang, Y.; Wang, F. Generation of beta-lactoglobulin knock-out goats using CRISPR/Cas9. PLOS ONE 2017, 12, e0186056. [CrossRef]
- Wei, J.; Wagner, S.; Maclean, P.; Brophy, B.; Cole, S.; Smolenski, G.; Carlson, D.F.; Fahrenkrug, S.C.; Wells, D.N.; Laible, G. Cattle with a precise, zygote-mediated deletion safely eliminate the major milk allergen beta-lactoglobulin. Scientific reports 2018, 8, 7661. [CrossRef]
- Tian, H.; Luo, J.; Zhang, Z.; Wu, J.; Zhang, T.; Busato, S.; Huang, L.; Song, N.; Bionaz, M. CRISPR/Cas9-mediated Stearoyl-CoA Desaturase 1 (SCD1) Deficiency Affects Fatty Acid Metabolism in Goat Mammary Epithelial Cells. Journal of Agricultural and Food Chemistry 2018, 66, 10041-10052. [CrossRef]
- Zhang, Y.; Wang, Y.; Wang, X.; Ji, Y.; Cheng, S.; Wang, M.; Zhang, C.; Yu, X.; Zhao, R.; Zhang, W.; et al. Acetyl-coenzyme A acyltransferase 2 promote the differentiation of sheep precursor adipocytes into adipocytes. Journal of cellular biochemistry 2019, 120, 8021-8031. [CrossRef]
- Ma, T.; Tao, J.; Yang, M.; He, C.; Tian, X.; Zhang, X.; Zhang, J.; Deng, S.; Feng, J.; Zhang, Z.; et al. An AANAT/ASMT transgenic animal model constructed with CRISPR/Cas9 system serving as the mammary gland bioreactor to produce melatonin-enriched milk in sheep. Journal of pineal research 2017, 63. [CrossRef]
- Fan, Z.; Yang, M.; Regouski, M.; Polejaeva, I.A. Gene Knockouts in Goats Using CRISPR/Cas9 System and Somatic Cell Nuclear Transfer. In Microinjection, Liu, C., Du, Y., Eds.; Springer New York: New York, NY, 2019; Volume 1874, pp. 373-390.
- Abstracts from the UC Davis Transgenic Animal Research Conference XI : August 13-17, 2017. Transgenic research 2018, 27, 467-487. [CrossRef]
- Menchaca, A.; Dos Santos-Neto, P.C.; Souza-Neves, M.; Cuadro, F.; Mulet, A.P.; Tesson, L.; Chenouard, V.; Guiffès, A.; Heslan, J.M.; Gantier, M.; et al. Otoferlin gene editing in sheep via CRISPR-assisted ssODN-mediated Homology Directed Repair. Scientific reports 2020, 10, 5995. [CrossRef]
- Li, X.; Hao, F.; Hu, X.; Wang, H.; Dai, B.; Wang, X.; Liang, H.; Cang, M.; Liu, D. Generation of Tβ4 knock-in Cashmere goat using CRISPR/Cas9. International Journal of Biological Sciences 2019, 15, 1743-1754. [CrossRef]
- Program and Abstracts of the 14th Transgenic Technology Meeting (TT2017) : Snowbird Resort, Salt Lake City, Utah, USA, 1-4 October 2017. Transgenic research 2017, 26, 1-45. [CrossRef]
- Williams, D.K.; Pinzón, C.; Huggins, S.; Pryor, J.H.; Falck, A.; Herman, F.; Oldeschulte, J.; Chavez, M.B.; Foster, B.L.; White, S.H.; et al. Genetic engineering a large animal model of human hypophosphatasia in sheep. Scientific reports 2018, 8, 16945. [CrossRef]
- Vilarino, M.; Suchy, F.P.; Rashid, S.T.; Lindsay, H.; Reyes, J.; McNabb, B.R.; Van Der Meulen, T.; Huising, M.O.; Nakauchi, H.; Ross, P.J. Mosaicism diminishes the value of pre-implantation embryo biopsies for detecting CRISPR/Cas9 induced mutations in sheep. Transgenic research 2018, 27, 525-537. [CrossRef]
- De Los Angeles, A.; Pho, N.; Redmond, D.E., Jr. Generating Human Organs via Interspecies Chimera Formation: Advances and Barriers. The Yale journal of biology and medicine 2018, 91, 333-342.
- Han, H.A.; Pang, J.K.S.; Soh, B.-S. Mitigating off-target effects in CRISPR/Cas9-mediated in vivo gene editing. Journal of Molecular Medicine 2020, 98, 615-632. [CrossRef]
- Chen, S.; Yao, Y.; Zhang, Y.; Fan, G. CRISPR system: Discovery, development and off-target detection. Cellular Signalling 2020, 70, 109577. [CrossRef]
- Yip, B. Recent Advances in CRISPR/Cas9 Delivery Strategies. Biomolecules 2020, 10, 839. [CrossRef]
- Wienert, B.; Wyman, S.K.; Richardson, C.D.; Yeh, C.D.; Akcakaya, P.; Porritt, M.J.; Morlock, M.; Vu, J.T.; Kazane, K.R.; Watry, H.L.; et al. Unbiased detection of CRISPR off-targets in vivo using DISCOVER-Seq. Science 2019, 364, 286-289. [CrossRef]
- Donohoue, P.D.; Pacesa, M.; Lau, E.; Vidal, B.; Irby, M.J.; Nyer, D.B.; Rotstein, T.; Banh, L.; Toh, M.S.; Gibson, J.; et al. Conformational control of Cas9 by CRISPR hybrid RNA-DNA guides mitigates off-target activity in T cells. Molecular Cell 2021, 81, 3637-3649.e3635. [CrossRef]
- Ali, A.; Aslam, S.; Tabasum, S.; Aslam, R. Overview of Delivery of CRISPR/Cas Systems, Its Types and Role in Genome Editing and Immunotherapy. 2021.
- Mout, R.; Ray, M.; Lee, Y.-W.; Scaletti, F.; Rotello, V.M. In Vivo Delivery of CRISPR/Cas9 for Therapeutic Gene Editing: Progress and Challenges. Bioconjugate Chemistry 2017, 28, 880-884. [CrossRef]
- Perez, A.R.; Sala, L.; Perez, R.K.; Vidigal, J.A. CSC software corrects off-target mediated gRNA depletion in CRISPR-Cas9 essentiality screens. Nature Communications 2021, 12, 6461. [CrossRef]
- 李国玲; 杨善欣; 吴珍芳; 张献伟. 提高CRISPR/Cas9介导的动物基因组精确插入效率研究进展 %J 遗传. 2020, 42, 641-656. [CrossRef]
- Schaefer, K.A.; Wu, W.-H.; Colgan, D.F.; Tsang, S.H.; Bassuk, A.G.; Mahajan, V.B. Unexpected mutations after CRISPR–Cas9 editing in vivo. Nature Methods 2017, 14, 547-548. [CrossRef]
- Wang, S.; Qu, Z.; Huang, Q.; Zhang, J.; Lin, S.; Yang, Y.; Meng, F.; Li, J.; Zhang, K. Application of Gene Editing Technology in Resistance Breeding of Livestock. Life 2022, 12, 1070. [CrossRef]
- Raza, S.H.A.; Hassanin, A.A.; Pant, S.D.; Bing, S.; Sitohy, M.Z.; Abdelnour, S.A.; Alotaibi, M.A.; Al-Hazani, T.M.; Abd El-Aziz, A.H.; Cheng, G.; et al. Potentials, prospects and applications of genome editing technologies in livestock production. Saudi Journal of Biological Sciences 2022, 29, 1928-1935. [CrossRef]
- 许美娜; 朱奕舟; 林思远; 陈瑶生; 何祖勇. CRISPR/Cas9基因编辑技术在猪育种中的研究进展 %J 广东农业科学. 2022, 49, 87-96. [CrossRef]
Table 1.
Comparison of advantages and disadvantages of CRISPR/Cas9, TALEN and ZFN gene editing technologies.
Table 1.
Comparison of advantages and disadvantages of CRISPR/Cas9, TALEN and ZFN gene editing technologies.
Characteristic |
CRISPR-Cas9 |
TALEN |
ZFN |
Price |
Low |
High |
Low |
Precision |
Pinpoint |
Moderate |
Low |
Combination mode |
RNA-DNA |
Protein-DNA |
Protein-DNA |
Design and construction |
Easy |
Difficulty |
Moderate |
Target fragment size |
20-50bp |
30-40bp |
18-36bp |
Application |
Wide |
Small |
Small |
Off-target effect |
High |
Low |
Low |
|
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).