CN105492608B - Method for specifically knocking out pig PDX1 gene by CRISPR-Cas9 and sgRNA for specifically targeting PDX1 gene - Google Patents

Method for specifically knocking out pig PDX1 gene by CRISPR-Cas9 and sgRNA for specifically targeting PDX1 gene Download PDF

Info

Publication number
CN105492608B
CN105492608B CN201580000468.3A CN201580000468A CN105492608B CN 105492608 B CN105492608 B CN 105492608B CN 201580000468 A CN201580000468 A CN 201580000468A CN 105492608 B CN105492608 B CN 105492608B
Authority
CN
China
Prior art keywords
sequence
pdx1 gene
pdx1
gene
sgrna
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
CN201580000468.3A
Other languages
Chinese (zh)
Other versions
CN105492608A (en
Inventor
蔡志明
牟丽莎
谢崇伟
陈鹏飞
张军方
陆赢
高汉超
刘璐
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 CN105492608A publication Critical patent/CN105492608A/en
Application granted granted Critical
Publication of CN105492608B publication Critical patent/CN105492608B/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
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • 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
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15041Use of virus, viral particle or viral elements as a vector
    • C12N2740/15045Special targeting system for viral vectors
    • 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
    • C12N2810/00Vectors comprising a targeting moiety
    • C12N2810/10Vectors comprising a non-peptidic targeting moiety

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Wood Science & Technology (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Virology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

The invention discloses a method for specifically knocking out pig PDX1 gene by using CRISPR-Cas9 and sgRNA for specifically targeting PDX1 gene. The sgRNA of the present invention specifically targeting PDX1 gene targets the PDX1 gene with a 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 PDX1 gene is located at the 1 st exon coding region at the N-terminus of the PDX1 gene or at the junction with the adjacent intron; the target sequence on the PDX1 gene is unique. The sgRNA disclosed by the invention is used in a method for specifically knocking out the pig PDX1 gene by CRISPR-Cas9, can quickly, accurately, efficiently and specifically knock out the pig PDX1 gene, and effectively solves the problems of long period and high cost in constructing PDX1 gene knock-out pigs.

Description

Method for specifically knocking out pig PDX1 gene by CRISPR-Cas9 and sgRNA for specifically targeting PDX1 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 PDX1 gene by using CRISPR-Cas9 and sgRNA for specifically targeting a PDX1 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.
The traditional technical route is to obtain a strain of swine that can be used for transplantation by reducing the differences in immunity between swine and humans. In recent years, the production of organs composed of human cells using organ development-deficient pigs as a culture environment has become a new idea. The gene for controlling the development of the pig organ is effectively interfered by genetic engineering, so that a certain organ of the pig is deleted in the development process, and a key culture environment is provided for the development of human cell organs.
The PDX1 gene is known to be an essential gene in pancreatic development and islet beta cell maturation. The pancreas requires the interaction of the anterior pancreatic epithelium with mesenchymal tissue during development. PDX1 is essential in this process as a transcription factor of the Hox type. Mice homozygous for the deletion of PDX1 develop a loss of pancreatic development and eventually die. Through artificially knocking out the PDX1 gene of the pig, the pig with pancreas development deficiency can be obtained and used for generating human cell-derived pancreatic islets. The accurate and efficient knockout of the PDX1 gene of the pig is the first step in the production of artificial pancreatic islets.
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 screening efficiency of PDX1 deletion cells and pancreas development deletion gene engineering pigs can be greatly improved by knocking out the PDX1 gene through the CRISPR technology. 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 PDX1 gene.
Disclosure of Invention
The invention aims to provide a method for specifically knocking out pig PDX1 gene by CRISPR-Cas9 and sgRNA for specifically targeting PDX1 gene.
According to a first aspect of the invention, the invention provides sgrnas in CRISPR-Cas 9-specific knockout of porcine PDX1 gene for specific targeting of PDX1 gene, the sgrnas having the following characteristics:
(1) the target sequence of the sgRNA on the PDX1 gene conforms to the sequence arrangement rule of 5 '-N (20) NGG-3', wherein N (20) represents 20 continuous bases, each N represents A or T or C or G, and the target sequence conforming to the rule is positioned on a sense strand or an antisense strand;
(2) the sgRNA target sequence on the PDX1 gene is located in the coding region of the 1 st exon at the N end of the PDX1 gene, or a part of the target sequence is located in the 1 st exon at the N end of the PDX1 gene, and the rest of the target sequence spans the boundary with the adjacent intron and is located in the adjacent intron;
(3) the sgRNA is unique in the target sequence on the PDX1 gene.
As a preferred scheme of the invention, the target sequence is SEQ ID NO: 1 to 20 in sequence list.
As a preferred scheme of the invention, the target sequence is SEQ ID NO: 3 or 10.
According to a second aspect of the invention, the invention provides a method for specifically knocking out a porcine PDX1 gene using CRISPR-Cas9, the method 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) packaging a pseudolentivirus simultaneously carrying sgRNA of a targeted PDX1 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; 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 PDX1 gene through denaturation, renaturation and enzyme digestion.
As a preferred scheme of the invention, the expression vector is SEQ ID NO: 21, or a vector having the sequence shown in figure 21.
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: 21, obtaining a linear vector by carrying out restriction enzyme digestion on an expression vector lentiCRISPR v2 with a sequence shown in 21 by BsmB I restriction enzyme to obtain a recombinant expression vector lentiCRISPR v2-PDX1 carrying sgRNA oligonucleotide, transforming competent bacteria, screening and identifying correct positive clone, shaking the positive clone, and extracting plasmid;
(3) the expression vector lentiCRISPR v2-PDX1, a packaging plasmid and a packaging cell line are used for packaging a pseudolentivirus which simultaneously carries sgRNA of a target PDX1 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 PDX1 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 PDX1 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 a PDX1 gene fragment containing the target sequence of the sgRNA by using the upstream and downstream primers of the PDX1 gene, and simultaneously amplifying the genomic DNA of wild-type cells not infected with the virus by using the same primers;
(b) purifying the PDX1 gene fragment amplified, and then heating and denaturing a PDX1 gene fragment from a target cell infected with a virus and a PDX1 gene fragment from a wild-type cell respectively to form hybrid DNA molecules;
(c) cutting the renatured hybrid DNA molecules by using Cruiser enzyme;
(d) detecting the enzyme digestion product by electrophoresis, and detecting the target sequence mediated PDX1 gene knockout effect.
According to a third aspect of the invention, the invention provides a recombinant expression vector lentiCRISPR v2-PDX1 used in a method for specifically knocking out porcine PDX1 gene by CRISPR-Cas9, wherein the sequence of a skeleton vector of the recombinant expression vector is shown as SEQ ID NO: 21 is shown in the figure; the carried target sequence is the target sequence of the sgRNA of the first aspect, preferably the sequence table SEQ ID NO: 3 or 10.
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-PDX1 of the third aspect in a method of CRISPR-Cas9 specific knockout of the porcine PDX1 gene.
The sgRNA of the pig PDX1 gene is successfully found by aiming at CRISPR-Cas9 specific knockout, and the sgRNA of the pig PDX1 gene is used in the method for CRISPR-Cas9 specific knockout of the pig PDX1 gene, so that the pig PDX1 gene can be quickly, accurately, efficiently and specifically knocked out, and the technical problems of long period and high cost in constructing the PDX1 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 gene knockout effect of enzyme digestion verification target sequence in the example of the present invention, where M represents DNA Marker, WT represents the results of Cruiser enzyme digestion detection of the PCR product of wild-type cells that have not undergone viral infection and Cas9 cleavage, 3 and 10 represent the targeted cleavage effect of target sequences No. 3 and No. 10 in Table 1 on PDX1 gene, and the arrow indicates small fragments obtained by cleavage with 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) PDX1 gene
sgRNA target sequence selection of PDX1 gene:
a suitable 20bp oligonucleotide sequence was found in the exon region of the PDX1 gene as a target sequence.
sgRNA target sequence design of PDX1 gene:
and (3) adding linkers to the target sequence and the complementary sequence to form a forward oligonucleotide sequence and a reverse oligonucleotide sequence.
Secondly, constructing CRISPR vector of PDX1 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-PDX 1.
Thirdly, obtaining the pseudotyped slow virus expressing PDX1sgRNA
A CRISPR pseudolentivirus expressing PDX1sgRNA is produced by using a packaging plasmid, a packaging cell line and a lentivirus CRISPR vector.
Fourthly, infecting target cells and detecting the gene knockout effect of PDX1
1. Lentivirus infection of cells of interest:
a pseudotyped lentivirus such as lentiCRISPR v2-PDX1 was added to the cell culture medium of interest for infection and further culture.
2. Detecting the effect of the PDX1 gene knockout:
collecting target cells, amplifying a gene segment containing a target sequence by using genome DNA as a template, and determining the knockout condition of the PDX1 gene through denaturation, renaturation and enzyme digestion.
Fifthly, selection and identification of PDX1 gene knockout monoclonal
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 PDX1 knock-out 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) PDX1 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 PDX1 Gene
For the PDX1 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 coding region of the exon of the PDX1 gene, wherein N (20) represents 20 continuous bases, each N represents A or T or C or G, and the target sequence conforming to the rule is positioned in a sense strand or an antisense strand;
(2) selecting a 1 st exon coding region sequence near the N-terminus, the target sequence may be located in the 1 st exon coding region at the N-terminus of the PDX1 gene, or a portion of the target sequence is located in the 1 st exon at the N-terminus of the PDX1 gene, and the remainder spans the junction with the adjacent intron and is located in the adjacent intron; such cleavage of the coding region sequence would result in a functional knock-out of the PDX1 gene, leaving the truncated sequence without the formation of a functional protein;
(3) if multiple splice bodies exist, the coding region of the common exon is selected, and the 1 st exon coding region sequence close to the N end is selected aiming at the PDX1 gene to meet the condition;
(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 PDX1 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 CGGCCCATGTACAGGCACGC
2 CCCAGGGCGCCGGCGAACGG
3 CCCCCTGCGTGCCTGTACAT
4 CCTCCGTTCGCCGGCGCCCT
5 GGGGCTGGCGGCCCATGTAC
6 CCGCCGCCGCCTCCGTTCGC
7 GGCCCATGTACAGGCACGCA
8 GCCTCCGTTCGCCGGCGCCC
9 AGGGCGCCGGCGAACGGAGG
10 GCCCATGTACAGGCACGCAG
11 CCCATGTACAGGCACGCAGG
12 GCCCTGCTCCAGCGCGCCCA
13 GCGCCCAGGGCGCCGGCGAA
14 GCGCCGGCGAACGGAGGCGG
15 CGCCCTGCTCCAGCGCGCCC
16 CGCCCTGGGCGCGCTGGAGC
17 CTCCAGCGCGCCCAGGGCGC
18 CCGGCGAACGGAGGCGGCGG
19 CGCCGGCGCCCTGGGCGCGC
20 GCCCTGGGCGCGCTGGAGCA
sgRNA target sequence design of PDX1 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 PDX1 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 PDX1 gene by the target sequences shown in SEQ ID Nos. 3 and 10 listed in Table 1 was examined.
The forward and reverse oligonucleotide sequences corresponding to target sequence No. 3 are as follows:
CACCGCCCCCTGCGTGCCTGTACAT(SEQ ID NO:22);
AAACATGTACAGGCACGCAGGGGGC(SEQ ID NO:23)。
the forward and reverse oligonucleotide sequences corresponding to target sequence No. 10 are as follows:
CACCGGCCCATGTACAGGCACGCAG(SEQ ID NO:24);
AAACCTGCGTGCCTGTACATGGGCC(SEQ ID NO:25)。
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: 21 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-PDX1 vector clone was used for seed preservation.
Example three obtaining a pseudotyped lentivirus expressing PDX1sgRNA
1. Material preparation
Amplifying and extractingExtracting packaging plasmids pLP1, pLP2, and pLP/VSVG (purchased from Invitrogen, maps of which are shown in FIG. 2, FIG. 3, and FIG. 4, respectively); amplifying and extracting a vector plasmid lentiCRISPR v2-PDX 1; 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-PDX1:6μg
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 (from Gibco)(ii) a lentiCRISPR v2-PDX1 pseudotyped lentiviruses of different target sequences (seq id No. 3 and seq id No. 10); 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: 1mL of lentiCRISPR v2-PDX1 pseudotyped lentiviral supernatant and 1mL of DMEM medium were added when the desired cells had a confluency of about 40%. 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 PDX1 Gene knockout Effect
(1) Designing an upstream primer and a downstream primer to amplify the PDX1 gene segment, wherein the sequences of the upstream primer and the downstream primer are shown as follows:
CATGAATGGCGAGGAGCAGTACTACG(SEQ ID NO:26)
GTGAGCCTTGGTAGACTTCATCCACG(SEQ ID NO:27)。
the target amplified fragment contains sgRNA target sequence and is 388bp in size. The positions of the target sequence from both ends of the fragment are not less than 100 bp.
(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 PDX1 gene fragment containing the target sequence (including the infected mutant and wild-type samples) was amplified using 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 target sequence mediated PDX1 gene knockout effect.
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. The mutant sample contains a part of wild-type DNA component, so that hybrid molecules containing local circular structures can be formed after renaturation.
As a result, as shown in FIG. 5, the wild-type cells which had not undergone viral infection did not produce cleavage, and thus small fragments were not detected; and the sequence 3 and the sequence 10 can effectively target the PDX1 gene to generate cleavage, so that the existence of a small fragment is detected, and the sequence 3 and the sequence 10 can be used as target sequences for CRISPR-Cas9 specific knockout of pig PDX1 gene.
EXAMPLE V selection and characterization of PDX1 Gene knockout monoclonals
1. Selection of a monoclonal (target sequence based on SEQ ID No. 3 and SEQ ID No. 10)
(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 PDX1 knockout
(1) And collecting the monoclonal and wild cells to be detected, and respectively extracting the genomic DNA.
(2) The PDX1 gene fragments of the monoclonal and wild type cells were amplified separately as described above, the amplified gene fragments comprising 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 8 monoclonals randomly selected from 100 single cells infect a target cell based on the lentiCRISPR v2-PDX1 pseudotype lentivirus of the target sequence shown in the sequence 3 and are detected by enzyme digestion electrophoresis of the Cruiser enzyme, wherein 7 monoclonals can detect a small cut fragment, which indicates that gene knockout occurs, and the gene knockout efficiency can reach more than 87%, thus indicating that the target sequence shown in the sequence 3 has a very high effect of targeted knockout of the PDX1 gene. The lentiCRISPR v2-PDX1 pseudotype lentivirus based on the target sequence shown in the sequence 10 infects a target cell, 6 monoclonals randomly selected from 100 single cells are detected by Cruiser enzyme digestion electrophoresis, wherein 6 monoclonals can detect a small cut fragment, which indicates that gene knockout occurs, the gene knockout efficiency can reach 100%, and the target sequence shown in the sequence 10 has a very high effect of targeted knockout of a PDX1 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 IDA0000811990500000011
Figure IDA0000811990500000021
Figure IDA0000811990500000031
Figure IDA0000811990500000041
Figure IDA0000811990500000051
Figure IDA0000811990500000061
Figure IDA0000811990500000071
Figure IDA0000811990500000081
Figure IDA0000811990500000091
Figure IDA0000811990500000101
Figure IDA0000811990500000111

Claims (8)

1. sgRNA for specifically targeting PDX1 gene in specific knockout of porcine PDX1 gene using CRISPR-Cas9, characterized in that:
(1) the sgRNA on the PDX1 gene has a target sequence conforming 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 is positioned on a sense strand or an antisense strand;
(2) the target sequence of the sgRNA on the PDX1 gene is located in the coding region of the 1 st exon at the N end of the PDX1 gene, or a part of the target sequence is located in the 1 st exon at the N end of the PDX1 gene, and the rest part of the target sequence spans the boundary with the adjacent intron and is located in the adjacent intron;
(3) the sgRNA is unique in the target sequence on the PDX1 gene;
the target sequence is SEQ ID NO: 3 or 10.
2. A method for specifically knocking out pig PDX1 gene by using CRISPR-Cas9, which is characterized by comprising the following steps:
(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 PDX1 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; 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 PDX1 gene through denaturation, renaturation and enzyme digestion.
3. The method for the CRISPR-Cas9 specific knockout of porcine PDX1 gene according to claim 2, wherein the expression vector is SEQ ID NO: 21, or a vector having the sequence shown in figure 21.
4. The method for CRISPR-Cas9 specific knockout of porcine PDX1 gene according to claim 2 or 3, characterized in that the method 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: 21, obtaining a linear vector by carrying out restriction enzyme digestion on an expression vector lentiCRISPR v2 with a sequence shown in 21 by BsmB I restriction enzyme to obtain a recombinant expression vector lentiCRISPR v2-PDX1 carrying sgRNA oligonucleotide, transforming competent bacteria, screening and identifying correct positive clone, shaking the positive clone, and extracting plasmid;
(3) the expression vector lentiCRISPR v2-PDX1, a packaging plasmid and a packaging cell line are used for packaging a pseudolentivirus which simultaneously carries sgRNA of a target PDX1 gene and 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 PDX1 gene through denaturation, renaturation and enzyme digestion.
5. The method for CRISPR-Cas9 specific knockout of porcine PDX1 gene 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 method for CRISPR-Cas9 specific knockout of porcine PDX1 gene 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 PDX1 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 a PDX1 gene fragment containing the target sequence of the sgRNA by using the upstream and downstream primers of the PDX1 gene, and simultaneously amplifying the genomic DNA of wild-type cells not infected with the virus by using the same primers;
(b) purifying the PDX1 gene fragment amplified, and then heating and denaturing a PDX1 gene fragment from a target cell infected with a virus and a PDX1 gene fragment from a wild-type cell respectively to form hybrid DNA molecules;
(c) cutting the renatured hybrid DNA molecules by using Cruiser enzyme;
(d) detecting the enzyme digestion product by electrophoresis, and detecting the target sequence mediated PDX1 gene knockout effect.
7. The recombinant expression vector lentiCRISPR v2-PDX1 used in the method for specifically knocking out pig PDX1 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: 21 is shown in the figure; the carried target sequence is SEQ ID NO: 3 or 10.
8. Use of the sgRNA of claim 1 or the recombinant expression vector lentiCRISPR v2-PDX1 of claim 7 in a method of CRISPR-Cas9 specific knockout of the porcine PDX1 gene.
CN201580000468.3A 2015-06-11 2015-06-11 Method for specifically knocking out pig PDX1 gene by CRISPR-Cas9 and sgRNA for specifically targeting PDX1 gene Active CN105492608B (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2015/081226 WO2016197354A1 (en) 2015-06-11 2015-06-11 Crispr-cas9 method for specific knockout of swine pdx1 gene and sgrna for use in targeting specifically pdx1 gene

Publications (2)

Publication Number Publication Date
CN105492608A CN105492608A (en) 2016-04-13
CN105492608B true CN105492608B (en) 2021-07-23

Family

ID=55678476

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201580000468.3A Active CN105492608B (en) 2015-06-11 2015-06-11 Method for specifically knocking out pig PDX1 gene by CRISPR-Cas9 and sgRNA for specifically targeting PDX1 gene

Country Status (2)

Country Link
CN (1) CN105492608B (en)
WO (1) WO2016197354A1 (en)

Families Citing this family (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3613852A3 (en) 2011-07-22 2020-04-22 President and Fellows of Harvard College Evaluation and improvement of nuclease cleavage specificity
US20150044192A1 (en) 2013-08-09 2015-02-12 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
US20150166985A1 (en) 2013-12-12 2015-06-18 President And Fellows Of Harvard College Methods for correcting von willebrand factor point mutations
EP4079847A1 (en) 2014-07-30 2022-10-26 President And Fellows Of Harvard College Cas9 proteins including ligand-dependent inteins
JP7067793B2 (en) 2015-10-23 2022-05-16 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 Nucleobase editing factors and their use
CN105886534A (en) * 2016-04-29 2016-08-24 苏州溯源精微生物科技有限公司 Tumor metastasis inhibition method
KR102547316B1 (en) 2016-08-03 2023-06-23 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 Adenosine nucleobase editing agents and uses thereof
US11661590B2 (en) 2016-08-09 2023-05-30 President And Fellows Of Harvard College Programmable CAS9-recombinase fusion proteins and uses thereof
US11542509B2 (en) 2016-08-24 2023-01-03 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
US11306324B2 (en) 2016-10-14 2022-04-19 President And Fellows Of Harvard College AAV delivery of nucleobase editors
US10745677B2 (en) 2016-12-23 2020-08-18 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
EP3592777A1 (en) 2017-03-10 2020-01-15 President and Fellows of Harvard College Cytosine to guanine base editor
IL306092A (en) 2017-03-23 2023-11-01 Harvard College Nucleobase editors comprising nucleic acid programmable dna binding proteins
WO2018209320A1 (en) 2017-05-12 2018-11-15 President And Fellows Of Harvard College Aptazyme-embedded guide rnas for use with crispr-cas9 in genome editing and transcriptional activation
CN111801345A (en) 2017-07-28 2020-10-20 哈佛大学的校长及成员们 Methods and compositions using an evolved base editor for Phage Assisted Continuous Evolution (PACE)
US11319532B2 (en) 2017-08-30 2022-05-03 President And Fellows Of Harvard College High efficiency base editors comprising Gam
JP2021500036A (en) 2017-10-16 2021-01-07 ザ ブロード インスティテュート, インコーポレーテッドThe Broad Institute, Inc. Use of adenosine base editing factors
CN108588071A (en) * 2018-04-25 2018-09-28 和元生物技术(上海)股份有限公司 The sgRNA of CRISPR-Cas9 targeting knock out people colon-cancer cell CNR1 genes and its specificity
AU2020242032A1 (en) 2019-03-19 2021-10-07 Massachusetts Institute Of Technology Methods and compositions for editing nucleotide sequences
BR112022022603A2 (en) 2020-05-08 2023-01-17 Broad Inst Inc METHODS AND COMPOSITIONS FOR SIMULTANEOUS EDITING OF BOTH DUAL-STRANDED NUCLEOTIDE TARGET SEQUENCE STRAINS

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102196722A (en) * 2008-08-22 2011-09-21 国立大学法人东京大学 Organ regeneration method utilizing iPS cell and blastocyst complementation
WO2015048577A2 (en) * 2013-09-27 2015-04-02 Editas Medicine, Inc. Crispr-related methods and compositions
CN104263754B (en) * 2014-08-29 2017-03-08 中国科学院广州生物医药与健康研究院 The reconstructed eggs of albinism swine model and its construction method of construction method and swine model
CN104480144B (en) * 2014-12-12 2017-04-12 武汉大学 CRISPR/Cas9 recombinant lentiviral vector for human immunodeficiency virus gene therapy and lentivirus of CRISPR/Cas9 recombinant lentiviral vector
CN104651399B (en) * 2014-12-31 2018-11-16 广西大学 A method of gene knockout being realized in Pig embryos cell using CRISPR/Cas system

Also Published As

Publication number Publication date
WO2016197354A1 (en) 2016-12-15
CN105492608A (en) 2016-04-13

Similar Documents

Publication Publication Date Title
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
CN105518139B (en) Method for specifically knocking out pig FGL2 gene by CRISPR-Cas9 and sgRNA for specifically targeting FGL2 gene
CN105518135B (en) Method for specifically knocking out pig CMAH gene by CRISPR-Cas9 and sgRNA for specifically targeting CMAH gene
CN105518138B (en) Method for specifically knocking out pig GFRA1 gene by CRISPR-Cas9 and sgRNA for specifically targeting GFRA1 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
CN107502608B (en) Construction method and application of sgRNA and ALDH2 gene-deleted cell strain for knocking out human ALDH2 gene
WO2016197361A1 (en) Method for specific knockout of swine ggta1 gene using crispr-cas9 specificity, and sgrna used for specifically targeting ggta1 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
CN115651927B (en) Methods and compositions for editing RNA
US20160362667A1 (en) CRISPR-Cas Compositions and Methods
WO2016174056A1 (en) Compositions and methods for the treatment of nucleotide repeat expansion disorders
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
CN111254164A (en) Method for rapidly establishing CRISPR gene editing liver cancer cell strain and cell strain
WO2019173248A1 (en) Engineered nucleic acid-targeting nucleic acids
CN110551762B (en) CRISPR/ShaCas9 gene editing system and application thereof
CN110499335B (en) CRISPR/SauriCas9 gene editing system and application thereof
CN110551763B (en) CRISPR/SlutCas9 gene editing system and application thereof
Sun et al. Organoid Easytag: an efficient workflow for gene targeting in human organoids
CN114891791B (en) sgRNA of specific targeting canine Rosa26 gene and application thereof
CN117844760A (en) Cell modification method for improving virus titer, gRNA, recombinant vector and modified cell

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