CN105518139B - Method for specifically knocking out pig FGL2 gene by CRISPR-Cas9 and sgRNA for specifically targeting FGL2 gene - Google Patents

Method for specifically knocking out pig FGL2 gene by CRISPR-Cas9 and sgRNA for specifically targeting FGL2 gene Download PDF

Info

Publication number
CN105518139B
CN105518139B CN201580000471.5A CN201580000471A CN105518139B CN 105518139 B CN105518139 B CN 105518139B CN 201580000471 A CN201580000471 A CN 201580000471A CN 105518139 B CN105518139 B CN 105518139B
Authority
CN
China
Prior art keywords
fgl2
gene
sequence
sgrna
target sequence
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
CN201580000471.5A
Other languages
Chinese (zh)
Other versions
CN105518139A (en
Inventor
蔡志明
牟丽莎
高汉超
谢崇伟
陆赢
刘璐
陈鹏飞
张军方
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.)
Shenzhen Second Peoples Hospital
Original Assignee
Shenzhen Second Peoples Hospital
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 Shenzhen Second Peoples Hospital filed Critical Shenzhen Second Peoples Hospital
Publication of CN105518139A publication Critical patent/CN105518139A/en
Application granted granted Critical
Publication of CN105518139B publication Critical patent/CN105518139B/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
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • 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
    • C12N15/86Viral vectors

Abstract

The invention discloses a method for specifically knocking out pig FGL2 gene by using CRISPR-Cas9 and sgRNA for specifically targeting FGL2 gene. The target sequence of sgRNA of the invention specifically targeting FGL2 gene on FGL2 gene conforms to the sequence arrangement rule of 5 '-N (20) NGG-3', wherein N (20) represents 20 consecutive bases, wherein each N represents a or T or C or G; the target sequence on the FGL2 gene is located in the exon coding region of the FGL2 gene; the target sequence on the FGL2 gene is unique. The sgRNA disclosed by the invention is used in a method for specifically knocking out the pig FGL2 gene by CRISPR-Cas9, can quickly, accurately, efficiently and specifically knock out the pig FGL2 gene, and effectively solves the problems of long period and high cost in constructing FGL2 gene knock-out pigs.

Description

Method for specifically knocking out pig FGL2 gene by CRISPR-Cas9 and sgRNA for specifically targeting FGL2 gene
Technical Field
The invention relates to the technical field of genetic engineering, in particular to the technical field of gene knockout, and specifically relates to a method for specifically knocking out a porcine FGL2 gene by using CRISPR-Cas9 and sgRNA for specifically targeting FGL2 gene.
Background
Organ transplantation is the most effective treatment for organ failure. To date, nearly a million patients worldwide have continued their lives through organ transplantation. With the aging population and the advancement of medical technology, more and more patients need to undergo organ transplantation, but the shortage of donor organs severely restricts the development of organ transplantation. Taking kidney transplantation as an example, as many as 30 million patients need kidney transplantation every year in China, but no more than 1 million donor kidneys are available for transplantation, and most patients die of renal failure. The need for organ transplantation has not been met by means of post-mortem organ donation. The genetic engineering of other species to provide organs suitable for human transplantation has become a major approach to the problem of organ shortage in human donors.
At present, pigs become the most ideal source of xenogeneic organs according to the evaluation in many aspects such as biosafety, physiological function indexes, economy, rare species protection and the like. However, there is a great difference between pigs and humans, and a strong immune rejection reaction is generated by directly transplanting the organs of pigs to humans. Therefore, the genetic engineering of pigs to produce organs suitable for human transplantation is the ultimate goal of xenotransplantation.
Fibrinogen-like protein 2(FGL2) has a highly conserved Fibrinogen domain that can be either expressed on the surface of the cell membrane or secreted extracellularly, FGL2 on the membrane surface is a direct prothrombinase, while FGL2 secreted extracellularly has immunomodulatory effects. FGL2 promotes the conversion of procoagulant enzymes to thrombin in the absence of the classical prothrombinase complex. Cellulose deposition is a typical feature of AVR. Deletion of FGL2 inhibits cellulose deposition using antibody-neutralized or FGL2 knockout mice. Thrombin is a potential immune activation factor that activates platelets and acts directly on vascular smooth muscle cells and vascular endothelial cells, which promote thrombus formation. Indeed, FGL 2-deficient mice as donors reduced AVR production in a mouse-to-rat xenograft model. Since FGL2 has such an important role in AVR, constructing FGL 2-deleted genetically modified pigs would likely make an important contribution to xenotransplantation.
Currently, common gene knockout technologies include Homologous Recombination (HR) technology, Transcription activation Effector-Like Nuclease (TALEN) technology, Zinc Finger Nuclease (ZFN) technology, and recently developed Regularly Clustered Short interspersed Palindromic Repeat (CRISPR) technology. HR technology is inefficient due to recombination (efficiency is only about 10)-6) The screening work for mutants is very time consuming and inefficient and has gradually been replaced. The cutting efficiency of the TALEN technology and the ZFN technology can generally reach 20%, but protein modules capable of identifying specific sequences need to be constructed, and the earlier work is complicated and time-consuming. The module design of the ZFN technology is complex and has high miss rate, and its application is limited.
CRISPR is an acquired immune system derived from prokaryotes, the complex that performs the interference function consisting of the protein Cas and CRISPR-rna (crrna). Three types of the system have been found, wherein the Cas9 system in the second type has simple composition and is actively applied to the field of genetic engineering. Cas9 targeted cleavage of DNA is achieved by the principle of complementary recognition of two small RNAs, crRNA (crispr RNA) and tracrRNA (trans-activating crRNA), to the target sequence. Two small RNAs have now been fused into one RNA strand, sgrna (single guide RNA) for short, capable of recognizing a specific gene sequence and guiding Cas9 protein cleavage. In eukaryotes, DNA is cleaved and end-linked by non-homologous recombination, resulting in frame shift mutations that ultimately result in functional gene knock-outs.
Compared with the 3 technologies, the CRISPR technology is simple to operate and high in screening efficiency, and can realize accurate targeted cutting. Therefore, the FGL2 gene knockout by the CRISPR technology can greatly improve the screening efficiency of FGL2 deleted cells and genetically engineered pigs. However, the key technical problem of the path is to design and prepare the sgRNA with accurate targeting, because the targeting accuracy of the gene is highly dependent on the sgRNA target sequence, and whether the sgRNA with accurate targeting can be successfully designed becomes the key technical problem of target gene knockout, and the invention aims to solve the technical problem so as to provide a solid foundation for knockout of the FGL2 gene.
Disclosure of Invention
The invention aims to provide a method for specifically knocking out pig FGL2 gene by CRISPR-Cas9 and sgRNA for specifically targeting FGL2 gene.
According to a first aspect of the invention, the invention provides a sgRNA for specifically targeting FGL2 gene in CRISPR-Cas9 specific knockout pig FGL2 gene, the sgRNA having the following features:
(1) the target sequence of the sgRNA on the FGL2 gene conforms to the sequence arrangement rule of 5 '-N (20) NGG-3', wherein N (20) represents 20 continuous bases, wherein each N represents A or T or C or G, and the target sequence conforming to the rule can be positioned on a sense strand or an antisense strand;
(2) the target sequence of the sgRNA on the FGL2 gene is positioned in the exon coding region of the FGL2 gene;
(3) the sgRNA is unique in the target sequence on FGL2 gene.
As a preferred scheme of the invention, the target sequence is SEQ ID NO: 1 to 78 in sequence list.
As a preferred scheme of the invention, the target sequence is SEQ ID NO: 3 or 25.
According to a second aspect of the invention, the invention provides a method for CRISPR-Cas9 specific knock-out of porcine FGL2 gene, comprising the steps of:
(1) adding a sequence for forming a sticky end to the 5' -end of the target sequence of the sgRNA in the first aspect, and synthesizing to obtain a forward oligonucleotide sequence; adding appropriate sequences for forming cohesive ends to both ends of a complementary sequence corresponding to the target sequence of the sgRNA described in the first aspect, and synthesizing to obtain an inverted oligonucleotide sequence; annealing and renaturing the synthesized forward oligonucleotide sequence and the reverse oligonucleotide sequence to form double-stranded oligonucleotide with a sticky end;
(2) connecting the double-stranded oligonucleotide into a linearized expression vector carrying a Cas9 gene to obtain an expression vector carrying sgRNA oligonucleotide containing a corresponding target sequence and a Cas9 gene, transforming competent bacteria, screening and identifying correct positive clone, shaking the positive clone, and extracting a plasmid;
(3) the expression vector, the packaging plasmid and the packaging cell line which carry the sgRNA oligonucleotide and the Cas9 gene are used for packaging the pseudotype lentivirus which simultaneously carries the sgRNA of the targeted FGL2 gene and the Cas 9;
(4) infecting a target cell with the pseudotyped lentivirus and further culturing; then collecting the infected target cell, amplifying a gene segment containing the target sequence by using the genome DNA of the infected target cell as a template, and determining the knockout condition of the FGL2 gene through denaturation, renaturation and enzyme digestion.
As a preferred scheme of the invention, the expression vector is SEQ ID NO: 79, or a vector comprising the sequence shown in seq id no.
As a preferred embodiment of the present invention, the above method comprises the steps of:
(1) adding a CACCG sequence to the 5' -end of the target sequence of the sgRNA in the first aspect, and synthesizing to obtain a forward oligonucleotide sequence; adding an AAAC sequence to the 5 '-end and adding a C to the 3' -end of a complementary sequence corresponding to the target sequence of the sgRNA in the first aspect, and synthesizing to obtain a reverse oligonucleotide sequence; annealing and renaturing the synthesized forward oligonucleotide sequence and the reverse oligonucleotide sequence to form double-stranded oligonucleotide with a sticky end;
(2) the double-stranded oligonucleotide is connected into a nucleotide sequence shown as SEQ ID NO: 79, obtaining a recombinant expression vector lentiCRISPR v2-FGL2 carrying sgRNA oligonucleotide by a linearized vector obtained by carrying out restriction enzyme digestion on an expression vector lentiCRISPR v2 with a sequence shown by 79 by BsmB I restriction enzyme, transforming competent bacteria, screening and identifying correct positive clone, shaking the positive clone, and extracting plasmid;
(3) the expression vector lentiCRISPR v2-FGL2, a packaging plasmid and a packaging cell line are used for packaging a pseudotype lentivirus which simultaneously carries sgRNA of a targeted FGL2 gene and Cas 9;
(4) infecting a target cell by using the CRISPR pseudotyped slow virus and further culturing; then collecting the infected target cell, amplifying a gene segment containing the target sequence by using the genome DNA of the infected target cell as a template, and determining the knockout condition of the FGL2 gene through denaturation, renaturation and enzyme digestion.
As a preferred embodiment of the present invention, the above-mentioned packaging plasmids are plasmid pLP1, plasmid pLP2 and plasmid pLP/VSVG; the packaging cell line is HEK293T cell.
In a preferred embodiment of the present invention, the target cell is a porcine PIEC cell.
In a preferred embodiment of the present invention, the gene fragment including the target sequence is amplified using the genomic DNA as a template, and the FGL2 gene knockout is determined by denaturation, renaturation and enzyme digestion, specifically:
(a) using the genomic DNA of the target cells infected with the virus as a template, amplifying an FGL2 gene fragment containing the target sequence of the sgRNA by using upstream and downstream primers of an FGL2 gene, and simultaneously amplifying the genomic DNA of wild-type cells not infected with the virus by using the same primer;
(b) purifying the amplified FGL2 gene fragment, mixing the FGL2 gene fragment from the target cell infected with virus and the FGL2 gene fragment from the wild cell in equal amount, heating for denaturation and renaturation to form hybrid DNA molecule;
(c) cutting the renatured hybrid DNA molecules by using Cruiser enzyme;
(d) detecting the enzyme digestion product by electrophoresis, and detecting the FGL2 gene knockout effect mediated by the target sequence.
According to the third aspect of the invention, the invention provides a recombinant expression vector lentiCRISPR v2-FGL2 used in a method for specifically knocking out porcine FGL2 gene by CRISPR-Cas9, wherein the sequence of the skeleton vector of the recombinant expression vector is shown as SEQ ID NO: 79; the carried target sequence is the target sequence of the sgRNA of the first aspect, preferably the sequence table SEQ ID NO: 3 or 25.
According to a fourth aspect of the invention, the invention provides the use of the sgRNA of the first aspect or the recombinant expression vector lentiCRISPR v2-FGL2 of the third aspect in a method of CRISPR-Cas9 specific knockout of the porcine FGL2 gene.
The sgRNA of the specific targeted FGL2 gene is successfully found by aiming at CRISPR-Cas9 specific knockout of the pig FGL2 gene, and the sgRNA is used in the method for CRISPR-Cas9 specific knockout of the pig FGL2 gene, so that the pig FGL2 gene can be quickly, accurately, efficiently and specifically knocked out, and the technical problems of long period and high cost in constructing the FGL2 gene knockout pig are effectively solved.
Drawings
Fig. 1 is a plasmid map of the vector plasmid lentiCRISPR v2 used in the examples of the present invention;
FIG. 2 is a plasmid map of the packaging plasmid pLP1 used in the examples of the present invention;
FIG. 3 is a plasmid map of the packaging plasmid pLP2 used in the examples of the present invention;
FIG. 4 is a plasmid map of the packaging plasmid pLP/VSVG used in the examples of the present invention;
FIG. 5 is a diagram showing the results of electrophoresis detection of the gene knockout effect of the enzyme digestion verification target sequence in the embodiment of the present invention, in which M represents DNA Marker, 1 and 2 respectively represent the targeted cleavage effect of the No. 3 and No. 25 target sequences in Table 1 on FGL2 gene, WT represents the detection result of the enzyme digestion of Cruiser, which is the PCR product of wild-type cells that have not undergone viral infection and Cas9 cleavage, and the arrow indicates small fragments obtained by the cleavage of Cruiser enzyme.
Detailed Description
The technical solution of the present invention is further explained with reference to the accompanying drawings and specific embodiments. The drawings and the detailed description are not intended to limit the scope of the invention. Unless otherwise specified, the technical means used in the examples are conventional means well known to those skilled in the art, and the raw materials used are commercially available products.
Test materials and reagents referred to in the following examples: the lentiCRISPR v2 plasmids were purchased from addge corporation, the packaging plasmids pLP1, pLP2 and pLP/VSVG were purchased from Invitrogen corporation, the packaging cell line HEK293T cells were purchased from american type culture collection bank (ATCC), the PIEC cells were purchased from chinese academy of sciences cell bank, the DMEM medium, Opti-MEM medium and fetal bovine serum FBS were purchased from Gibco corporation, and the Lipofectamine2000 was purchased from Invitrogen corporation.
The molecular biological experiments, which are not specifically described in the following examples, were performed by referring to the specific methods described in molecular cloning, a laboratory manual (third edition) j. sambrook, or according to the kit and product instructions.
The general technical scheme of the invention comprises the following five parts:
selection and design of sgRNA target sequence of Sus scrofa (pig) FGL2 gene
sgRNA target sequence selection of FGL2 gene:
an appropriate 20bp oligonucleotide sequence was found in the exon region of FGL2 gene as a target sequence.
sgRNA target sequence design of FGL2 gene:
and (3) adding linkers to the target sequence and the complementary sequence to form a forward oligonucleotide sequence and a reverse oligonucleotide sequence.
Second, construct CRISPR vector of FGL2 gene
1. The forward oligonucleotide sequence and the reverse oligonucleotide sequence are synthesized and renatured to form double-stranded DNA fragments (i.e., double-stranded target sequence oligonucleotides, which may also be referred to as double-stranded oligonucleotides) with sticky ends.
2. Constructing a CRISPR-sgRNA expression vector:
the double-stranded DNA fragment is constructed into a target vector (such as lentiCRISPR v2, and the plasmid map of the double-stranded DNA fragment is shown in figure 1) to form a lentivirus CRISPR vector such as lentiCRISPR v2-FGL 2.
Thirdly, obtaining the pseudotyped slow virus expressing FGL2sgRNA
And (3) producing the CRISPR pseudotyped lentivirus expressing FGL2sgRNA by using the packaging plasmid, the packaging cell line and the lentivirus CRISPR vector.
Fourthly, infecting target cells and detecting the knockout effect of FGL2 gene
1. Lentivirus infection of cells of interest:
a pseudotyped lentivirus such as lentiCRISPR v2-FGL2 is added to a cell culture medium of interest for infection and further culture.
2. Detecting the knockout effect of FGL2 gene:
collecting target cells, amplifying a gene segment containing a target sequence by using the genomic DNA as a template, and determining the knockout condition of the FGL2 gene through denaturation, renaturation and enzyme digestion.
Fifthly, selecting and identifying FGL2 gene knockout monoclone
1. For a target cell group with a definite knockout effect, a plurality of cell strains with single cell sources are separated through dilution and monoclonal culture.
2. A monoclonal FGL2 knockout was identified.
The technical solution and the advantageous effects thereof of the present invention will be described in detail by examples below.
Example one selection and design of sgRNA target sequence of Sus scrofa (pig) FGL2 Gene
The target sequence determines the targeting specificity of the sgRNA and the efficiency of inducing Cas9 to cleave the gene of interest. Therefore, efficient and specific target sequence selection and design are a prerequisite for constructing sgRNA expression vectors.
sgRNA target sequence selection of FGL2 Gene
For the FGL2 gene, the following principles should be followed in the selection of target sequences:
(1) searching for a target sequence conforming to the 5 '-N (20) NGG-3' rule in the exon coding region of the FGL2 gene, wherein N (20) represents 20 consecutive bases, wherein each N represents A or T or C or G, and the target sequence conforming to the rule can be positioned in a sense strand or an antisense strand;
(2) selecting exon coding region sequences, particularly selecting exon coding region sequences near the N end, wherein the functional knockout of the FGL2 gene can be caused by the cutting of the coding region sequences, and functional proteins cannot be formed by residual truncated sequences;
(3) if multiple spliceosomes are present, selection is made in the consensus exon coding region, and this condition can be met by selecting the exon coding region sequence near the N-terminus for the FGL2 gene;
(4) the homology of the target sequence in the pig genome is analyzed by using an online sequence analysis tool (http:// crispr. mit. edu /), the target sequence with significant homologous sequence is discarded, and the selected target sequence is further selected according to the score, and is unique on the FGL2 gene.
Based on the above principle, the target sequence set shown in Table 1 was selected.
TABLE 1 set of target sequences
Numbering Sequence of
1 TCCGCTGTCCTCGCGGCTTA
2 CCAAAAAGCCATAAGCCGCG
3 GCTTTCTAGTCTCACCGGGC
4 CCTCGCGGCTTATGGCTTTT
5 TGAAGCTGAAGCTGTCGAAC
6 CGTGGCCAACAATGAGACGG
7 TGAAGCTGTCGAACTGGTGC
8 GGCTTATGGCTTTTTGGTCG
9 GGTAGGCGGGTGTCCCTACC
10 TCTCACCGGGCAGGCGTCCT
11 GGTCGTGGCCAACAATGAGA
12 GCCATAAGCCGCGAGGACAG
13 AATTTCCTCCGTCTCATTGT
14 TCAAGGGGGGCAGGTTCACC
15 AGCTCCCCAAGCAGTTTGGC
16 TCAATCCTGCCAAACTGCTT
17 CAATCCTGCCAAACTGCTTG
18 GCTGAGCTCCGCTGTCCTCG
19 CTCAATCCTGCCAAACTGCT
20 CAAGCAGTTTGGCAGGATTG
21 CTCTGCTTTCTAGTCTCACC
22 GGAAATGCGAAGAGGTAGGC
23 AGCAGCCAAGGACGCCTGCC
24 CTTCAGCTCCCCAAGCAGTT
25 GCAGTTTGGCAGGATTGAGG
26 CCTCTGCTTTCTAGTCTCAC
27 GGGGGGCAGGTTCACCTGGT
28 GCTGAAGAGTCAAGGGGGGC
29 AAGCAGAGGGAAATGCGAAG
30 CCGGTGAGACTAGAAAGCAG
31 GATTGAGGAGGTGTTCAAAG
32 CTTGGGGAGCTGAAGAGTCA
33 GGGAAATGCGAAGAGGTAGG
34 CGGTGAGACTAGAAAGCAGA
35 GGGGGCAGGTTCACCTGGTA
36 AGAGGGAAATGCGAAGAGGT
37 TTGGGGAGCTGAAGAGTCAA
38 TGGGGAGCTGAAGAGTCAAG
39 GGGGAGCTGAAGAGTCAAGG
40 GAAGAGATCGACGGGCTTCA
41 GCTGACGACAACCGAGACCC
42 ATGCCAAGGAAGAGATCGAC
43 GGAAGAGATCGACGGGCTTC
44 AAGCCCGTCGATCTCTTCCT
45 CAGGAATGGACTGCTGTCAC
46 AATGCCAAGGAAGAGATCGA
47 TTGCCAAGACTGCAGACTGC
48 CTCTGACCTGAAGAATGCCA
49 GGACTGCTGTCACCGGGCAC
50 ACGACAACCGAGACCCAGGC
51 AGGAATGGACTGCTGTCACC
52 AGTTAGAGAATTGGAGAACG
53 AGTCTTGGCAAGTCTTCTTC
54 GACAGCAGTCCATTCCTGCC
55 GGCTGTTCACAATTTCCTTG
56 CTTGGCATTCTTCAGGTCAG
57 TGGATGGCAAATGTTCATCG
58 TGAGATCTACAGAGTTACAC
59 AACGTTTGCTGTCAATAGTT
60 AGAACAAATACAGTCACGTC
61 AACTATTGACAGCAAACGTT
62 TGCTCTGAATACTACACAAT
63 AAATTACGTTGATAACAAGG
64 TTTGCTGTCAATAGTTTGGA
65 GACTGTATTTGTTCTTGACT
66 TTATATATAAGATGTTGAAC
67 TGACTGTATTTGTTCTTGAC
68 GTGCTGCAGGCACGTCTTGA
69 TTTGTAGTCTCGCCATGTTC
70 CGAGACTACAAAGTTGGCTT
71 ACATGGCGAGACTACAAAGT
72 TCGCCATGTTCTGGTGAAGT
73 AAGCTACTGTTTTTGGGATC
74 TCATCTTCTGACTAAGAGTA
75 GCACCAACTTCACCAGAACA
76 TACTGTGACATGGAGACCAT
77 CCTCAGAAGAGAATTTTGGT
78 GATTCTAAGAATAGATCTTG
sgRNA target sequence design of FGL2 gene:
(1) a lentiCRISPR v2 plasmid is used as an expression vector, and a CACCG sequence is added to the 5' -end of the N (20) target sequence according to the characteristics of the lentiCRISPR v2 plasmid to form a forward oligonucleotide sequence:
5’-CACCGNNNNNNNNNNNNNNNNNNNN-3’;
(2) adding sequences to both ends of the reverse complement of the N (20) target sequence to form a reverse oligonucleotide sequence:
5’-AAACNNNNNNNNNNNNNNNNNNNNC-3’;
the forward oligonucleotide sequence and the reverse oligonucleotide sequence may be complementary to form a double-stranded DNA fragment with sticky ends:
5’-CACCGNNNNNNNNNNNNNNNNNNNN-3’
3’-CNNNNNNNNNNNNNNNNNNNNCAAA-5’。
example two construction of sgRNA expression vector for FGL2 Gene
1. Synthesis of DNA insert
(1) Synthesis of the designed Forward and reverse oligonucleotide sequences
Oligonucleotide sequences can be specifically synthesized by commercial companies (e.g., Invitrogen corporation) based on the sequences provided. In this example and the following examples, the effect of knocking out the FGL2 gene by the target sequences shown in SEQ ID Nos. 3 and 25 listed in Table 1 was examined.
The forward and reverse oligonucleotide sequences corresponding to target sequence No. 3 are as follows:
GACCGGCTTTCTAGTCTCACCGGGC(SEQ ID NO:80);
AAACGCCCGGTGAGACTAGAAAGCC(SEQ ID NO:81)。
the forward and reverse oligonucleotide sequences corresponding to target sequence No. 25 are as follows:
CACCGGCAGTTTGGCAGGATTGAGG(SEQ ID NO:82);
AAACCCTCAATCCTGCCAAACTGCC(SEQ ID NO:83)。
the corresponding forward and reverse oligonucleotide sequences are annealed and renatured to form double-stranded DNA fragments having sticky ends.
The reaction system (20. mu.L) is shown below:
forward oligonucleotide (10. mu.M): 1 μ L
Reverse oligonucleotide (10 μ M): 1 μ L
10×PCR buffer:2μL
ddH2O:16μL
The reaction system was placed in a PCR apparatus and the reaction was carried out according to the following procedure.
Reaction procedure:
95℃,5min;
80℃,5min;
70℃,5min;
60℃,5min;
50℃,5min;
naturally cooling to room temperature.
2. Construction of sgRNA expression vector
(1) The BsmB I restriction enzyme is used for cutting the objective vector lentiCRISPR v2 plasmid (the sequence of which is shown as SEQ ID NO: 79 in the sequence table).
The preparation method comprises the following steps:
the lentiscrispr v2 plasmid: 1 μ g
10 Xenzyme digestion buffer: 2 μ L
BsmB I restriction enzyme: 2 μ L
Supplemental ddH2O to a total volume of 20. mu.L
The enzyme digestion reaction system is placed at 37 ℃ for reaction for 4 h.
(2) Electrophoretic separation and purification of vector fragments
After completion of the digestion, the digestion mixture was separated by agarose gel electrophoresis, and the vector fragment (about 12kb) was selected for cleavage and recovered by a DNA gel recovery column.
(3) Connecting the synthesized double-stranded DNA fragment with the vector main fragment and transforming the double-stranded DNA fragment into escherichia coli
Performing ligation reaction on the double-stranded DNA fragment obtained by renaturation and the recovered vector fragment, and preparing according to the following reaction system:
the LentiCRISPR v2 vector fragment: 100ng
Double-stranded DNA fragment: 200ng
T4 ligase: 1 μ L
T4 ligation reaction buffer: 1 μ L
Supplemental ddH2O to a total volume of 10. mu.L
The ligation mixture was left to react for 2h at 25 ℃.
After the reaction was complete, the ligation mixture was transformed into E.coli strain DH5 α: add 100. mu.L E.coli DH 5. alpha. competent cells to the ligation mixture and incubate for 30min on ice; putting the mixture into a water bath at 42 ℃, performing heat shock for 90s, and then putting the mixture on ice for cooling; adding 100 μ L LB medium into the mixture, and shake culturing at 37 deg.C for 20 min; the mixture was spread on Amp LB plates and incubated at 37 ℃ for 14 h.
(4) Identification of the correct transformed clones
And selecting a plurality of colonies from the Amp LB plate for amplification culture, and extracting plasmids for enzyme digestion identification. Clones that are likely to be correct are selected for sequencing, and the correct insertion sequence is verified. The correct lentiCRISPR v2-FGL2 vector clone was used for seed preservation.
Example three obtaining a pseudotyped Lentivirus expressing FGL2sgRNA
1. Material preparation
The packaging plasmids pLP1, pLP2, and pLP/VSVG (purchased from Invitrogen, maps shown in FIG. 2, FIG. 3, and FIG. 4, respectively) were amplified and extracted; amplifying and extracting a vector plasmid lentiCRISPR v2-FGL 2; culturing packaging cell line HEK293T cells (purchased from ATCC); DMEM medium, Opti-MEM medium and fetal bovine serum FBS (purchased from Gibco); lipofectamine2000 (ex Invitrogen); HEK293T cells cultured in 5% CO2The culture environment of (1) is 37 ℃, and the culture medium is a DMEM medium containing 10% FBS.
2. Transfection and viral packaging
The first day: the packaging cell line HEK293T was passaged to 10cm dish, approximately 30% confluence;
the next day: transfection was performed at 80% confluence of HEK293T according to the following recipe:
formulation mixture 1, comprising:
lentiCRISPR v2-FGL2:6μg
pLP1:6μg
pLP2:6μg
pLP/VSVG:3μg
Opti-MEM:500μL。
formulation mixture 2, comprising:
Lipofectamine 2000:30μL
Opti-MEM:500μL。
after standing for 5min, mix 1 and mix 2 were mixed well to form a transfection mixture, and left to stand for 20 min.
The HEK293T medium was changed to serum-free DMEM medium, the transfection mixture was added, and the medium was changed to 20% FBS DMEM medium after 8 hours at 37 ℃ to continue the culture.
3. Virus collection and preservation
And on the third day: after transfection for 48h, HEK293T medium supernatant containing virus was collected, filtered through 0.45 μm filter tip, split charged, and stored at-80 ℃.
Example four infection of cells of interest and detection of the knockout Effect of the target sequence
1. Material preparation
Culturing porcine hip arterial endothelial cells PIEC (purchased from cell bank of Chinese academy of sciences) of a target cell line; DMEM medium and fetal bovine serum FBS (purchased from Gibco); lentiCRISPR v2-FGL2 pseudotyped lentiviruses of different target sequences (seq id No. 3 and seq id No. 25); PIEC cells cultured in 5% CO2The culture environment of (1) is 37 ℃, and the culture medium is a DMEM medium containing 10% FBS.
2. Lentiviral infection of target cells
The first day: cells of interest were passaged to 6-well plates at approximately 20% confluency density. One 6 well per virus was required, while one 6 well efficiency control was required.
The next day: when the fusion density of the target cells is about 40%, 1mL lentiCRISPR v2-FGL2 pseudotype lentivirus supernatant and 1mL DMEM medium are added. The efficiency control did not require addition of lentivirus.
And on the third day: after 24h of infection, the virus-containing medium was removed, replaced with normal medium, puromycin was added to a final concentration of 2. mu.g/mL, and puromycin was added as a control to the efficiency control sample without virus infection for 48 h.
3. Cell infection efficiency detection and culture
The fifth day: uninfected efficiency control cells should all apoptosis (> 95%) under the action of puromycin. The infection efficiency of the cells is judged according to the apoptosis of infected lentivirus cells, and can generally reach more than 90 percent (the apoptosis rate is less than 10 percent). If necessary, the virus supernatant may be concentrated or diluted in a gradient and then infected to achieve a suitable infection efficiency.
After puromycin screening, uninfected cells were apoptotic. The cells of interest were re-passaged and replaced with normal medium for 48 h.
4. Detection of FGL2 gene knockout effect
(1) Designing an upstream primer and a downstream primer to amplify the FGL2 gene fragment, wherein the sequences of the upstream primer and the downstream primer are shown as follows:
GCTGGGGTGAGCGCCACCGC(SEQ ID NO:84);
CAGGCGACCCTGAAGCCCGT(SEQ ID NO:85)。
CCAACAATGAGACGGAGGAA(SEQ ID NO:86);
GCGATGAACATTTGCCATCC(SEQ ID NO:87)。
primers SEQ ID NO: 84-85 for detecting sequence No. 3, primer SEQ ID NO: 86-87 were used to detect sequence number 25.
(2) A part of the cells of interest was collected, and genomic DNA was extracted using a promega genomic DNA kit. Meanwhile, the genome DNA of the wild type target cell is extracted.
(3) The FGL2 gene fragment containing the target sequence (including the infected mutant sample and the wild-type sample) was amplified using the genomic DNA as a template.
The amplification reaction (20. mu.L) was as follows:
upstream primer (10 μ M): 1 μ L
Downstream primer (10 μ M): 1 μ L
2×PCR Mix:10μL
Genomic DNA: 100ng
The above reaction system was prepared, placed in a PCR apparatus, and reacted according to the following procedure.
Reaction procedure:
95℃,3min
95℃,30s
58℃,20s
72℃,20s
72℃,3min;
wherein the second through fourth steps are repeated for 35 cycles.
(4) Electrophoresis detection of PCR product and recovery and purification
(5) And (3) respectively heating and denaturing the purified DNA fragments to form hybrid DNA molecules (including mutant samples and wild samples).
The reaction system is as follows:
genomic PCR fragment: 200ng
5 × reaction buffer: 2 μ L
Reaction system totally 9. mu.L
The above reaction system was prepared, placed in a PCR apparatus, and reacted according to the following procedure.
Reaction procedure:
95℃,5min;
80℃,5min;
70℃,5min;
60℃,5min;
50℃,5min;
naturally cooling to room temperature.
(6) Cleavage of renatured hybrid DNA (including mutant and wild type samples) with Cruiser enzyme
mu.L of Cruiser enzyme was added to the denatured, renatured reaction mixture and incubated at 45 ℃ for 20 min.
(7) Detecting the enzyme digestion product by electrophoresis, and detecting the FGL2 gene knockout effect mediated by the target sequence.
The digested DNA fragment was analyzed by electrophoresis on a 2% agarose gel at 100V for 25 min. Determining the cutting condition of the target segment and judging the gene knockout effect of the target sequence.
The recognition of the cleavage of the mutated DNA is based on the following principle: infected cells express sgRNA and Cas 9. If targeted cleavage of genomic DNA by sgRNA mediated Cas9 protein, a mutation (wild type to mutant) is introduced near the cleavage site after repair. Because the wild type and the mutant type sequences are not matched at the position, a hybrid molecule formed by the wild type DNA and the mutant type DNA amplified by taking the wild type and the mutant type sequences as templates through renaturation can generate a local loop structure. The latter can be recognized and cleaved by the Cruiser enzyme, resulting in the cleavage of the hybrid DNA molecule into small fragments.
As a result, as shown in FIG. 5, no small fragments were detected from the PCR product of the virus-uninfected wild-type cells; and the sequence 3 and the sequence 25 can effectively target FGL2 gene to generate cleavage, so that the existence of a small fragment is detected, and the sequence 3 and the sequence 25 can be used as target sequences for CRISPR-Cas9 specific knockout of pig FGL2 gene.
EXAMPLE V selection and characterization of FGL2 Gene knockout monoclonals
1. Selection of a monoclonal (target sequence based on SEQ ID No. 3 and SEQ ID No. 25)
(1) The partially infected target cell population was passaged, and 100 single cells were transferred to 10cm dish culture.
(2) After about 10 days of culture, a significant number of the single clones grew to macroscopic levels.
(3) Individual clones were scraped with a pipette tip and cells were transferred to 24-well plates for culture, one clone per well.
(4) After about one week of culture, some clones grew to a sufficient number and were ready for further characterization.
2. Identification of a monoclonal FGL2 knockout
(1) And collecting the monoclonal and wild cells to be detected, and respectively extracting the genomic DNA.
(2) FGL2 gene fragments of monoclonal and wild type cells were amplified separately as described above, the amplified gene fragments containing the sgRNA target sequence.
(3) Mixing the same amount of monoclonal PCR fragment with wild PCR fragment, heating to denature and renature to form hybrid DNA molecule.
(4) The annealed hybrid DNA was cleaved with Cruiser enzyme and incubated at 45 ℃ for 20 min.
(5) Detecting the enzyme digestion product by electrophoresis, and determining whether effective mutation occurs in the monoclonal according to whether the cutting fragment exists.
The result shows that 20 monoclonals randomly selected from 100 single cells infect target cells based on the lentiCRISPR v2-FGL2 pseudotype lentivirus of the target sequence shown in the sequence 3 and are detected by the enzyme digestion electrophoresis of the Cruiser enzyme, wherein 19 monoclonals can detect the small cut fragment, which indicates that gene knockout occurs, and the gene knockout efficiency can reach more than 95%, thus indicating that the target sequence shown in the sequence 3 has a very high effect of targeted knockout of the FGL2 gene. 20 monoclones randomly selected from 100 single cells infect a target cell based on the lentiCRISPR v2-FGL2 pseudotype lentivirus of a target sequence shown in a sequence 25, and are detected by Cruiser enzyme digestion electrophoresis, wherein 18 monoclones can detect a small cut fragment, which indicates that gene knockout occurs, and the gene knockout efficiency can reach more than 90 percent, thus indicating that the target sequence shown in the sequence 25 has a very high effect of targeted knockout of an FGL2 gene.
The foregoing is a more detailed description of the present invention that is presented in conjunction with specific embodiments, and the practice of the invention is not to be considered limited to those descriptions. It will be apparent to those skilled in the art that a number of simple derivations or substitutions can be made without departing from the inventive concept.
Figure IDA0000812020540000011
Figure IDA0000812020540000021
Figure IDA0000812020540000031
Figure IDA0000812020540000041
Figure IDA0000812020540000051
Figure IDA0000812020540000061
Figure IDA0000812020540000071
Figure IDA0000812020540000081
Figure IDA0000812020540000091
Figure IDA0000812020540000101
Figure IDA0000812020540000111
Figure IDA0000812020540000121
Figure IDA0000812020540000131
Figure IDA0000812020540000141
Figure IDA0000812020540000151
Figure IDA0000812020540000161
Figure IDA0000812020540000171
Figure IDA0000812020540000181
Figure IDA0000812020540000191
Figure IDA0000812020540000201

Claims (8)

1. sgRNA for specifically targeting FGL2 gene in CRISPR-Cas9 specific knockout pig FGL2 gene, characterized by:
(1) the target sequence of the sgRNA on the FGL2 gene conforms to the sequence arrangement rule of 5 '-N (20) NGG-3', wherein N (20) represents 20 continuous bases, wherein each N represents A or T or C or G, and the target sequence conforming to the rule can be positioned on a sense strand or an antisense strand;
(2) the target sequence of the sgRNA on the FGL2 gene is positioned in an exon coding region of the FGL2 gene;
(3) the sgRNA is unique for its target sequence on FGL2 gene;
the target sequence is SEQ ID NO: 3 or 25.
Use of sgRNA or its target sequence on FGL2 gene for the preparation of a kit for specific knockout of porcine FGL2 gene using CRISPR-Cas9, characterized in that the use comprises the steps of:
(1) synthesizing a forward oligonucleotide sequence by adding a sequence for forming a sticky end to the 5' -end of the target sequence of the sgRNA of claim 1; adding appropriate sequences for forming cohesive ends to both ends of a complementary sequence corresponding to a target sequence of the sgRNA according to claim 1, and synthesizing to obtain an inverted oligonucleotide sequence; annealing and renaturing the synthesized forward oligonucleotide sequence and the synthesized reverse oligonucleotide sequence to form double-stranded oligonucleotide with a sticky end;
(2) connecting the double-stranded oligonucleotide into a linearized expression vector carrying a Cas9 gene to obtain an expression vector carrying sgRNA oligonucleotide containing a corresponding target sequence and a Cas9 gene, converting competent bacteria, screening and identifying correct positive clone, shaking the positive clone, and extracting a plasmid;
(3) packaging a pseudolentivirus simultaneously carrying sgRNA of a targeted FGL2 gene and Cas9 by using the expression vector, the packaging plasmid and the packaging cell line which carry the sgRNA oligonucleotide and the Cas9 gene;
(4) infecting a target cell with the pseudotyped lentivirus and further culturing; and then collecting infected target cells, amplifying a gene segment containing the target sequence by using the genome DNA of the infected target cells as a template, and determining the knockout condition of the FGL2 gene through denaturation, renaturation and enzyme digestion.
3. The use of claim 2, wherein the expression vector is SEQ ID NO: 79, or a vector comprising the sequence shown in seq id no.
4. Use according to claim 2 or 3, characterized in that it comprises the following steps:
(1) adding a CACCG sequence to the 5' -end of the target sequence of the sgRNA of claim 1, and synthesizing to obtain a forward oligonucleotide sequence; adding an AAAC sequence to the 5 '-end and C to the 3' -end of a complementary sequence corresponding to the target sequence of the sgRNA of claim 1, and synthesizing to obtain an inverse oligonucleotide sequence; annealing and renaturing the synthesized forward oligonucleotide sequence and the synthesized reverse oligonucleotide sequence to form double-stranded oligonucleotide with a sticky end;
(2) and (2) connecting the double-stranded oligonucleotide into a nucleotide sequence shown as SEQ ID NO: 79, obtaining a recombinant expression vector lentiCRISPR v2-FGL2 carrying sgRNA oligonucleotide by a linearized vector obtained by digesting an expression vector lentiCRISPR v2 with BsmB I restriction enzyme, transforming competent bacteria, screening and identifying correct positive clone, shaking the positive clone, and extracting plasmid;
(3) the expression vector lentiCRISPR v2-FGL2, a packaging plasmid and a packaging cell line are used for packaging a pseudotype lentivirus which simultaneously carries sgRNA of a targeted FGL2 gene and Cas 9;
(4) infecting a target cell with the pseudotyped lentivirus and further culturing; and then collecting infected target cells, amplifying a gene segment containing the target sequence by using the genome DNA of the infected target cells as a template, and determining the knockout condition of the FGL2 gene through denaturation, renaturation and enzyme digestion.
5. Use according to claim 4, characterized in that the packaging plasmids are plasmid pLP1, plasmid pLP2 and plasmid pLP/VSVG; the packaging cell line is HEK293T cell.
6. The use according to claim 4, wherein the cell of interest is a porcine PIEC cell;
amplifying a gene segment containing the target sequence by taking the genome DNA as a template, and determining the knockout condition of the FGL2 gene through denaturation, renaturation and enzyme digestion, wherein the method specifically comprises the following steps:
(a) using the genomic DNA of target cells infected with the virus as a template, amplifying an FGL2 gene fragment containing a target sequence of the sgRNA by using upstream and downstream primers of an FGL2 gene, and simultaneously amplifying the genomic DNA of wild-type cells not infected with the virus by using the same primer;
(b) purifying the amplified FGL2 gene fragment, mixing the FGL2 gene fragment from the target cell infected with virus and the FGL2 gene fragment from the wild cell in equal amount, heating for denaturation, and renaturing to form a hybrid DNA molecule;
(c) cutting the renatured hybrid DNA molecules by using Cruiser enzyme;
(d) detecting the enzyme digestion product by electrophoresis, and detecting the FGL2 gene knockout effect mediated by the target sequence.
7. The recombinant expression vector lentiCRISPR v2-FGL2 used in the method for specifically knocking out pig FGL2 gene by CRISPR-Cas9 is characterized in that the sequence of the skeleton vector of the recombinant expression vector is shown as SEQ ID NO: 79; the carried target sequence is selected from SEQ ID NO: 3 or 25.
8. Use of the recombinant expression vector lentiCRISPR v2-FGL2 of claim 7 in the preparation of a kit for specific knockout of porcine FGL2 gene using CRISPR-Cas 9.
CN201580000471.5A 2015-06-11 2015-06-11 Method for specifically knocking out pig FGL2 gene by CRISPR-Cas9 and sgRNA for specifically targeting FGL2 gene Active CN105518139B (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2015/081230 WO2016197358A1 (en) 2015-06-11 2015-06-11 Method for specific knockout of swine fgl-2 gene using crispr-cas9 specificity, and sgrna used for specifically targeting fgl-2 gene

Publications (2)

Publication Number Publication Date
CN105518139A CN105518139A (en) 2016-04-20
CN105518139B true CN105518139B (en) 2021-02-02

Family

ID=55724976

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201580000471.5A Active CN105518139B (en) 2015-06-11 2015-06-11 Method for specifically knocking out pig FGL2 gene by CRISPR-Cas9 and sgRNA for specifically targeting FGL2 gene

Country Status (2)

Country Link
CN (1) CN105518139B (en)
WO (1) WO2016197358A1 (en)

Families Citing this family (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10323236B2 (en) 2011-07-22 2019-06-18 President And Fellows Of Harvard College Evaluation and improvement of nuclease cleavage specificity
US9163284B2 (en) 2013-08-09 2015-10-20 President And Fellows Of Harvard College Methods for identifying a target site of a Cas9 nuclease
US9359599B2 (en) 2013-08-22 2016-06-07 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
US9340800B2 (en) 2013-09-06 2016-05-17 President And Fellows Of Harvard College Extended DNA-sensing GRNAS
US9322037B2 (en) 2013-09-06 2016-04-26 President And Fellows Of Harvard College Cas9-FokI fusion proteins and uses thereof
US9526784B2 (en) 2013-09-06 2016-12-27 President And Fellows Of Harvard College Delivery system for functional nucleases
US9068179B1 (en) 2013-12-12 2015-06-30 President And Fellows Of Harvard College Methods for correcting presenilin point mutations
AU2015298571B2 (en) 2014-07-30 2020-09-03 President And Fellows Of Harvard College Cas9 proteins including ligand-dependent inteins
IL258821B (en) 2015-10-23 2022-07-01 Harvard College Nucleobase editors and uses thereof
KR102547316B1 (en) 2016-08-03 2023-06-23 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 Adenosine nucleobase editing agents and uses thereof
CN109804066A (en) 2016-08-09 2019-05-24 哈佛大学的校长及成员们 Programmable CAS9- recombination enzyme fusion proteins and application thereof
WO2018039438A1 (en) 2016-08-24 2018-03-01 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
KR102622411B1 (en) 2016-10-14 2024-01-10 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 AAV delivery of nucleobase editor
WO2018119359A1 (en) 2016-12-23 2018-06-28 President And Fellows Of Harvard College Editing of ccr5 receptor gene to protect against hiv infection
WO2018165504A1 (en) 2017-03-09 2018-09-13 President And Fellows Of Harvard College Suppression of pain by gene editing
US11542496B2 (en) 2017-03-10 2023-01-03 President And Fellows Of Harvard College Cytosine to guanine base editor
GB2575930A (en) 2017-03-23 2020-01-29 Harvard College Nucleobase editors comprising nucleic acid programmable DNA binding proteins
US11560566B2 (en) 2017-05-12 2023-01-24 President And Fellows Of Harvard College Aptazyme-embedded guide RNAs for use with CRISPR-Cas9 in genome editing and transcriptional activation
AU2018288048A1 (en) * 2017-06-20 2019-12-19 Jiangsu Hengrui Medicine Co., Ltd. Method for knocking out target gene in T cell in vitro and crRNA used in the method
CN109207515A (en) * 2017-07-03 2019-01-15 华中农业大学 A method of design and building pig full-length genome CRISPR/Cas9 knock out library
JP2020534795A (en) 2017-07-28 2020-12-03 プレジデント アンド フェローズ オブ ハーバード カレッジ Methods and Compositions for Evolving Base Editing Factors Using Phage-Supported Continuous Evolution (PACE)
EP3676376A2 (en) 2017-08-30 2020-07-08 President and Fellows of Harvard College High efficiency base editors comprising gam
WO2019079347A1 (en) 2017-10-16 2019-04-25 The Broad Institute, Inc. Uses of adenosine base editors
CN111100876A (en) * 2018-10-25 2020-05-05 立沃生物科技(深圳)有限公司 Method for specifically knocking out FAH gene by CRISPR-Cas9 and specific sgRNA
WO2020191249A1 (en) 2019-03-19 2020-09-24 The Broad Institute, Inc. Methods and compositions for editing nucleotide sequences
CN110438128B (en) * 2019-08-20 2023-04-25 山西农业大学 Method for knocking out pig CCAR1 gene by using CRISPR/Cas9 system
MX2022014008A (en) 2020-05-08 2023-02-09 Broad Inst Inc Methods and compositions for simultaneous editing of both strands of a target double-stranded nucleotide sequence.
CN113151274B (en) * 2021-04-15 2024-01-19 贵州省畜牧兽医研究所 New LncRNA application, excavation and identification method for regulating and controlling fat deposition performance of pigs
CN113564165B (en) * 2021-07-19 2023-06-27 武汉市工程科学技术研究院 Cell strain for intracellular editing of pseudorabies virus key genes, construction method and application thereof
CN114350615B (en) * 2021-12-20 2024-04-16 北京镁伽科技有限公司 STAT2 gene deletion cell strain and preparation method and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998051335A1 (en) * 1997-05-15 1998-11-19 Transplantation Technologies Inc. Methods of modulating immune coagulation
CN102085378A (en) * 2010-12-29 2011-06-08 华中科技大学同济医学院附属同济医院 Application of hfgl2 (Human Fibrinogen-like protein 2) inhibitor in preparation of medicaments for treating liver cancer
CN104480144A (en) * 2014-12-12 2015-04-01 武汉大学 CRISPR/Cas9 recombinant lentiviral vector for human immunodeficiency virus gene therapy and lentivirus of CRISPR/Cas9 recombinant lentiviral vector

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2932421A1 (en) * 2012-12-12 2015-10-21 The Broad Institute, Inc. Methods, systems, and apparatus for identifying target sequences for cas enzymes or crispr-cas systems for target sequences and conveying results thereof
US9783616B2 (en) * 2013-04-02 2017-10-10 University Of Connecticut Regulating transplant rejection of donor and embryonic stem cell-derived tissues and organs

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998051335A1 (en) * 1997-05-15 1998-11-19 Transplantation Technologies Inc. Methods of modulating immune coagulation
CN102085378A (en) * 2010-12-29 2011-06-08 华中科技大学同济医学院附属同济医院 Application of hfgl2 (Human Fibrinogen-like protein 2) inhibitor in preparation of medicaments for treating liver cancer
CN104480144A (en) * 2014-12-12 2015-04-01 武汉大学 CRISPR/Cas9 recombinant lentiviral vector for human immunodeficiency virus gene therapy and lentivirus of CRISPR/Cas9 recombinant lentiviral vector

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
Characterization of fibrinogen-like protein 2 (FGL2): Monomeric FGL2 has enhanced immunosuppressive activity in comparison to oligomeric FGL2;Hao Liu;《The International Journal of Biochemistry》;20121102;第408-418页 *
Efficient generation of targeted and controlled mutational events in porcine cells using nuclease-directed homologous recombination;James R. Butler;《journal of surgical research》;20170129;第239-246页 *
猪FGL2基因cDNA末端序列检测及结构分析;刘浩;《遗传》;20031231;第25卷(第1期);第17-21页 *
登录号:AY112657;佚名;《GenBank》;20040422;参见序列部分 *

Also Published As

Publication number Publication date
CN105518139A (en) 2016-04-20
WO2016197358A1 (en) 2016-12-15

Similar Documents

Publication Publication Date Title
CN105518139B (en) Method for specifically knocking out pig FGL2 gene by CRISPR-Cas9 and sgRNA for specifically targeting FGL2 gene
CN105492608B (en) Method for specifically knocking out pig PDX1 gene by CRISPR-Cas9 and sgRNA for specifically targeting PDX1 gene
CN105518137B (en) Method for specifically knocking out pig SALL1 gene by CRISPR-Cas9 and sgRNA for specifically targeting SALL1 gene
CN105518138B (en) Method for specifically knocking out pig GFRA1 gene by CRISPR-Cas9 and sgRNA for specifically targeting GFRA1 gene
CN105518135B (en) Method for specifically knocking out pig CMAH gene by CRISPR-Cas9 and sgRNA for specifically targeting CMAH gene
WO2016197357A1 (en) Method for specific knockout of swine sla-3 gene using crispr-cas9 specificity, and sgrna used for specifically targeting sla-3 gene
WO2016197356A1 (en) Method for knockout of swine sla-2 gene using crispr-cas9 specificity, and sgrna used for specifically targeting sla-2 gene
WO2016187904A1 (en) Method for pig cmah gene specific knockout by means of crispr-cas9 and sgrna for specially targeting cmah gene
WO2016197361A1 (en) Method for specific knockout of swine ggta1 gene using crispr-cas9 specificity, and sgrna used for specifically targeting ggta1 gene
CN107502608B (en) Construction method and application of sgRNA and ALDH2 gene-deleted cell strain for knocking out human ALDH2 gene
WO2016197359A1 (en) Method for specific knockout of swine sla-1 gene using crispr-cas9 specificity, and sgrna used for specifically targeting sla-1 gene
WO2016197362A1 (en) Method for specific knockout of swine vwf gene using crispr-cas9 specificity, and sgrna used for specifically targeting vwf gene
US20160362667A1 (en) CRISPR-Cas Compositions and Methods
WO2017190664A1 (en) Use of chemosynthetic crrna and modified crrna in crispr/cpf1 gene editing systems
CN105886498A (en) Method for specifically knocking out human PCSK9 gene by virtue of CRISPR-Cas9 and sgRNA for specifically targeting PCSK9 gene
US20210198699A1 (en) Kit for reparing fbn1t7498c mutation, combination for making and repairing mutation, and method of repairing thereof
CN106987560B (en) Construction method of RK-13 cell HBB gene knockout stable strain
JP2019514379A (en) Methods for in vivo high-throughput evaluation of RNA-inducible nuclease activity
CN111254164A (en) Method for rapidly establishing CRISPR gene editing liver cancer cell strain and cell strain
CN111484994B (en) Method for specifically knocking out pig Fah and Rag2 double genes by CRISPR-Cas9
CN110551762B (en) CRISPR/ShaCas9 gene editing system and application thereof
CN114990093B (en) Protein sequence MINI RFX-CAS13D with small amino acid sequence
CN112979823B (en) Product and fusion protein for treating and/or preventing beta-hemoglobinopathy
CN110551763B (en) CRISPR/SlutCas9 gene editing system and application thereof
US20240066116A1 (en) Fully synthetic, long-chain nucleic acid for vaccine production to protect against coronaviruses

Legal Events

Date Code Title Description
C06 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