CN111334529B - Method for preparing accurate BLG gene knockout cattle by using third generation base editor - Google Patents

Method for preparing accurate BLG gene knockout cattle by using third generation base editor Download PDF

Info

Publication number
CN111334529B
CN111334529B CN202010426973.7A CN202010426973A CN111334529B CN 111334529 B CN111334529 B CN 111334529B CN 202010426973 A CN202010426973 A CN 202010426973A CN 111334529 B CN111334529 B CN 111334529B
Authority
CN
China
Prior art keywords
gene
blg
knockout
seq
cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202010426973.7A
Other languages
Chinese (zh)
Other versions
CN111334529A (en
Inventor
戴蕴平
王明
孙照霖
丁方荣
王海萍
李玲
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
China Agricultural University
Original Assignee
China Agricultural University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by China Agricultural University filed Critical China Agricultural University
Priority to CN202010426973.7A priority Critical patent/CN111334529B/en
Publication of CN111334529A publication Critical patent/CN111334529A/en
Application granted granted Critical
Publication of CN111334529B publication Critical patent/CN111334529B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
    • A01K67/027New or modified breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/873Techniques for producing new embryos, e.g. nuclear transfer, manipulation of totipotent cells or production of chimeric embryos
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/101Bovine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/02Animal zootechnically ameliorated

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Environmental Sciences (AREA)
  • Molecular Biology (AREA)
  • Plant Pathology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • Biodiversity & Conservation Biology (AREA)
  • Animal Husbandry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Developmental Biology & Embryology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The invention discloses a method for preparing an accurate BLG gene knockout cattle by using a third generation base editor. The preparation method of the protective gene knockout dairy cow comprises the following steps: mutating 277 th basic group from C to T of the 5' end of the BLG gene in the genome of the starting dairy cow; the BLG gene is a DNA molecule shown in SEQ ID No. 1. The invention establishes a large animal point mutation technology, can carry out accurate single base mutation, and can achieve the purpose of gene knockout by skillfully designing C-T mutation to cause a stop codon to terminate protein translation in advance. Compared with the traditional gene editing technology, the point mutation technology based on BE3 does not generate genome double-strand break, thereby greatly avoiding random mutation and even genome damage and having higher safety. The invention provides important technical support for accurate gene editing and gene knockout research of cattle.

Description

Method for preparing accurate BLG gene knockout cattle by using third generation base editor
Technical Field
The invention relates to a method for preparing an accurate BLG gene knockout cattle by using a third generation base editor.
Background
Milk is an important nutritionally well-balanced health food for worldwide consumption, comprising abundant proteins, carbohydrates, fats and minerals. Although milk yield has been improved by natural breeding strategies over the years, nutrient management and quantitative genetics, milk composition has not changed significantly. With the development of biotechnology and genetic engineering, the development of animal husbandry provides more opportunities for the preparation of "designer milk" particularly suitable for human beings.
To date, various high quality transgenic cattle have been reported, such as cows expressing recombinant human lactoferrin, cows with increased milk casein content, cows with resistance to mastitis, cows with resistance to mad cow disease, and calves with no horns that improve animal welfare. However, the problem of allergic reactions in milk is not well solved at present.
Beta-lactoglobulin is a main allergen in milk, the milk allergy is most common in infant food allergy, and foreign epidemiological investigation results show that the incidence rate of infant milk protein allergy is 0.3% -3.5%. And tends to rise continuously. The absorption and utilization of the nutrient substances in the dairy products by the infants are seriously influenced. To date, researchers have tried various techniques to reduce the sensitization of BLG in cow's milk, mainly including heat treatment, high pressure, enzymatic, and glycosylation modification, among others. Although these methods reduce the sensitization of BLG to some extent, the structure and function of other proteins in milk are destroyed, the nutritional effect of milk is greatly reduced, and the sensitization of BLG cannot be completely eliminated. How to effectively reduce BLG allergenicity and simultaneously keep the original structure and function of protein as much as possible, no effective and feasible technical method exists at home and abroad at present.
The gene site-directed modification is one of the important means for editing genes, namely, the target gene can be modified and can also be used for treating human genetic diseases, so that the technology becomes a research hotspot of modern molecular biology. However, the gene targeting technology based only on homologous recombination is extremely inefficient and has limited applications. The advent of artificial endonuclease technology has changed this situation. Zinc finger endonucleases (ZFNs) were the first generation of artificial endonucleases. Zinc fingers are proteins capable of binding DNA, about half of the transcription factors of human cells contain zinc finger structures, ZFNs are endonucleases formed by fusing zinc finger proteins with the cleavage domain of the endonuclease Fok1, and it is possible to make double-stranded nicks of DNA at specific positions of various complex genomes using the same. To date, ZFNs have been successfully applied to black-tailed monkeys, rats, mice, chinese hamsters, zebrafish, drosophila, sea urchins, silkworms, arabidopsis thaliana, tobacco, corn, pigs, cattle, human iPS cells. However, ZFNs are complex to prepare, expensive, and their technical patents are controlled by a few commercial companies. Soon, the advent of a second generation artificial nuclease-like transcription activator-like effector nuclease (TALEN) largely replaced ZFNs. In 2009, scientists deciphered the base correspondence between transcription activator-like effectors (TALEs) encoded by pathogenic bacteria (Xanthamonas) of rice and DNA. In 2010, the successful application of the TALEN protein in yeast is reported for the first time, and then the TALEN protein is rapidly applied to plants, human cells, mice, zebra fish, pigs and cattle. TALEN can carry out meticulous modification to complicated genome like ZFN, and its structure is comparatively simple simultaneously, and the specificity is higher, has consequently received scientific research worker's favor. In 2012, TALENs were rated as one of ten scientific breakthroughs by the journal of science. The principles of the two artificial nucleases are the same regardless of ZFN or TALEN, and the two artificial nucleases are formed by fusing DNA binding protein and endonuclease Fok1, so that a large amount of vector construction work is required, and the multi-gene knockout operation is difficult to perform.
In the beginning of 2013, a brand-new artificial endonuclease (CRISPR)/CRISPR-associated) Cas9 appears, which is mainly formed by modifying an acquired immune system based on bacteria, and has the characteristics of simple manufacture, low cost and high effect. Two subject groups of Feng Zhang and George m. Church of MIT in 2013 successfully edited human cells first using this technique and could perform multigene knockout, and then this technique was rapidly applied to mice, zebrafish, nematodes, rats, rabbits, dogs, pigs, cattle and plants. However, the technology only determines the target specificity and has 20bp sequence and off-target phenomenon. Although researchers subsequently upgraded nickase versions of dCas9 and high fidelity Cas9 in this technology to greatly reduce off-target effects, these technologies all utilize DSBs made on host genomic DNA, which are then progressively repaired by NHEJ or HDR, with certain drawbacks. The efficiency of NHEJ is far higher than HDR, however, the repair randomness of NHEJ is strong, random fragment deletion or insertion of a target gene is often caused, and accurate point mutation preparation is difficult to perform; HDR repair, while highly accurate, is extremely inefficient and restricted to mitotic cells, limiting the application of HDR repair. Therefore, the precise point mutation technique remains a big problem. Because most of mutations existing in nature are point mutations, establishing an efficient point mutation technology for repair and quality improvement is significant. The problem group of David R. Liu at Harvard university in the United states is published in Nature, and a single-Base Editor (BE) based on CRISPR/Cas9 is innovatively developed to effectively solve the problems. This group connects rat cytosine nucleoside deaminase rAPOBEC1 converting cytosine (C) to uracil (U) to the N-terminus of catalytically inactive Cas9 (catharticy end Cas9, dCas9) via linker sequence XTEN, constructing first generation single base editor (BE1) (rAPOBEC 1-XTEN-dCas 9), followed by further structural optimization such as adding UGI and restoring a cleavage domain of Cas9 (rAPOBEC 1-XTEN-dCas 9-UGI), successfully preparing third generation single base editor base editors (BE3) that can efficiently mutate C to T at the human cellular level. This technology was subsequently successfully applied in mice, zebrafish and plants.
Disclosure of Invention
The invention aims to provide a method for preparing a precise BLG gene knockout cattle by using a third generation base editor.
In a first aspect, the method for preparing a knockout bovine having a protected gene of the present invention comprises the steps of: mutating 277 th base of BLG gene from 5' end to T; the BLG genes on two homologous chromosomes of the gene knockout cattle have the same point mutation; the BLG gene is a DNA molecule shown in SEQ ID No. 1.
The difference between the gene knockout cattle and the starting cattle is only that single-point mutation is introduced into the BLG gene of the starting cow (namely the 277 th nucleotide of the DNA molecule shown in SEQ ID No.1 from 5' end is changed from C mutation to T mutation, and the two chromosomes are subjected to the same mutation).
Further, the method comprises the steps (1) and (2):
(1) preparing a gene knockout cell: introducing a third generation single base editor BE3 and gRNA for identifying BLG gene into bovine fibroblasts, and preparing gene knockout cells through gene editing; all 277 bases of the BLG genes on two homologous chromosomes of the gene knockout cell from the 5' end are mutated from C to T; the target sequence of the gRNA is shown as 273 to 295 th positions from the 5' end of SEQ ID No. 1;
(2) and (2) cloning the single cell of the gene knockout cell obtained in the step (1) by a somatic cell nuclear transfer technology to obtain the gene knockout cattle.
In the step (2), the gene knockout cell single cell is used as a donor cell to be injected into the bovine oocyte with the cell nucleus removed, a reconstructed embryo is developed and formed, then the reconstructed embryo is transferred into the cow uterus, and the gene knockout cow is obtained through delivery.
Further, the present invention also provides a method for knocking out a BLG gene in a cell, comprising the steps of: introducing a third generation single base editor BE3 and gRNA for identifying BLG gene into a target cell, and preparing a gene knockout cell through gene editing; the 277 th base of the BLG gene on two homologous chromosomes of the gene knockout cell from the 5' end is mutated from C to T; the target sequence of the gRNA is shown as 273 to 295 th positions from the 5' end of SEQ ID No. 1; the BLG gene is a DNA molecule shown in SEQ ID No. 1.
The target cell may be specifically a bovine fibroblast.
Any one of the gRNAs is shown in SEQ ID No. 4. Any one of the above gRNAs can be obtained by in vitro transcription of a template, which is shown in SEQ ID No. 3.
Any one of the above third generation single base editors BE3 is mRNA transcribed in vitro from a vector expressing the third generation single base editor BE3 or a protein translated from the mRNA.
The vector for expressing the third generation single base editor BE3 can BE pCMV-BE3 vector.
The gRNA and the mRNA are introduced into bovine fibroblasts in an electric shock transformation mode, and the mass ratio of the gRNA to the mRNA is 1: 1.
After the gRNA and mRNA are introduced into bovine fibroblasts, a single cell clone (gene knockout positive cell) is prepared by an infinite dilution method.
In a second aspect, the invention provides the use of a method according to any one of the first aspects for breeding cows producing hypoallergenic milk.
The method for breeding the dairy cattle producing hypoallergenic milk comprises the following steps: the gene knockout dairy cow prepared by the method in the first aspect is the dairy cow producing hypoallergenic milk.
In a third aspect, the present invention provides a protection kit comprising any one of the grnas described above, or a gene encoding the gRNA, or an expression cassette containing the encoding gene; the kit is used for preparing gene knockout cattle.
The kit also comprises a vector for expressing the third generation single base editor BE3 or mRNA obtained by in vitro transcription of the vector or protein obtained by translation of the mRNA.
In a fourth aspect, the invention protects the application of the kit in the third invention in breeding cows producing hypoallergenic milk.
In each of the above aspects, the cow may be a holstein cow.
The possibility that an exogenous BE3 plasmid is integrated into a receptor genome exists when a BE3 plasmid is used for preparing a gene editing animal, so that the transfection is carried out by using mRNA of BE3, the problem is avoided, the biological safety problem caused by screening of a marker gene is avoided, and then the BLG double allele C-T point mutation positive cell clone without exogenous DNA integration is finally obtained by using a gene editing method without a screening marker, and the early termination of AA is caused by the mutation of C-T, so that the purpose of BLG gene knockout is achieved. The positive cells are subjected to somatic cloning to prepare the BLG gene knockout cattle with accurate editing. When the cow normally lactates, the experiment proves that the milk of the cow does not contain BLG protein.
The invention establishes a large animal point mutation technology which can carry out accurate single base mutation and achieve the purpose of gene knockout by skillfully designing C-T mutation to cause a stop codon to terminate protein translation in advance. Compared with the traditional gene editing technology, the point mutation technology based on BE3 does not generate genome double-strand break, thereby greatly avoiding random mutation and even genome damage and having higher safety. The invention provides important technical support for accurate gene editing and gene knockout research of cattle.
Drawings
FIG. 1 is a technical scheme of the present invention.
FIG. 2 is a schematic diagram showing the target region sequence of BE3 single base modification technique and the achievement of BLG gene knockout by the single base modification technique of BLG gene.
FIG. 3 shows the results of the sequencing alignment in example 2.
FIG. 4 shows the results of SDS-PAGE in example 3.
FIG. 5 shows the Western blot detection results in example 3.
FIG. 6 is the statistical results of the anal temperature changes of the mice in example 4.
FIG. 7 shows the results of ELISA assay in example 4.
Detailed Description
The following examples are given to facilitate a better understanding of the invention, but do not limit the invention. The experimental procedures in the following examples are conventional unless otherwise specified. The test materials used in the following examples were purchased from a conventional biochemical reagent store unless otherwise specified. The quantitative tests in the following examples, all set up three replicates and the results averaged.
pCMV-BE3 vector: addgen, Cat number: # 73021.
In the pCMV-BE3 vector, the CMV promoter drives the expression of the third generation base editor BE3 (rAPOBEC 1-XTEN-Cas9 n-UGI-NLS).
Example 1 preparation of BLG Diallelic mutant cell lines Using third Generation base editor
This example utilizes the third generation base editor (BE3) to prepare BLG biallelic mutant cell lines. Considering the later industrialization and avoiding the biosafety problem, the invention does not use a screening marker gene and simultaneously uses the mRNA of BE3, so that a BLG biallelic gene mutant cell line without exogenous DNA integration can BE prepared, and the technical route is shown in figure 1. The target region sequence of BE3 single base modification technology and the schematic diagram of BLG gene knockout achieved by the single base modification technology of BLG gene are shown in FIG. 2.
According to the BLG gene sequence (SEQ ID No. 1), the region of the target sequence with base editing is positioned on the exon 1 (SEQ ID No. 2) of the BLG gene sequence, and the binding sequence of the target region is cacccagaccatgaagggccTGG (SEQ ID No.1 at position 273 and 295 from the 5' end,97-119 th from the 5' end of SEQ ID No. 2), wherein tgg is a PAM sequence, and the underlined site is the expected mutation site.
First, preparation of in vitro transcribed BE3 mRNA
1. In vitro transcribed mRNA systems were configured at room temperature (table 1). After the prepared in vitro transcription mRNA system is completely mixed, the mixture is incubated for 1 h at 37 ℃, 1 mu L of TURBO DNase is added into the reaction system, and the incubation is carried out for 15min at 37 ℃.
TABLE 1
Components Volume of
Linearized pCMV-BE3 vector treated with Ahd1 1μL(0.1-1μg)
2×NTP/CAP 10μL
10×Buffer 2μL
RNA synthetase 2μL
ddH2O Make up to 20 mu L
All of the above reagents were obtained from MEGAscript T7 Transcription Kit, Ambion, cat # SEQ ID NO: AM 1333.
2. Taking the reaction product obtained in the step 1, configuring an in vitro transcription mRNA and polyA system (table 2), and incubating for 1 h at 37 ℃ after complete mixing.
TABLE 2
Components Volume of
Reaction product of step 1 20μL
5×E-PAP Buffer 20μL
MnCl2(25mM) 10μL
ATP(10mM) 10μL
E-PAP 4μL
ddH2O 36μL
Total of 100μL
All from Poly (A) labeling Kit, Ambion, cat # K: AM 1350.
3. In vitro transcription of mRNA purification by column chromatography
(1) And (3) adding 350 mu L of binding buffer into the reaction product obtained in the step (2), blowing, sucking and uniformly mixing, transferring the sample into a column, and centrifuging at 10000g for 1min at room temperature.
(2) Discarding the filtrate, re-packing the column, rinsing the column with 500 μ L of eluent, centrifuging at 10000g at room temperature for 1min, and repeatedly rinsing once; the filtrate was discarded and centrifuged on an empty column for 15 s.
(3) The column was placed in a new centrifuge tube, 50. mu.L of the eluent was added to the center of the column, the lid was closed and incubated at 60 ℃ for 10min, and 10000g was centrifuged at room temperature for 1 min.
Both the reagents and the column were from clear ­ Transcription clear-Up Kit, Ambion, cat #: AM 1908.
BE3 mRNA was obtained according to the procedure described above.
Second, preparation of in vitro transcribed gRNA
1. A gRNA in vitro transcription template (SEQ ID No. 3) was synthesized.
In SEQ ID No.3, the T7 promoter is located at positions 1 to 20 from the 5' end, the transcription initiation site is located at position 19, the target sequence is located at positions 21 to 40, and the sgRNA is located at positions 41 to 120.
2. Taking the gRNA in-vitro transcription template obtained in the step 1, configuring an in-vitro transcription system (table 3), and incubating for 2 h at 37 ℃.
TABLE 3
Components Volume of
Form panel 8 μL(2μg)
2×ATP/CTP/UTP/GTP 2/2/2/2μL
10×Reaction Buffer 2 μL
Enzyme Mix
2 μL
Total of 20 μL
3. After completion of step 2, 1. mu.L of TURBO DNase was added to the reaction system and incubated at 37 ℃ for 15 min.
4. After the step 3 is completed, 2 times of volume of absolute ethyl alcohol is added to precipitate the transcription product, the transcription product is kept stand at the temperature of minus 20 ℃ for 30min, the transcription product is centrifuged at 13000rpm for 30min, the supernatant is discarded, and the precipitate is taken.
5. And (3) fully washing the precipitate obtained in the step (4) by using 75% (v/v) ethanol water solution, removing supernatant, fully drying the precipitate by using an ultra-clean bench, and fully dissolving the gRNA (the gRNA sequence is shown as SEQ ID No. 4) by using nucleic-free water.
All of the above reagents were obtained from MEGASHORTCRIPT-T7 Transcription Kit, Ambion, cat #: AM 1354.
Third, obtaining bovine fibroblasts
Collecting ear skin tissue of Holstein cow, removing hair from lower edge back side of ear, cleaning with 70% (v/v) ethanol water solution, and picking with blade with area of 1cm2The skin was transported to the laboratory as soon as possible in DMEM/F12 medium at 0 deg.C, washed several times with PBS buffer and 70% (v/v) alcohol in water and chopped to 1mm3The left and right small blocks were washed with DMEM medium for 2 times and then planted in batches in 1mL DMEM medium flasks (25 cm in specification) containing 10% (v/v) FBS2) After the tissue blocks adhere firmly, the DMEM medium containing 10% (v/v) FBS is supplemented to 6mL, the temperature is 37 ℃, and the CO content is 5 percent2The culture box is cultured for 6-7 days, the culture solution is changed 1 time every 2 days, after the cells grow and are confluent, the cells are digested and passaged for 2-3 times by 0.25% trypsin, and the cells are frozen in DMEM culture medium containing 20% (v/v) FBS and 10% (v/v) DMSO in batches. Thus, the Holstein cow fibroblast cell line is established through in vitro culture operations such as primary culture, subculture, freezing and the like.
IV, BE3 mRNA transfection
(1) 2 days before transfection, the bovine fibroblast cell line obtained in step three was trypsinized into single cells, and 1 × 10 was added6The bovine fibroblasts were transferred to a culture flask (100 mL), 4mL of DMEM medium containing 10% (v/v) fetal bovine serum was added thereto, and the mixture was incubated at 37 ℃ with 5% CO2Culturing to logarithmic growth phase.
(2) After the step (1) is completed, taking bovine fibroblasts in logarithmic growth phase, digesting the bovine fibroblasts with trypsin, centrifuging the bovine fibroblasts for 5min at 1000g, and collecting precipitates.
(3) After completion of step (2), the pellet was taken, washed 1 time with PBS buffer, and then resuspended with 100 μ L of shock solution to obtain a cell suspension.
(4) Taking cell suspension (containing 1 × 10)6Cell), 4. mu.g of BE3 mRNA obtained in the first step and 4. mu.g of gRNA obtained in the second step were thoroughly mixed and transferred into an electric shock cup for electric shock (electric field strength 1.2KV/cm, pulse time 1 ms).
(5) After the step (4) is completed, the cells after electric shock are transferred into a culture bottle (the specification is 100 mL), 4mL of DMEM culture solution containing 10% (v/v) fetal calf serum is added, the temperature is 37 ℃, and 5% CO is added2Culturing for 1-2 days to obtain transfected cells.
Five, infinite dilution method for preparing single cell clone and genotype identification
(1) And (3) culturing the transfected cells obtained in the fourth step until the fusion rate reaches 80-90%, digesting the cells by using 0.1% pancreatin at 37 ℃, stopping digestion by using a DMEM medium containing 10% (v/v) FBS, and centrifuging to collect the cells.
(2) The cells were resuspended in a DMEM medium containing 20% (v/v) FBS, a portion of the cells were counted, and the cells were diluted to 100 cells/mL to obtain a cell suspension. To 20 dishes to which 9mL of 20% (v/v) FBS-containing DMEM medium had been added, 1mL of each cell suspension was added at 37 ℃ with 5% CO2And culturing under saturated humidity condition.
(3) When the cell clone in the culture dish grows to be more than 2mm in diameter, removing the culture medium, washing with DPBS, covering the clone cluster with a clone ring, dripping about 100 mu L of 0.1% pancreatin at 37 ℃ into the clone cluster, digesting for about 3min, dripping DMEM culture medium containing 20% (v/v) FBS to stop digestion, slightly blowing and then transferring the cell clone into a 48-well plate for culture expansion.
(4) When the cell fusion rate in the 48-well plate reaches 90%, half of the cells are taken for cell clone genotype identification after digestion, and the rest half are continuously cultured in the well plate.
(5) After the cells for identifying the genotype are centrifuged for 5min at 1000g, the supernatant is discarded, and 10-20 mul of cell lysate is added according to the amount of the cell precipitate.
(6) mu.L of cell lysate was pipetted as a template for PCR identification, and PCR amplification was performed using a primer pair consisting of primer P1 and primer P2 (designed and synthesized based on the bovine BLG gene).
Primer P1: 5'-AGGCCTCCTATTGTCCTCGT-3', respectively;
primer P2: 5'-GCAAAGGACACAGGGAGAAG-3' are provided.
The reaction system was 20. mu.L, consisting of 1.0. mu.L of DNA template, 0.4. mu.L of primer P1 (10. mu.M), 0.4. mu.L of primer P2 (10. mu.M), 0.4. mu.L of dNTP, 0.3. mu.L of LA DNA polymerase, 2.0. mu.L of 10 XPCR Buffer, and 15.5. mu.L of ddH 2O.
Reaction procedure: 5min at 94 ℃; 30s at 94 ℃, 30s at 52 ℃, 1min at 72 ℃ and 35 cycles; preserving at 72 deg.C for 5min and 4 deg.C.
And purifying and sequencing the PCR amplification product. If a mutation is generated at the target site, a bimodal phenomenon appears in the sequencing result, and the cell line is determined to be a mutant. And connecting the PCR product of the mutant to a pMD-19t plasmid vector, transforming the competence of escherichia coli, selecting a plurality of single colonies for sequencing, and comparing the sequencing result with the wild type BLG gene to obtain detailed mutation information.
The results showed that of the 32 single cell clones, one biallelic C-T mutation was obtained and AA prematurely terminated. The sequencing is carried out, and the BLG gene in the genome of the wild-type fibroblast is the wild-type BLG gene (shown in SEQ ID No. 1). The BLG gene in the genome of mutant fibroblast cells differs from the wild-type BLG gene only in that a single point mutation (i.e., the 277 th nucleotide from the 5' end of the DNA molecule shown in SEQ ID No.1 is mutated from C to T, and the same mutation occurs in both chromosomes) is introduced into the wild-type BLG gene.
Example 2 preparation of BLG double allele knock-out cattle
1. Taking the mutant fibroblast cells in logarithmic growth phase, and digesting with 0.25% trypsin for 5min to obtain single cells.
2. Collecting ovaries of adult Holstein cows from slaughter houses, cleaning in PBS (phosphate buffer solution) at 37 ℃ for three times, extracting follicles with the diameter of 2-8mm by using a needle with the diameter of 0.7mm, recovering cumulus-oocyte-complex (COCs) with uniform shape and compact structure, washing twice by using mature liquid (M199+10% FBS +0.01U/mL bFSH +0.01U/mL bLH +1 mu g/mL estradiol), putting the cumulus-oocyte-complex into a four-well plate containing the mature liquid at 38.5 ℃ and 5% CO for 50-60 holes, and washing by using a washing machine2After the mature culture in the incubator for 18-20h, placing the mature oocyte in a tube of 0.1% hyaluronidase, oscillating for 2-3min, and lightly blowing and beating with a glass tube to completely separate the cumulus cell from the oocyte, and selecting the oocyte with complete shape, uniform cytoplasm and discharged first polar body as a cytoplasmic receptor.
Transferring the oocyte with the first polar body into an operation liquid containing M199+10% FBS +7.5 mug/mL cytochalasin B, cutting a small opening on the upper part of the polar body by using a glass needle under a 200-time microscope, sucking off chromosomes in the first polar body and the oocyte under the first polar body by using a glass tube with the inner diameter of 20 mug, washing the first polar body and the oocyte under the first polar body for three times by using a solution of M199+20% FBS, and placing the first polar body in an incubator for later use.
3. Transferring the single cell obtained in the step 1 into an enucleated oocyte transparent band, firstly placing the enucleated oocyte transparent band into Zimmerman liquid (100 mL Zimmerman liquid is composed of sucrose 0.9854g, magnesium acetate tetrahydrate 10.7mg, calcium acetate monohydrate 1.8mg, dipotassium hydrogen phosphate 7.4mg, reduced glutathione 3.1mg, bovine serum albumin 1.0mg and water) for balancing for 3-5min, then placing the enucleated oocyte transparent band into a fusion tank to rotate the oocyte to enable donor cells to be in contact with the oocyte prepared in the step 2 to be vertical to an electric field, simultaneously, rapidly transferring the oocyte into M199 liquid (product of Gibco company) containing 10% (v/v) FBS in a direct current pulse field with the field intensity of 2.5kV/cm under the conditions that the pulse time is 10 mu s, the pulse frequency is 2 times and the pulse interval is 1s, and culturing for 30min at 37 ℃ and 5% CO2 to obtain the embryo.
4. Taking the reconstructed embryo obtained in the step 3, adding CR1aa culture solution (100 mL of CR1aa culture solution is composed of 0.67g of sodium chloride, 0.023g of potassium chloride, 0.22g of sodium bicarbonate, 2mg of sodium pyruvate, 100 mul of phenol red and water) of calcium ionophore A23178 with the concentration of 5 mul, and treating for 5 min; discarding the liquid phase, adding CR1aa culture solution containing 5 μ g/mL cytochalasin B and 10 μ g/mL cycloheximide, and treating for 5h (for activating recombinant embryo); discarding the liquid phase, adding 5% (v/v) FBS-containing CR1aa culture solution, 37 deg.C, 5% CO2Culturing for 48h, observing the cleavage rate, and observing the blastocyst development rate in 7-8 days.
5. The morphologically excellent transgenic cloned blastocysts cultured for 7 days in step 4 were transferred into the uterine horns of the contemporary recipient cows (3-head recipient cow was co-transplanted), and BLG double allele knock-out cows (i.e., No. #170923 cows) were obtained.
The genomic DNA of the ear tissue of a cattle to be detected (cattle No. 170923 or a wild Holstein cow) is extracted by adopting a blood and tissue genomic kit (QIAGEN company), and is used as a template, and a primer pair consisting of a primer P1 and a primer P2 (shown in Table 1) is adopted for PCR amplification, so that a PCR amplification product is obtained. And purifying and sequencing the PCR amplification product.
Sequencing results show that the genotype of a wild Holstein cow (i.e. a non-transgenic cow) based on the BLG gene is wild type (BLG +/+), and the genotype of a cattle #170923 based on the BLG gene is biallelic mutant type (BLG-/-). The biallelic mutant is characterized in that 277 nucleotides of wild-type two homologous chromosomes (the wild-type BLG gene is shown as a sequence 1 in a sequence table) are mutated from C to T from 5' end, and the transcription and translation of mRNA of the BLG gene are terminated early due to the base mutation of C-T, so that the function of BLG protein is lost.
The sequencing alignment results are shown in FIG. 3.
Cattle #170923 survived normally at present, see FIG. 3.
Example 3 analysis of BLG protein expression in milk from BLG double allele knock-out cattle
First, SDS-PAGE detects whether BLG protein exists in milk of BLG double allele knock-out cattle
1. Extracting total protein from milk produced from 1 st to 7 th days of lactation period of cattle to be detected (BLG double allele knock-out cattle or wild Holstein cow).
2. And (3) taking the total protein of the cattle to be detected obtained in the step (1) and a BLG protein standard (a product of Sigma company) respectively, and performing SDS-PAGE.
The results are shown in FIG. 4 (M is protein Marker and BLG is BLG protein standard). The results show that the total protein of the BLG double allele knock-out cattle has no expression of BLG protein detected, and the milk produced by the BLG double allele knock-out cattle has no BLG protein, so that the BLG gene is knocked out at a DNA level, and the BLG is also effectively removed at a protein level.
Secondly, detecting whether BLG protein exists in milk of cattle with BLG double allele knockout by Western blot
1. Extracting total protein from milk produced on 1 st, 3 rd, 5 th or 7 th day of lactation period of cattle (BLG double allele knock-out cattle or wild Holstein cow) to be detected.
2. And (3) respectively taking the total protein of the cattle to be detected obtained in the step (1) and the BLG protein standard substance, and carrying out Western blot. The BLG protein was detected using a Bovine beta-lactoglobulin Antibody (product of BETHYL corporation) as a primary Antibody and a goat Antibody (product of China fir Jinqiao corporation) as a secondary Antibody.
The results of the experiment are shown in FIG. 5 (in FIG. 5, BLG is a standard BLG protein). The results show that the total protein of the BLG double allele knock-out cattle can not detect the expression of the BLG protein, and the milk produced by the BLG double allele knock-out cattle can not have the BLG protein.
The results show that the milk produced by the BLG gene disruption of the cattle with the BLG double allele knockout does not contain BLG protein.
Example 4 sensitization assay of BLG double allele knock-out cow milk
To further verify whether the milk produced by BLG knockout cattle had low allergenicity, the milk sample from #170923 was made into dry powder for later sensitization experimental studies, while milk powder from wild type milk was used as a control.
Animal model: clean-grade female Balb/c mice, Beijing Wittingle laboratory animal technology Ltd, certification number SCXK (Jing) 2012-0001.
The clean-grade common feed is prepared by mixing the levels of main nutritional ingredients and processing into pellet feed; then pass through60Co radiation sterilization is carried out, so that the feed reaches a clean level. The level of the clean-grade common feed reaches the experimental animal nutrition standard listed in GB 14924-2010.
Feeding conditions are as follows: the experiment is carried out in SPF animal houses of food and nutrition engineering college of China agricultural university, and the qualification number is as follows: SYXK (Kyoto) 2010-0036. The environment temperature is 22-25 ℃, the relative humidity is 40% -60%, the artificial illumination/dark cycle is carried out for 12 h, and the ventilation frequency is 15 times/h. Animals in the same group were fed 4 animals per cage, with free access to food and water.
Taking 96 clean-grade female Balb/c mice with the weight of 16-20g, after the mice are fed with clean-grade common feed adaptively for 5 days, randomly dividing the mice into 8 groups of a positive control group, a negative control group, a non-transgenic milk powder group 1, a non-transgenic milk powder group 2, a non-transgenic milk powder group 3, a beta-LG double allele knockout milk powder group 1, a beta-LG double allele knockout milk powder group 2 and a beta-LG double allele knockout milk powder group 3 according to the weight, and carrying out the following treatment (the average weight of the mice needs to be weighed before each gastric lavage for adjusting the concentration of gastric lavage fluid):
positive control group (allergen Ovalbumin (OVA) solution): each mouse was gavaged orally with 0.2 mL OVA solution on day 0, 7, 14, 21 and 28 of the experiment, respectively; on day 42 of the experiment, each mouse was gavaged with 2 mL OVA solution orally; the concentration of OVA solution is 5 mg/mL, the solvent is PBS buffer solution with pH7.2 and 0.01 mM;
negative control group (non-allergen PBS buffer): on experiment days 0, 7, 14, 21 and 28, respectively, each mouse was gavaged with 0.2 mL PBS buffer per oral route; on day 42 of the experiment, each mouse was gavaged with 2 mL PBS buffer per mouth;
non-transgenic milk powder group 1: on the 0 th day, the 7 th day, the 14 th day, the 21 st day and the 28 th day of the experiment, each mouse was orally gavaged with 0.2 mL of wild type non-transgenic milk powder solution (total protein content in wild type non-transgenic milk powder is used as a test object, and the dosage is 0.5 mg total protein/g BW); on the 42 th day of the experiment, each mouse was orally gavaged with 0.2 mL of wild type non-transgenic milk powder solution (total protein content in wild type non-transgenic milk powder is used as a test substance, and the dosage is 5 mg total protein/g BW);
non-transgenic milk powder group 2: on the 0 th day, the 7 th day, the 14 th day, the 21 st day and the 28 th day of the experiment, each mouse was orally gavaged with 0.2 mL of wild type non-transgenic milk powder solution (total protein content in wild type non-transgenic milk powder is used as a test object, and the dosage is 1mg total protein/g BW); on the 42 th day of the experiment, each mouse was orally gavaged with 0.2 mL of wild type non-transgenic milk powder solution (total protein content in wild type non-transgenic milk powder is used as a test substance, and the dosage is 10mg total protein/g BW);
non-transgenic milk powder group 3: on the 0 th day, the 7 th day, the 14 th day, the 21 st day and the 28 th day of the experiment, each mouse was orally gavaged with 0.2 mL of wild type non-transgenic milk powder solution (total protein content in wild type non-transgenic milk powder is used as a test object, and the dosage is 2mg total protein/g BW); on the 42 th day of the experiment, each mouse was orally gavaged with 0.2 mL of wild type non-transgenic milk powder solution (total protein content in wild type non-transgenic milk powder is used as a test substance, and the dosage is 20 mg total protein/g BW);
BLG biallelic knockout milk powder group 1: on the 0 th day, the 7 th day, the 14 th day, the 21 st day and the 28 th day of the experiment, each mouse is orally gavaged with 0.2 mL of BLG double allele knockout milk powder solution (the total protein content in the BLG double allele knockout milk powder is used as a test object, and the dosage is 0.5 mg of total protein/g BW); on the 42 th day of the experiment, each mouse was orally gavaged with 0.2 mL of BLG double allele knockout milk powder solution (total protein content in BLG double allele knockout milk powder is used as a test substance, and the dose is 5 mg total protein/g BW);
BLG double allele knock-out milk powder group 2: orally intragastrically perfusing 0.2 mL of BLG double allele knockout milk powder solution (total protein content in BLG double allele knockout milk powder is used as a test object, and the dosage is 1mg total protein/g BW) for each mouse on days 0, 7, 14, 21 and 28 respectively; on the 42 th day of the experiment, each mouse was orally gavaged with 0.2 mL of BLG double allele knockout milk powder solution (total protein content in BLG double allele knockout milk powder is used as a test substance, and the dose is 10mg total protein/g BW);
BLG biallelic knockout milk powder group 3: orally intragastrically perfusing 0.2 mL of BLG double allele knockout milk powder solution (total protein content in BLG double allele knockout milk powder is taken as a test object, and the dosage is 2mg total protein/g BW) for each mouse on days 0, 7, 14, 21 and 28 respectively; on the 42 th day of the experiment, each mouse was orally gavaged with 0.2 mL of BLG double allele knock-out milk powder solution (total protein content in BLG double allele knock-out milk powder was used as the test substance, and the dose was 20 mg total protein/g BW).
Allergen Ovalbumin (OVA), purchased from sigma.
During the experiment, the growth conditions of the mice (including the appearance, the feed intake amount, the mental state, and the like of the mice) were observed every week.
On day 42 of the experiment, the body temperature change of the mice before and after the stimulation was measured using an electronic thermometer with a probe coated with appropriate vaseline. The results are shown in FIG. 6. The results showed that mice in each protein-treated group, except negative control group non-allergenic PBS, had a significant decrease in anal temperature after stimulation (. about.p < 0.05). However, the temperature drop of the wild non-transgenic milk powder is higher, and the temperature drop of the BLG double allele knock-out milk powder after treatment is lower than that of the positive control milk powder and the common milk powder group, and the result proves that the sensitization of the BLG double allele knock-out milk powder is reduced compared with that of the wild non-transgenic milk powder.
Subsequently, the mice were tested by ELISA for the important immunoglobulin IgE at day 42 (purchased from Thermo, PA 1-84764).
The results are shown in FIG. 7. The result shows that the immunoglobulin level of the positive control group is obviously higher than that of the negative control group (P < 0.05), and the successful establishment of the sensitization animal model is proved. Meanwhile, 3 doses of the BLG double allele knock-out milk powder group all caused a significant increase in IgE level, but the IgE level was low compared with that of the wild type non-transgenic milk powder and the positive control group, and these results indicate that the allergenicity of the BLG double allele knock-out milk powder is lower than that of the wild type non-transgenic milk powder.
Sequence listing
<110> university of agriculture in China
<120> method for preparing accurate BLG gene knockout cattle by using third generation base editor
<160>4
<170>SIPOSequenceListing 1.0
<210>1
<211>4894
<212>DNA
<213> Artificial Sequence (Artificial Sequence)
<400>1
tcctgcttgg cctcgaatgg aagaaggcct cctattgtcc tcgtagagga agcaacccca 60
gggcccaagg ataggccagg ggggattcgg ggaaccgcgt ggctgggggc ccggcccggg 120
ctggctggct ggccctcctc ctgtataagg ccccgagccc actgtctcag ccctccactc 180
cctgcagagc tcagaagcgt gaccccagct gcagccatga agtgcctcct gcttgccctg 240
gccctcactt gtggcgccca ggccctcatt gtcacccaga ccatgaaggg cctggatatc 300
cagaaggttc gagggtgccc gggtgggtgg tgagttgcag ggcaggcagg ggagctgggc 360
ctcagagacc aagggaggct gtgacgtctg ggattcccat cagtcagcta gagccgcctg 420
acaaatcgcc cgccagggca gcttcaacca ggcgtttagt gtcttgcatt ctggaggctg 480
gaagcctgca atccgggcat cggcccagct ggcttctcct gcggccactc tccggggagc 540
agacagccat cttctccctg tgtcctttgc gtgccctggt ttcctcttcc tgtgaggtca 600
ccaggcctgc tggatccacg cccgcccaca cagcctcacg taacctttgt catctcttta 660
aaggccgtgt ctccagtcct gtgttgaggt tctgggggtt aatgggacac agttcagccc 720
ctaaaagagt cactctgccc ctcaaatttt ccccacctcc agctatgtct ccccaagatc 780
caaatgttgc cacgtgtgcg ggggctcatc tgggtccctc tttgggctca gagtgagtct 840
ggggagagca ttcctcaggg tgccgagttg gggggagcat ctcagggctg cccaggccag 900
ggtgggacag agagcccact gtggggctgg gggccccttc ccgcccctgg agtgcagctc 960
aaggtccctc cccaggtggc ggggacttgg tactccttgg ccatggcggc cagcgacatc 1020
tccctgctgg acgcccagag tgcccccctg agagtgtatg tggaggagct gaagcccacc 1080
cctgagggcg acctggagat cctgctgcag aaatggtggg cgtccccccc aaaaaaagca 1140
tggaaccccc actccccagg gatatggacc cccccggggt ggggtgcagg agggaccagg 1200
gccccagggc tggggaacgg ggcttggagt ttcctggtac ccctggaggt ccacccaagg 1260
ctgcttatcc agggctttct ctttcttttt ttcccccaac ttttattaat ttgatgcttc 1320
agaacatcat caaacaaatg aacacaaaac atcattttcg ttaacttgga aggggagata 1380
aaatccactg aagtggaaat gcataggaaa gatacataca gtaaggcagg tattctgaat 1440
tcgctgttag tttgaggatt acaaatgcac ttgagcaaca gagagacgtt ttcattattt 1500
ctggtctgaa cagctcagta tctaaaatga acaagatgtc atggagacaa agccggcggg 1560
ggagaggccc gtgtgaaggc cgctgggcgg ctgcagacct gggtcctcgg ggcccaggca 1620
gttcccacta ccagccctgt ccaccctcag acgggggtca gagtgcagga gagagctggg 1680
tgggtgtggg ggcagagatg gggacctgaa ccccaggact gccttttggg gtgcctgtgg 1740
tcaaggctct ccccaacctt ttctccctgg ctccatctga cttctcctgg cccatccacc 1800
cggtcacctg tggccccaga ggtgacagtg agtgcagcca aggccggttg gccagccggc 1860
cccctatgcc cacgccaccc gcctccagcc cctcctgggg ccgccttctg cccctggccc 1920
tcagttcatc ctgatgaaaa tggtccatgc ccgtggctca gaaagcagct gtctttcagg 1980
gagaacggtg agtgtgctca gaagaagatc attgcagaaa aaaccaagat ccctgcggtg 2040
ttcaagatcg atggtgagtg ctgggtcccc aggggacgcc caccaccccc cagggactgt 2100
gggcaggtgc agggggctgg cgtcaggccc cgagatgcta aggggctggt ggtgatgaag 2160
acactgccgt gccacctgct tccctggcct ccctgccacc tgcccggggc cttggggccg 2220
gtggccgtgg gcaggtcccg gctgggcagg tctgacaccc cagggtgaca cccgagctct 2280
ctttgctgag ggtggggtgg tgctcggggc cctcaggctg agctcaggag gtccctgtgc 2340
ccacccaggg gtaaccgaga gccgctgccc gctccagggg tccaggtgcc ccacgatccc 2400
agcccacccc acggctcctt catctcctga agacgaactc tgtccgccct cgctcattca 2460
cttgtttgtc ctaaatccaa gatgagaaag cttcgaggtg gggttggggt tccatcaggg 2520
cctgcccttc cgccgggcag cctgggccac atctgccctt ggcctcttca ggactcactc 2580
tgactggagg ccctgcactg actgatgcca gggtgcccag cccagggtct cctgtgccat 2640
ccggctgcac ggggtttgga tgctggtcct gcccccaagc tgcccagaca ctgcagggca 2700
gctggggcca cccgcaggcc tcggtcaggg agagccccag ctgcccccgc tcagcgctgc 2760
cccccaacaa ttccccagtc ctcaggacgc atccctcttc ccttgctggg cagtgttcag 2820
ccccacccga gatcggggga agccctattt cttgaccact ccggtccctg ggggagggcg 2880
gcctcagact gagtggtgag tgttcccaag tccaggaggt ggtggagggt ccctggcgga 2940
tccagagttg ggcttccaga gtgagggctt cctgggcccc atgtgcctgg cagtggcagc 3000
agggaagggg ccacaccatt ttggggctgg gggatgccag agggcgctcc ccaccccgtc 3060
ctcaccaagt ggtgaccccg ggggagcccc gctggttgtg gggggcgctg ggggctgacc 3120
agaaaccccc ctcctgctgg aactcacttt cctcctgtct tgatctctac cagccttgaa 3180
cgagaacaaa gtccttgtgc tggacaccga ctacaaaaag tacctgctct tctgcatgga 3240
gaacagtgct gagcccgagc aaagcctggt ctgccagtgc ctgggtgggt gccaaccctg 3300
gctgcccagg gagaccagct gtgtggtcct cgctgcaacg gggccggggg ggacggtggg 3360
agcagggagc ttgattccca ggaggaggag ggatgggggg tccccgagtc ccgccaggag 3420
agggtggtca tataccggga gccggtgtcc tgggggtctg tgggtgactg gggacggggg 3480
ccagacacac aggctgggag acggggggct gcagcgctct ggtgtgacca tcacgatgga 3540
gccggcggtc actatgaatc taacagcctt tgttaccggg gagtttcaat tatttcatca 3600
aataagaact caggcacaaa gctgtctttc aactgtcacg tcctgaaaac aaatggcagg 3660
tgacattttc catgccatag cagtgccact gggcattttc agggcccatg tgccaggagg 3720
gcgtgggcat cggcgagtgg aggctcctgg ccgtgtcagc tggcccaggg ggaggagggg 3780
acccagacag ccagaggtgg ggagcaggct ttccccctgt gacgctgcag acccaccgca 3840
ctgccctggg aggaagggga gggaactggg ccaaggggga agggcaggtg tgctggaggc 3900
caaggcagac ctgcacacca ccctggagag caggggttga ccccgtcccg gccccacagt 3960
caggaccccg gaggtggacg acgaggccct ggagaaattc gacaaagccc tcaaggccct 4020
gcccatgcac atccggctgt ccttcaaccc aacccagctg gagggtgagc acccaggccc 4080
caccctgctc ctggggcagg aagccacccg gcccaggacc acctcctccc atggtgaccc 4140
ccagctcccc aggcctcccg ggaggatgga gacggggtgc agggccccga ggtggccccc 4200
tccccacccc ctccccagct ccctctgtcc tggggtgtcc agtcccatcc tgacgctccc 4260
ccgccacggc tctccctccc ccacagagca gtgccacatc taggtgagcc cctgccggcg 4320
cctctggggt aagctgcctg ccctgcccca cgtcctgggc acacacatgg ggtaggggtt 4380
cttggttggg cccgggagcc cccattaggc cctggggtcc ccccgtagga atggctggaa 4440
gctggggtcc ttcctggaga ctacagagcc ggctggccac atgctcgctc ttgtggggtg 4500
acctgtgtcc tggcctcact cacacgctga tctcctccac ctccttcctg gcagacctaa 4560
gggccaaggt ggaggctcag gaagtggaca cctaaggggg aggctagggg ggtccttctc 4620
ccaaggaggg gccgtcctga atccccagcc acggacaggc tggcaagggt ctggcaggta 4680
ccccaggaat cacaggggag ccccatgtcc atttcagagc ccgggagcct tggcccctct 4740
ggggacagac gatgtcatcc ccgcctgccc catcagggga ccaggaggaa ccgggaccac 4800
attcacccct cctgggaccc aggcccctcc aggcccctcc tggggcctcc tgcttggggc 4860
cgctcctcct tcagcaataa aggcataaac ctgt 4894
<210>2
<211>132
<212>DNA
<213> Artificial Sequence (Artificial Sequence)
<400>2
actccctgca gagctcagaa gcgtgacccc agctgcagcc atgaagtgcc tcctgcttgc 60
cctggccctc acttgtggcg cccaggccct cattgtcacc cagaccatga agggcctgga 120
tatccagaag gt 132
<210>3
<211>120
<212>DNA
<213> Artificial Sequence (Artificial Sequence)
<400>3
ttaatacgac tcactatagg cacccagacc atgaagggcc gttttagagc tagaaatagc 60
aagttaaaat aaggctagtc cgttatcaac ttgaaaaagt ggcaccgagt cggtgctttt 120
<210>4
<211>102
<212>RNA
<213> Artificial Sequence (Artificial Sequence)
<400>4
gcacccagac caugaagggc cguuuuagag cuagaaauag caaguuuaaa auaaggcuag 60
uccguuauca acuugaaaaa guggcaccga gucggugcuu uu 102

Claims (8)

1. The preparation method of the knockout cattle comprises the following steps (1) and (2):
(1) preparing a gene knockout cell: introducing a third generation single base editor BE3 and gRNA for identifying BLG gene into bovine fibroblasts, and preparing gene knockout cells through gene editing; all 277 bases of the BLG genes on two homologous chromosomes of the gene knockout cell from the 5' end are mutated from C to T;
the BLG gene is a DNA molecule shown as SEQ ID No. 1;
the target sequence of the gRNA is shown as 273 to 295 th positions from the 5' end of SEQ ID No. 1;
the gRNA is shown in SEQ ID No. 4;
(2) and (2) cloning the single cell of the gene knockout cell obtained in the step (1) by a somatic cell nuclear transfer technology to obtain the gene knockout cattle.
2. The method of claim 1, wherein: the third generation single base editor BE3 is mRNA obtained by in vitro transcription of a vector for expressing the third generation single base editor BE 3.
3. A method for knocking out a BLG gene in a cell, comprising the steps of: introducing a third generation single base editor BE3 and gRNA for identifying BLG gene into a target cell, and preparing a gene knockout cell through gene editing; the 277 th base of the BLG gene on two homologous chromosomes of the gene knockout cell from the 5' end is mutated from C to T; the target sequence of the gRNA is shown as the 273 to 295 th positions from the 5' end of SEQ ID No. 1; the BLG gene is a DNA molecule shown as SEQ ID No. 1; the gRNA is shown in SEQ ID No. 4.
4. Use of the method according to any one of claims 1 to 3 for breeding cows producing hypoallergenic milk.
5. The method for breeding the dairy cattle producing hypoallergenic milk comprises the following steps: the method of any one of claims 1 to 3, wherein the knockout dairy cow is a dairy cow producing hypoallergenic milk.
6. A kit for preparing a knockout cow comprises gRNAs shown as SEQ ID No.4 or an expression cassette containing gRNAs shown as SEQ ID No. 4.
7. The kit of claim 6, wherein: the kit also comprises a vector for expressing the third generation single base editor BE3 or mRNA obtained by in vitro transcription of the vector.
8. Use of the kit according to claim 6 or 7 for breeding cows producing hypoallergenic milk.
CN202010426973.7A 2020-05-20 2020-05-20 Method for preparing accurate BLG gene knockout cattle by using third generation base editor Active CN111334529B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010426973.7A CN111334529B (en) 2020-05-20 2020-05-20 Method for preparing accurate BLG gene knockout cattle by using third generation base editor

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010426973.7A CN111334529B (en) 2020-05-20 2020-05-20 Method for preparing accurate BLG gene knockout cattle by using third generation base editor

Publications (2)

Publication Number Publication Date
CN111334529A CN111334529A (en) 2020-06-26
CN111334529B true CN111334529B (en) 2020-09-29

Family

ID=71181103

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010426973.7A Active CN111334529B (en) 2020-05-20 2020-05-20 Method for preparing accurate BLG gene knockout cattle by using third generation base editor

Country Status (1)

Country Link
CN (1) CN111334529B (en)

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BR122023023211A2 (en) * 2014-04-28 2024-01-23 Recombinetics, Inc. METHOD OF MAKING MULTIPLEX GENE EDITS IN A VERTEBRATE OR PRIMARY NON-HUMAN EMBRYO CELL
CN109321600A (en) * 2018-10-19 2019-02-12 中国农业大学 A kind of method and its application of ox that cultivating production low irritability milk

Also Published As

Publication number Publication date
CN111334529A (en) 2020-06-26

Similar Documents

Publication Publication Date Title
JP6878482B2 (en) Targeted genome editing in large livestock conjugations
WO2014041327A1 (en) Genetically edited animal
Zhu et al. CRISPR/Cas9‐mediated MSTN disruption accelerates the growth of Chinese Bama pigs
CN111808887B (en) Method for preparing double-muscle gluteal beef cattle similar to natural mutation Belgian blue cattle
Tanihara et al. Current status of the application of gene editing in pigs
US11535850B2 (en) Methods for improving the health of porcine species by targeted inactivation of CD163
Hirata et al. One-step genome editing of porcine zygotes through the electroporation of a CRISPR/Cas9 system with two guide RNAs
Whitworth et al. Improvements in pig agriculture through gene editing
Kang et al. Apancreatic pigs cloned using Pdx1-disrupted fibroblasts created via TALEN-mediated mutagenesis
CN112094868B (en) Method for preparing CD163 gene edited pig by using single base editor SpRY-BE4
WO2021057806A1 (en) Method for establishing diabetes disease model dog
CN110938629B (en) Complete sgRNA for specifically recognizing pig Wip1 gene and application and product thereof
US20030115618A1 (en) Method of generating a transgenic livestock animal
CN111334529B (en) Method for preparing accurate BLG gene knockout cattle by using third generation base editor
WO2012135981A1 (en) Method for knocking out bovine beta-lactoglobulin gene by using zinc finger nucleases
CN110283847A (en) A kind of while site-directed integration FAD3 and FABP4 gene carrier and recombinant cell
Pan et al. Cloned pigs derived from somatic cell nuclear transfer embryos cultured in vitro at low oxygen tension
WO2022012663A1 (en) Virus insusceptible animal and construction method therefor
CN109321600A (en) A kind of method and its application of ox that cultivating production low irritability milk
CN111849977B (en) Method for preparing transgenic animals by sperm vector, sgRNA for preparing short and small transgenic chickens and preparation method
CN117143916A (en) Method for establishing WNT10A gene function-deficiency mutation disease model dog
WO2019184937A1 (en) Method for producing pig having improved properties
CN116904513A (en) Method for establishing SCN5A gene edited arrhythmia model dog
CN115820733A (en) Method for establishing CADASIL disease model dog
CN115786398A (en) Method for establishing DRD2 gene editing model dog

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant