WO2023165613A1 - Use of 5'→3' exonuclease in gene editing system, and gene editing system and gene editing method - Google Patents

Use of 5'→3' exonuclease in gene editing system, and gene editing system and gene editing method Download PDF

Info

Publication number
WO2023165613A1
WO2023165613A1 PCT/CN2023/079632 CN2023079632W WO2023165613A1 WO 2023165613 A1 WO2023165613 A1 WO 2023165613A1 CN 2023079632 W CN2023079632 W CN 2023079632W WO 2023165613 A1 WO2023165613 A1 WO 2023165613A1
Authority
WO
WIPO (PCT)
Prior art keywords
gene editing
exonuclease
editing system
cells
site
Prior art date
Application number
PCT/CN2023/079632
Other languages
French (fr)
Chinese (zh)
Inventor
李寅青
王沛喆
富晶晶
Original Assignee
清华大学
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 清华大学 filed Critical 清华大学
Publication of WO2023165613A1 publication Critical patent/WO2023165613A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases RNAses, DNAses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y301/00Hydrolases acting on ester bonds (3.1)
    • C12Y301/16Exonucleases active with either ribo- or deoxyribonucleic acids and producing 3'-phosphomonoesters (3.16)
    • C12Y301/16001Spleen exonuclease (3.1.16.1), i.e. 5->3 exoribonuclease
    • 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
    • C12N2310/20Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPRs]

Definitions

  • the invention relates to the field of biotechnology, in particular to a use of a 5' ⁇ 3' exonuclease in a gene editing system, a gene editing system and an editing method thereof.
  • the present invention aims to solve at least one of the technical problems existing in the prior art at least to a certain extent.
  • the present invention provides a use of a 5' ⁇ 3' exonuclease in a gene editing system, a gene editing system and an editing method thereof, by introducing the 5' ⁇ 3' exonuclease into the gene editing system , can improve homologous recombination repair (HDR) efficiency, reduce non-homologous end-joining repair (NHEJ), so as to realize precise gene editing.
  • HDR homologous recombination repair
  • NHEJ non-homologous end-joining repair
  • the inventors found through a large number of experiments that the exonucleic acid excision efficiency in the 5' ⁇ 3' direction of the nucleic acid damage site is the main factor limiting the editing efficiency in the dsDNA HDR technical route; and the non-mammalian endogenous 5' ⁇ 3 Exonucleases in the 'direction, especially those derived from the 5' ⁇ 3' direction of bacteriophage, have a wide range of highly active in vivo activities.
  • the present invention proposes the use of a 5' ⁇ 3' exonuclease in a gene editing system.
  • the 5' ⁇ 3' exonuclease is in a non-fused state with the site-specific nuclease in the gene editing system.
  • the present invention provides a gene editing system.
  • the gene editing system includes: a site-specific nuclease, a 5' ⁇ 3' exonuclease, and a donor DNA; wherein, the 5' ⁇ 3' exonuclease and the The site-specific nucleases are non-fused.
  • the present invention provides a method for gene editing of cells.
  • the method includes: introducing the above-mentioned gene editing system into cells.
  • Figure 17 is a histogram of gene editing efficiency at the DNMT1 locus of the U2OS cell line in Example 9 of the present invention.
  • Figure 18 is a histogram of gene editing efficiency at the mESC cell line EMX1 site in Example 9 of the present invention.
  • Figure 20 is a histogram of gene editing efficiency at TLR sites in Example 11 of the present invention.
  • Figure 24 is a histogram of gene editing efficiency at the VEGFA site in Example 11 of the present invention.
  • Figure 26 is a histogram of gene editing efficiency at the HEK3 locus in Example 12 of the present invention.
  • Figure 29 is a histogram of gene editing efficiency at the VEGFA site in Example 12 of the present invention.
  • Fig. 36 is a histogram of gene editing efficiency in the introduction of replacement mutations in Example 17 of the present invention.
  • Figure 38 is a diagram of the results of NPM1 double-color labeling immunofluorescence verification in Example 14 of the present invention.
  • Fig. 39 is a histogram of high-throughput screening results in Example 15 of the present invention.
  • gene encompasses not only chromosomal DNA present in the nucleus but also organelle DNA present in subcellular components of cells such as mitochondria.
  • the present invention proposes the use of a 5' ⁇ 3' exonuclease in a gene editing system.
  • the 5' ⁇ 3' exonuclease is in a non-fused state with the site-specific nuclease in the gene editing system.
  • the inventors improved the competitiveness of HDR pathway repair relative to NHEJ repair by adding 5' ⁇ 3' exonuclease to the gene editing system.
  • HDR pathway repair also occurs in other periods, thereby improving the efficiency of HDR pathway repair, while greatly inhibiting NHEJ; and, the present invention also found that through the addition of 5' ⁇ 3' exonuclease, it can be achieved in non-dividing cells HDR pathway.
  • 5' ⁇ 3' exonuclease in gene editing system is the use of 5' ⁇ 3' exonuclease in gene editing.
  • it refers to a A method for gene editing, the method comprising providing a 5' ⁇ 3' exonuclease, and using the 5' ⁇ 3' exonuclease for gene editing.
  • the 5' ⁇ 3' exonuclease is T7 exonuclease.
  • the T7 exonuclease has the amino acid sequence shown in SEQ ID NO: 1 or an amino acid sequence having at least 80% homology therewith.
  • the inventors have found through a large number of experiments that using an exonuclease homologous to T7 exonuclease can reduce the repair efficiency of the NHEJ pathway and improve the repair efficiency of the HDR pathway.
  • the added amount of the site-specific nuclease is 2-50 ng.
  • the inventor obtained the above-mentioned optimal addition amount through a large number of experiments, thus, the repair efficiency of the improved HDR channel can be further filled.
  • the site-specific nuclease is selected from endonucleases, specifically, the site-specific nuclease can be selected from clustered regularly interspaced short palindromic repeats, transcription activation-like effectors At least one of nuclease, zinc finger nuclease, homing endonuclease, and restriction endonuclease.
  • the above-mentioned site-specific nucleases can be used to produce DSBs according to requirements, and the specific types are not limited.
  • the specific nucleases for the above sites can be selected from endonucleases such as clustered regularly interspaced short palindromic repeats, transcription activator-like effector nucleases, zinc finger nucleases, and homing endonucleases. At least one of them may also be a nuclease modified on the basis of any of the above-mentioned site-specific nucleases (for example, introducing a point mutation), and the specific type is not limited.
  • the gRNA includes at least one selected from crRNA/tracrRNA, sgRNA, and pegRNA.
  • the guide RNA in the gene editing system, as long as the guide RNA is capable of forming a complex with the site-specific nuclease and can target the complex to the target due to certain complementarity with the target sequence
  • the sequence is enough, and at least one of the above-mentioned guide RNAs (the guide RNAs in the present invention include traditional guide RNAs and guide RNAs optimized and improved on traditional guide RNA sequences) can be used, wherein the specific type is not limited.
  • the inventors found through experiments that by introducing end protection means to protect the end of the donor DNA as a replication template for the HDR pathway, the donor DNA can be stably present in the cell before editing begins, further improving the efficiency of gene editing, and making the editing efficiency Significantly increased.
  • the above-mentioned donor DNA used in the present invention has higher HDR pathway repair efficiency and HDR/NHEJ ratio.
  • the technical route of the gene editing system includes homologous recombination mediated by an oligonucleotide editing template, single base editing, guided editing, and homologous recombination mediated by a double-stranded long-chain nucleic acid editing template One of.
  • the DSB is produced by the CRISPR/Cas9 system, and the exonuclease is enriched by the MS2 recruitment system, wherein the exonuclease is T7 bacteriophage exonuclease, T5 phage exonuclease (the amino acid sequence is shown in SEQ ID NO: 2 ), lambda bacteriophage exonuclease (as shown in SEQ ID NO: 3), Escherichia coli exonuclease DCLRE1B (as shown in SEQ ID NO: 4), Escherichia coli exonuclease RecE (amino acid The sequence is shown in SEQ ID NO: 5) or human exonuclease EXO1 (amino acid sequence is shown in SEQ ID NO: 6), and the amino acid sequence of T7 bacteriophage exonuclease is shown in SEQ ID NO: 1.
  • the Cas9/sgRNA expression vector and the MCP-exonuclease fusion protein expression vector can be constructed using conventional techniques in the art, for example, see “Gibson, D.G. et al. Enzymatic assembly of DNA molecules up to several hundred kilobases. Nat Methods 6,343 -345, doi:10.1038/nmeth.1318(2009)” and "Potapov, V. et al. Comprehensive Profiling of Four Base Overhang Ligation Fidelity by T4DNA Ligase and Application to DNA Assembly. ACS Synth Biol 7, 2665-2674, do i :10.1021/acssynbio.8b00333 (2016)”.
  • the repair efficiency of the HDR pathway is shown in Figure 2, where the abscissa indicates the species source of the exonuclease, and all the exonucleases form fusion proteins with MS2, from left to right: no exonuclease negative control, T7 phage Exonuclease, T5 bacteriophage exonuclease, lambda bacteriophage exonuclease, E. coli exonuclease DCLRE1B, E. coli exonuclease RecE and human exonuclease EXO1 active domain; ordinate indicates repair efficiency.
  • the exonuclease is T7 exonuclease.
  • different donor DNAs are selected for experiments, that is, unmodified linear double-stranded DNA, sulfo Phosphodiester bond (PS)-modified linear double-stranded DNA (nPS refers to the 5' end of linear double-stranded DNA modified by n consecutive phosphorothioate bonds) and circular double-stranded DNA in the form of HMEJ were used as donors.
  • PS sulfo Phosphodiester bond
  • nPS refers to the 5' end of linear double-stranded DNA modified by n consecutive phosphorothioate bonds
  • circular double-stranded DNA in the form of HMEJ were used as donors.
  • the HMEJ double-stranded DNA donor is used to introduce consecutive three-base mutations at different positions within 100 bp from the cutting site (nBP refers to the distance between the introduced mutation position and the Cas9 cutting site by n base pairs).
  • nBP refers to the distance between the introduced mutation position and the Cas9 cutting site by n base pairs.
  • the gene editing efficiency was characterized by next-generation sequencing analysis, and the rest of the steps were the same as in Example 1.
  • Donor DNA was not used in this example, only Cas9/sgRNA and T7 exonuclease were introduced as the experimental group (i.e., T7+), and samples were taken at different time points after transfection, and single samples were analyzed by enzyme digestion-fluorescent quantitative PCR method. stranded DNA is detected, and the control uses MCP protein instead of T7 exonuclease (being T7-) (in this embodiment, a 6-well cell culture plate is used, and during transfection, the contents of Cas9/sgRNA and T7 exonuclease are respectively 500ng, 1500ng, PEI dosage is 12 ⁇ L).
  • the results showed that within a period of time after transfection, the total amount of single-stranded DNA near the DSB of the sample added with T7 exonuclease was significantly higher than that of the control group.
  • HMEJ double-stranded DNA donors were used to edit different sites in the cell genome, and the gene editing efficiency was analyzed and characterized by next-generation sequencing (refer to Example 3).
  • the content of Cas9/sgRNA was 20ng, and other steps were the same as in Example 1.
  • the analysis results are shown in Figure 25-30.
  • This embodiment is an extended application of Embodiment 5.
  • the donor DNA with the split-sfGFP tag introduced at the C-terminal of the NPM1 protein that is, NPM1 WT
  • the donor DNA with the pathogenic +4 mutation and the FlAsH short peptide tag introduced at the C-terminal of the NPM1 protein that is, the NPM1 mut
  • the Cas9/sgRNA content is 50ng
  • the T7 exonuclease content is 80ng
  • the donor DNA is 100ng.
  • Other steps are the same as in Example 5.
  • Example 16 Study on the effect of polyQ structure length of ATXN2 protein on protein function by means of gene editing

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Cell Biology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Mycology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

A use of a 5'→3' exonuclease in a gene editing system, and the gene editing system and a gene editing method. The 5'→3' exonuclease and a site-specific nuclease in the gene editing system are in a non-fusion state; and the gene editing system comprises the site-specific nuclease, the 5'→3' exonuclease, and a donor DNA, wherein the 5'→3' exonuclease and the site-specific nuclease are in a non-fusion state.

Description

5’→3’核酸外切酶在基因编辑系统中的用途和基因编辑系统及其编辑方法Use of 5'→3' exonuclease in gene editing system and gene editing system and editing method thereof 技术领域technical field
本发明涉及生物技术领域,特别涉及一种5’→3’核酸外切酶在基因编辑系统中的用途和基因编辑系统及其编辑方法。The invention relates to the field of biotechnology, in particular to a use of a 5'→3' exonuclease in a gene editing system, a gene editing system and an editing method thereof.
背景技术Background technique
基因编辑技术通过修改基因序列,包括核酸碱基的插入、缺失和替换,改变蛋白质的表达和功能,从而影响生物体的各种性状。在生物技术领域,使用基因编辑技术改变特定核酸序列,优化蛋白质的已有功能或产生新的功能,以此增强生物体的性状表现,已成为生物育种等生物产业的基础。在生物医疗领域,利用基因编辑技术修复特定位点突变到正常序列,是基因疾病彻底治愈的可能唯一有效途径。因此,能够高效、准确的产生特定核酸碱基的插入、缺失和替换的基因编辑技术(精准基因编辑技术),是生物产业、生物医疗等重要领域的关键技术,其中,提高同源重组修复(HDR)效率、降低非同源末端链接修复(NHEJ)是精准基因编辑的关键技术需求。Gene editing technology changes the expression and function of proteins by modifying the gene sequence, including the insertion, deletion and substitution of nucleic acid bases, thereby affecting various traits of organisms. In the field of biotechnology, the use of gene editing technology to change specific nucleic acid sequences, optimize the existing functions of proteins or generate new functions, so as to enhance the traits of organisms, has become the basis of biological industries such as biological breeding. In the field of biomedicine, the use of gene editing technology to repair specific site mutations to normal sequences may be the only effective way to completely cure genetic diseases. Therefore, gene editing technology (precision gene editing technology), which can efficiently and accurately generate insertion, deletion and replacement of specific nucleic acid bases, is a key technology in important fields such as bioindustry and biomedicine. Among them, improving homologous recombination repair ( HDR) efficiency and reduction of non-homologous end-joining repair (NHEJ) are key technical requirements for precise gene editing.
因此,亟需一种提高HDR效率的精准基因编辑系统。Therefore, there is an urgent need for a precise gene editing system that improves HDR efficiency.
发明内容Contents of the invention
本发明旨在至少在一定程度上解决现有技术中存在的技术问题至少之一。The present invention aims to solve at least one of the technical problems existing in the prior art at least to a certain extent.
为此,本发明提供了一种5’→3’核酸外切酶在基因编辑系统中的用途和基因编辑系统及其编辑方法,通过将5’→3’核酸外切酶引入基因编辑系统中,能够提高同源重组修复(HDR)效率、降低非同源末端链接修复(NHEJ),以实现精准基因编辑需要说明的是,本发明是基于发明人的下列工作而完成的:To this end, the present invention provides a use of a 5'→3' exonuclease in a gene editing system, a gene editing system and an editing method thereof, by introducing the 5'→3' exonuclease into the gene editing system , can improve homologous recombination repair (HDR) efficiency, reduce non-homologous end-joining repair (NHEJ), so as to realize precise gene editing. It should be noted that the present invention is completed based on the following work of the inventors:
精准基因编辑技术由两个相对独立的部分组成:第一部分是通过核酸酶,识别并切割目的基因位点,在待编辑位点附近制造出核酸损伤;第二部分是利用细胞内源的核酸损伤修复过程引入特定的核酸改变。精准基因编辑技术的技术差别集中在第二部分,其中主要的技术路线包括寡核苷酸编辑模板介导的同源重组(ssODN HDR)、单碱基编辑(Base Editing,BE)、引导编辑(Prime Editing,PE)和双链长链核酸编辑模板介导的同源重组(dsDNA HDR)等。上述技术路线有比较明确的局限性:(1)编辑范围小,ssODN HDR、BE、PE主要在核酸损伤附近(10个碱基以内)产生编辑,因而绝大多数基因组位点不能被该方法有效编辑;其中,BE除了编辑范围小之外,只能引入非常有限类型的碱基改变,因而只限于特殊情况的基因编辑应用;(2)效率低,虽然dsDNA HDR技术路线具有实现全基因组位点的各种类型的编辑的潜力,但该方法效率极低,且伴随不可控的随机基因突变(非同源重组,NHEJ),限制其实际使用。The precision gene editing technology consists of two relatively independent parts: the first part is to identify and cut the target gene site through nuclease, and create nucleic acid damage near the site to be edited; the second part is to use the endogenous nucleic acid damage of the cell The repair process introduces specific nucleic acid changes. The technical differences of precision gene editing technology are concentrated in the second part, in which the main technical routes include oligonucleotide editing template-mediated homologous recombination (ssODN HDR), single base editing (Base Editing, BE), guided editing ( Prime Editing, PE) and double-stranded long-chain nucleic acid editing template-mediated homologous recombination (dsDNA HDR), etc. The above technical route has relatively clear limitations: (1) The editing range is small, and ssODN HDR, BE, and PE are mainly edited near nucleic acid damage (within 10 bases), so most genomic sites cannot be effectively edited by this method Editing; Among them, BE can only introduce very limited types of base changes in addition to the small editing range, so it is limited to gene editing applications in special cases; (2) The efficiency is low, although the dsDNA HDR technology route has the ability to realize genome-wide sites However, this method is extremely inefficient and accompanied by uncontrollable random gene mutation (non-homologous recombination, NHEJ), which limits its practical use.
基于此,发明人通过大量试验发现,核酸损伤位点5’→3’方向的核酸外切效率是dsDNA HDR技术路线中编辑效率被限制的主要因素;且非哺乳动物内源的5’→3’方向的核酸外切酶,尤其是来源于噬菌体的5’→3’方向的核酸外切酶,具有广泛的高度活跃的生物体内活性。发明人通过试验还发现,在基因编辑过程中,通过外源表达并招募噬菌体的5’→3’方向的核酸外切酶到目的基因编辑位点,可实现dsDNA HDR效率显著提高,同时极大的抑制NHEJ。Based on this, the inventors found through a large number of experiments that the exonucleic acid excision efficiency in the 5'→3' direction of the nucleic acid damage site is the main factor limiting the editing efficiency in the dsDNA HDR technical route; and the non-mammalian endogenous 5'→3 Exonucleases in the 'direction, especially those derived from the 5'→3' direction of bacteriophage, have a wide range of highly active in vivo activities. The inventor also found through experiments that during the gene editing process, the efficiency of dsDNA HDR can be significantly improved by exogenously expressing and recruiting the exonuclease in the 5'→3' direction of the phage to the target gene editing site, and at the same time greatly Inhibition of NHEJ.
在本发明的一个方面,本发明提出了一种5’→3’核酸外切酶在基因编辑系统中的用途。根据本发明的实施例,所述5’→3’核酸外切酶与所述基因编辑系统中的位点特异性核酸酶为非融合状态。In one aspect of the present invention, the present invention proposes the use of a 5'→3' exonuclease in a gene editing system. According to an embodiment of the present invention, the 5'→3' exonuclease is in a non-fused state with the site-specific nuclease in the gene editing system.
在本发明的第二方面,本发明提出了一种基因编辑系统。根据本发明的实施例,所述基因编辑系统包括:位点特异性核酸酶、5’→3’核酸外切酶和供体DNA;其中,所述5’→3’核酸外切酶与所述位点特异性核酸酶为非融合状态。In the second aspect of the present invention, the present invention provides a gene editing system. According to an embodiment of the present invention, the gene editing system includes: a site-specific nuclease, a 5'→3' exonuclease, and a donor DNA; wherein, the 5'→3' exonuclease and the The site-specific nucleases are non-fused.
在本发明的第三方面,本发明提出了一种对细胞进行基因编辑的方法。根据本发明的实施例,所述方法包括:将上述的基因编辑系统引入细胞。In the third aspect of the present invention, the present invention provides a method for gene editing of cells. According to an embodiment of the present invention, the method includes: introducing the above-mentioned gene editing system into cells.
本发明的附加方面和优点将在下面的描述中部分给出,部分将从下面的描述中变得明显,或通过本发明的实践了解到。 Additional aspects and advantages of the invention will be set forth in the description which follows, and in part will be obvious from the description, or may be learned by practice of the invention.
附图说明Description of drawings
本发明的上述和/或附加的方面和优点从结合下面附图对实施例的描述中将变得明显和容易理解,其中:The above and/or additional aspects and advantages of the present invention will become apparent and understandable from the description of the embodiments in conjunction with the following drawings, wherein:
图1为本发明实施例中TLR报告系统工作原理示意;Fig. 1 is a schematic diagram of the working principle of the TLR reporting system in the embodiment of the present invention;
图2为本发明实施例1中基因编辑效率柱状图;Figure 2 is a histogram of gene editing efficiency in Example 1 of the present invention;
图3为本发明实施例2中基因编辑效率柱状图;3 is a histogram of gene editing efficiency in Example 2 of the present invention;
图4为本发明实施例3中基因编辑效率柱状图;Figure 4 is a histogram of gene editing efficiency in Example 3 of the present invention;
图5为本发明实施例4中基因编辑效率柱状图;Figure 5 is a histogram of gene editing efficiency in Example 4 of the present invention;
图6为本发明实施例5中细胞NPM1位点的蛋白标记图;Figure 6 is a protein labeling map of the cell NPM1 site in Example 5 of the present invention;
图7为本发明实施例5中细胞FUS位点的蛋白标记图;Figure 7 is a protein labeling map of the cell FUS site in Example 5 of the present invention;
图8为本发明实施例6中PE-T7核酸外切酶的编辑系统示意图;8 is a schematic diagram of the editing system of PE-T7 exonuclease in Example 6 of the present invention;
图9为本发明实施例6中PE系统编辑效率柱形图;Fig. 9 is a histogram of editing efficiency of the PE system in Embodiment 6 of the present invention;
图10为本发明实施例7中有/无MS2招募系统的HDR通路修复效率柱形图;Figure 10 is a histogram of HDR pathway repair efficiency with/without MS2 recruitment system in Example 7 of the present invention;
图11为本发明实施例8中T7核酸外切酶在细胞内DSB处制造的不同长度单链DNA的热图;11 is a heat map of different lengths of single-stranded DNA produced by T7 exonuclease at DSB in the cell in Example 8 of the present invention;
图12为本发明实施例9中在Hela细胞系HEK3位点基因编辑效率柱状图;Figure 12 is a histogram of gene editing efficiency at the HEK3 locus of the Hela cell line in Example 9 of the present invention;
图13为本发明实施例9中在Hela细胞系DNMT1位点基因编辑效率柱状图;Figure 13 is a histogram of gene editing efficiency at the DNMT1 locus of the Hela cell line in Example 9 of the present invention;
图14为本发明实施例9中在HCT116细胞系HEK3位点基因编辑效率柱状图;Figure 14 is a histogram of gene editing efficiency at the HEK3 locus in the HCT116 cell line in Example 9 of the present invention;
图15为本发明实施例9中在HCT116细胞系DNMT1位点基因编辑效率柱状图;Figure 15 is a histogram of gene editing efficiency at the DNMT1 locus of the HCT116 cell line in Example 9 of the present invention;
图16为本发明实施例9中在U2OS细胞系HEK3位点基因编辑效率柱状图;Figure 16 is a histogram of gene editing efficiency at the HEK3 locus in the U2OS cell line in Example 9 of the present invention;
图17为本发明实施例9中在U2OS细胞系DNMT1位点基因编辑效率柱状图;Figure 17 is a histogram of gene editing efficiency at the DNMT1 locus of the U2OS cell line in Example 9 of the present invention;
图18为本发明实施例9中在mESC细胞系EMX1位点基因编辑效率柱状图;Figure 18 is a histogram of gene editing efficiency at the mESC cell line EMX1 site in Example 9 of the present invention;
图19为本发明实施例10中在大鼠原代神经元EMX1位点编辑效率柱状图;Figure 19 is a histogram of editing efficiency at the EMX1 site in rat primary neurons in Example 10 of the present invention;
图20为本发明实施例11中在TLR位点基因编辑效率柱状图;Figure 20 is a histogram of gene editing efficiency at TLR sites in Example 11 of the present invention;
图21为本发明实施例11中在HEK3位点基因编辑效率柱状图;Figure 21 is a histogram of gene editing efficiency at the HEK3 locus in Example 11 of the present invention;
图22为本发明实施例11中在DNMT1位点基因编辑效率柱状图;Figure 22 is a histogram of gene editing efficiency at the DNMT1 site in Example 11 of the present invention;
图23为本发明实施例11中在RNF2位点基因编辑效率柱状图;Figure 23 is a histogram of gene editing efficiency at the RNF2 locus in Example 11 of the present invention;
图24为本发明实施例11中在VEGFA位点基因编辑效率柱状图;Figure 24 is a histogram of gene editing efficiency at the VEGFA site in Example 11 of the present invention;
图25为本发明实施例12中在TLR位点基因编辑效率柱状图;Figure 25 is a histogram of gene editing efficiency at TLR sites in Example 12 of the present invention;
图26为本发明实施例12中在HEK3位点基因编辑效率柱状图;Figure 26 is a histogram of gene editing efficiency at the HEK3 locus in Example 12 of the present invention;
图27为本发明实施例12中在DNMT1位点基因编辑效率柱状图;Figure 27 is a histogram of gene editing efficiency at the DNMT1 site in Example 12 of the present invention;
图28为本发明实施例12中在RNF2位点基因编辑效率柱状图;Figure 28 is a histogram of gene editing efficiency at the RNF2 site in Example 12 of the present invention;
图29为本发明实施例12中在VEGFA位点基因编辑效率柱状图;Figure 29 is a histogram of gene editing efficiency at the VEGFA site in Example 12 of the present invention;
图30为本发明实施例13中在TLR位点基因编辑效率柱状图;Figure 30 is a histogram of gene editing efficiency at TLR sites in Example 13 of the present invention;
图31为本发明实施例13中在HEK3位点基因编辑效率柱状图;Figure 31 is a histogram of gene editing efficiency at the HEK3 locus in Example 13 of the present invention;
图32为本发明实施例13中在DNMT1位点基因编辑效率柱状图;Figure 32 is a histogram of gene editing efficiency at the DNMT1 site in Example 13 of the present invention;
图33为本发明实施例13中在RNF2位点基因编辑效率柱状图;Figure 33 is a histogram of gene editing efficiency at the RNF2 locus in Example 13 of the present invention;
图34为本发明实施例13中在VEGFA位点基因编辑效率柱状图;Figure 34 is a histogram of gene editing efficiency at the VEGFA site in Example 13 of the present invention;
图35为本发明实施例17中在引入插入突变基因编辑效率柱状图;Figure 35 is a histogram of gene editing efficiency in the introduction of insertion mutations in Example 17 of the present invention;
图36为本发明实施例17中在引入替换突变基因编辑效率柱状图;Fig. 36 is a histogram of gene editing efficiency in the introduction of replacement mutations in Example 17 of the present invention;
图37为本发明实施例14中NPM1双色标记结果图; Figure 37 is a graph showing the result of NPM1 double-color marking in Example 14 of the present invention;
图38为本发明实施例14中NPM1双色标记免疫荧光验证结果图;Figure 38 is a diagram of the results of NPM1 double-color labeling immunofluorescence verification in Example 14 of the present invention;
图39为本发明实施例15中高通量筛选结果柱状图;Fig. 39 is a histogram of high-throughput screening results in Example 15 of the present invention;
图40为本发明实施例15中高通量筛选结果代表性结果图;Figure 40 is a representative result diagram of the high-throughput screening results in Example 15 of the present invention;
图41为本发明实施例15中高通量筛选结果免疫荧光验证结果图;Figure 41 is a diagram of the results of immunofluorescence verification of the high-throughput screening results in Example 15 of the present invention;
图42为本发明实施例16中polyQ编辑验证结果图;Figure 42 is a diagram of the verification results of polyQ editing in Example 16 of the present invention;
图43为本发明实施例16中polyQ长度对ATXN2应激反应影响结果柱状图。Fig. 43 is a histogram of the effect of polyQ length on ATXN2 stress response in Example 16 of the present invention.
具体实施方式Detailed ways
下面详细描述本发明的实施例。下面描述的实施例是示例性的,仅用于解释本发明,而不能理解为对本发明的限制。Embodiments of the present invention are described in detail below. The embodiments described below are exemplary only for explaining the present invention and should not be construed as limiting the present invention.
需要说明的是,术语“第一”、“第二”仅用于描述目的,而不能理解为指示或暗示相对重要性或者隐含指明所指示的技术特征的数量。由此,限定有“第一”、“第二”的特征可以明示或者隐含地包括一个或者更多个该特征。进一步地,在本发明的描述中,除非另有说明,“多个”的含义是两个或两个以上。It should be noted that the terms "first" and "second" are only used for descriptive purposes, and cannot be understood as indicating or implying relative importance or implicitly indicating the quantity of indicated technical features. Thus, a feature defined as "first" and "second" may explicitly or implicitly include one or more of these features. Further, in the description of the present invention, unless otherwise specified, "plurality" means two or more.
如本文中所使用的术语“5’→3’核酸外切酶”是指从5’末端,即以5’至3’方向降解DNA的核酸外切酶。所关注的5’→3’核酸外切酶可以在平端处以及在某些实施方式中在3’和/或5’突出端处从dsDNA链的5’末端除去核苷酸。The term "5'→3' exonuclease" as used herein refers to an exonuclease that degrades DNA from the 5' end, ie in the 5' to 3' direction. The 5'→3' exonuclease of interest can remove nucleotides from the 5' end of a dsDNA strand at blunt ends and, in certain embodiments, at 3' and/or 5' overhangs.
在本文中所使用的术语“非融合状态”,指5’→3’核酸外切酶与基因编辑系统中的位点特异性核酸酶分别独立存在,两者之间并未存在任何连接关系,连接关系包括调控元件(例如但不限于,启动子序列、转录终止序列等)、核酸序列(例如,编码序列或开放读码框)连接和/或通过肽链/共价键连接。换句话说,在本文中所采用的5’→3’核酸外切酶和位点特异性核酸酶的最终发挥功能的状态是不构成融合蛋白。The term "non-fusion state" used herein refers to the independent existence of the 5'→3' exonuclease and the site-specific nuclease in the gene editing system, without any connection relationship between the two, Linkages include linkages to regulatory elements (eg, but not limited to, promoter sequences, transcription termination sequences, etc.), nucleic acid sequences (eg, coding sequences or open reading frames) and/or linkages via peptide chains/covalent bonds. In other words, the final functional state of the 5'→3' exonuclease and site-specific nuclease used herein does not constitute a fusion protein.
在本文中所使用的术语“gRNA”和“向导RNA”可互换使用,指的是能够与CRISPR核酸酶形成复合物并由于与靶序列具有一定互补性而能够将所述复合物靶向靶序列的RNA分子。例如,在基于Cas9的基因编辑系统中,gRNA通常由部分互补形成复合物的crRNA和tracrRNA分子构成,其中crRNA包含与靶序列具有足够相同性并且指导CRISPR复合物(Cas9+crRNA+tracrRNA)与该靶序列序列特异性地结合的序列。然而,本领域已知可以设计单向导RNA(sgRNA),其同时包含crRNA和tracrRNA的特征。而在基于Cas12a的基因编辑系统中,gRNA通常仅由成熟crRNA分子构成,其中crRNA包含的序列与靶序列具有足够相同性并且指导复合物(Cas12a+crRNA)与该靶序列序列特异性结合。基于所使用的CRISPR核酸酶和待编辑的靶序列设计合适的gRNA序列属于本领域技术人员的能力范围内。本领域已知可以设计pegRNA,pegRNA是sgRNA的一种衍生形式,在sgRNA的3’末端延伸出一小段RNA序列,该段序列包含Cas9切割位点两侧各约15~20nt的序列及中间待编辑的目的序列(若用于删除则无中间序列),一般适用于PE编辑系统,在执行sgRNA基本功能同时可作为逆转录酶的复制模板。As used herein, the terms "gRNA" and "guide RNA" are used interchangeably and refer to a gene that is capable of forming a complex with a CRISPR nuclease and targeting the complex to a target sequence due to a certain complementarity to the target sequence. sequence of RNA molecules. For example, in a Cas9-based gene editing system, the gRNA is usually composed of partially complementary crRNA and tracrRNA molecules that form a complex, where the crRNA contains sufficient identity to the target sequence and guides the CRISPR complex (Cas9+crRNA+tracrRNA) to the target sequence. target sequence A sequence to which a sequence specifically binds. However, it is known in the art that single-guide RNAs (sgRNAs) can be designed that contain features of both crRNAs and tracrRNAs. In the Cas12a-based gene editing system, the gRNA is usually only composed of mature crRNA molecules, where the crRNA contains a sequence that has sufficient identity with the target sequence and guides the complex (Cas12a+crRNA) to specifically bind to the target sequence. It is within the purview of those skilled in the art to design appropriate gRNA sequences based on the CRISPR nuclease used and the target sequence to be edited. It is known in the art that pegRNA can be designed. PegRNA is a derivative form of sgRNA. A short RNA sequence extends from the 3' end of sgRNA. The edited target sequence (if used for deletion, there is no intermediate sequence) is generally suitable for PE editing system, and can be used as a replication template for reverse transcriptase while performing the basic functions of sgRNA.
在本文中所使用的术语“招募系统”是指将多个一种蛋白富集到另一目的蛋白或目的区域的系统。该招募系统包括通过蛋白-蛋白相互作用、蛋白-小分子相互作用或蛋白-核酸相互作用,通过非共价键的分子间力,将多个一种蛋白富集到另一目的蛋白或目的区域的系统;该招募系统还可以包括通过化学修饰进行共价连接,将多个一种蛋白富集到另一目的蛋白或目的区域的系统。常用的蛋白-蛋白相互作用如suntag系统;蛋白-核酸相互作用如MS2-MCP、PP7-PCP系统等。As used herein, the term "recruitment system" refers to a system that enriches more than one protein into another protein or region of interest. The recruitment system involves the enrichment of multiple one protein to another target protein or target region through protein-protein interaction, protein-small molecule interaction or protein-nucleic acid interaction through non-covalent intermolecular force system; the recruitment system may also include a system for covalently linking multiple proteins to another target protein or target region through chemical modification. Commonly used protein-protein interactions such as the suntag system; protein-nucleic acid interactions such as MS2-MCP, PP7-PCP systems, etc.
在本文中所使用的术语“基因组”不仅涵盖存在于细胞核中的染色体DNA,而且还包括存在于细胞的亚细胞组分(如线粒体)中的细胞器DNA。The term "genome" as used herein encompasses not only chromosomal DNA present in the nucleus but also organelle DNA present in subcellular components of cells such as mitochondria.
在本文中所使用的术语“动物细胞”包括适于基因组编辑的任何动物体的细胞。动物体的实例包括但不限于,哺乳动物如人、小鼠、大鼠、猴、犬、猪、羊、牛、猫;家禽如鸡、鸭、鹅。The term "animal cell" as used herein includes cells of any animal body suitable for genome editing. Examples of animal bodies include, but are not limited to, mammals such as humans, mice, rats, monkeys, dogs, pigs, sheep, cows, and cats; poultry such as chickens, ducks, and geese.
本发明提出了一种5’→3’核酸外切酶在基因编辑系统中的用途和基因编辑系统及其编辑方法,下面将分别对其进行详细描述。The present invention proposes a use of a 5'→3' exonuclease in a gene editing system, a gene editing system and an editing method thereof, which will be described in detail below.
5’→3’核酸外切酶在基因编辑系统中的用途 Use of 5'→3' exonuclease in gene editing system
在本发明的一个方面,本发明提出了一种5’→3’核酸外切酶在基因编辑系统中的用途。根据本发明的实施例,该5’→3’核酸外切酶与基因编辑系统中的位点特异性核酸酶为非融合状态。In one aspect of the present invention, the present invention proposes the use of a 5'→3' exonuclease in a gene editing system. According to an embodiment of the present invention, the 5'→3' exonuclease is in a non-fused state with the site-specific nuclease in the gene editing system.
HDR通路修复在基因编辑系统中是最精准的一种修复通路,相比于其他修复通路,HDR通路更合适用于基因治疗的修复通路,但目前却没有被开发出高效的基因编辑工具。其原因为,HDR修复通路只活跃在细胞周期的S/G2期,而在细胞的其他时期都被多种调控机制抑制,但即使在S/G2期,NHEJ也会与HDR通路竞争。且通过试验发现,利用Cas9基于HDR通路的基因编辑在细胞系中只有大约1%的效率,而在没有细胞周期的不分裂细胞中几乎不发生HDR修复。因此,针对如此低的效率,使其无法作为实验工具,更无法作为治疗手段。HDR pathway repair is the most accurate repair pathway in the gene editing system. Compared with other repair pathways, the HDR pathway is more suitable for gene therapy repair pathways, but no efficient gene editing tools have been developed yet. The reason is that the HDR repair pathway is only active in the S/G2 phase of the cell cycle, and is inhibited by various regulatory mechanisms in other phases of the cell, but even in the S/G2 phase, NHEJ will compete with the HDR pathway. And it was found through experiments that the gene editing based on the HDR pathway using Cas9 has only about 1% efficiency in cell lines, and HDR repair hardly occurs in non-dividing cells without a cell cycle. Therefore, for such a low efficiency, it cannot be used as an experimental tool, let alone a treatment.
基于此,发明人通过对基因编辑技术的深入研究,发现核酸损伤位点5’方向的核酸外切效率是基因编辑系统的技术路线中编辑效率被限制的主要因素;并且,非哺乳动物内源的5’方向的核酸外切酶,具有广泛的高度活跃的生物体内活性。Based on this, the inventors, through in-depth research on gene editing technology, found that the exonucleic acid excision efficiency in the 5' direction of the nucleic acid damage site is the main factor that limits the editing efficiency in the technical route of the gene editing system; and, non-mammalian endogenous Exonuclease in the 5' direction, has a wide range of highly active in vivo activities.
具体地,发明人通过试验发现,在基因编辑过程中,通过外源表达并招募5’→3’核酸外切酶到目的基因编辑位点,5’→3’核酸外切酶可对DNA双链断裂(DNA Double Strand Break,DSB)末端进行切割,以补充/代替细胞内源的核酸酶的切割作用,能够提高在HDR通路中间状态的长片段单链DNA的产生,进而提高HDR通路修复效率,同时极大的抑制NHEJ。Specifically, the inventors found through experiments that in the process of gene editing, through exogenous expression and recruitment of 5'→3' exonuclease to the target gene editing site, the 5'→3' exonuclease can double DNA DNA double strand break (DNA Double Strand Break, DSB) is cleaved at the end to supplement/replace the cleavage of endogenous nucleases in cells, which can increase the production of long fragments of single-stranded DNA in the middle state of the HDR pathway, thereby improving the repair efficiency of the HDR pathway , while greatly inhibiting NHEJ.
同时,发明人通过对分裂细胞(例如HEK293细胞)和不分裂细胞(例如神经细胞)的RNA测序分析发现,对于两种细胞类型,NHEJ修复通路的相关因子都有较高表达。但是,HDR通路发生几乎只存在于分裂细胞中,而不分裂细胞中缺乏CtIP等启动HDR通路末端切割步骤所需的蛋白,导致不分裂细胞中不发生HDR通路。并且,发明人发现,不分裂细胞中存在一定表达量的HDR通路下游链插入和复制等步骤的蛋白,而针对不分裂细胞的基因编辑系统中,发现若添加5’→3’核酸外切酶可完成对末端的切割,可以诱导HDR通路在不分裂细胞中发生。At the same time, the inventors found through RNA sequencing analysis of dividing cells (such as HEK293 cells) and non-dividing cells (such as nerve cells), that for both cell types, the related factors of the NHEJ repair pathway are highly expressed. However, the HDR pathway occurs almost only in dividing cells, and non-dividing cells lack CtIP and other proteins required to initiate the end-cleavage step of the HDR pathway, resulting in the absence of the HDR pathway in non-dividing cells. Moreover, the inventors found that in non-dividing cells, there is a certain expression level of proteins in steps such as insertion and replication of the downstream chain of the HDR pathway. Cleavage of the ends can be accomplished, and the HDR pathway can be induced in non-dividing cells.
因此,发明人通过于基因编辑系统中添加5’→3’核酸外切酶,提高了HDR通路修复相对于NHEJ修复的竞争力,同时,还可以使分裂细胞中除S/G2期之外的其他时期中也发生HDR通路修复,从而提高HDR通路修复效率,同时极大的抑制NHEJ;并且,本发明还发现,通过5’→3’核酸外切酶的添加,可在不分裂细胞中实现HDR通路。Therefore, the inventors improved the competitiveness of HDR pathway repair relative to NHEJ repair by adding 5'→3' exonuclease to the gene editing system. HDR pathway repair also occurs in other periods, thereby improving the efficiency of HDR pathway repair, while greatly inhibiting NHEJ; and, the present invention also found that through the addition of 5'→3' exonuclease, it can be achieved in non-dividing cells HDR pathway.
需要说明的是,5’→3’核酸外切酶在基因编辑系统中的用途,即为5’→3’核酸外切酶在基因编辑中的用途,换句话说,在本文中是指一种基因编辑的方法,该方法包含提供5’→3’核酸外切酶,和使用5’→3’核酸外切酶进行基因编辑。It should be noted that the use of 5'→3' exonuclease in gene editing system is the use of 5'→3' exonuclease in gene editing. In other words, it refers to a A method for gene editing, the method comprising providing a 5'→3' exonuclease, and using the 5'→3' exonuclease for gene editing.
根据本发明的实施例,所述基因编辑系统中采用的细胞为动物细胞,优选为哺乳动物细胞。发明人通过大量试验发现,通过添加5’→3’核酸外切酶提高HDR通路修复效率,适用于动物细胞的基因编辑系统中,尤其是适用于哺乳动物细胞中。According to an embodiment of the present invention, the cells used in the gene editing system are animal cells, preferably mammalian cells. The inventors have found through a large number of experiments that the repair efficiency of the HDR pathway can be improved by adding 5'→3' exonuclease, which is suitable for the gene editing system of animal cells, especially for mammalian cells.
根据本发明的实施例,所述述5’→3’核酸外切酶为T7核酸外切酶。According to an embodiment of the present invention, the 5'→3' exonuclease is T7 exonuclease.
发明人通过大量试验,对多种5’→3’核酸外切酶(简称核酸外切酶)加入到基因编辑系统中进行比较,最终发现,T7核酸外切酶(又称T7噬菌体核酸外切酶)可在细胞内DSB处制造出长片段的单链DNA,该长片段的单链DNA出有利于HDR通路修复、而无法使用NHEJ通路修复,从而降低NHEJ通路修复效率,同时提高了HDR通路修复效率。Through a large number of experiments, the inventors compared various 5'→3' exonucleases (referred to as exonucleases) to the gene editing system, and finally found that T7 exonuclease (also known as T7 phage exonuclease) Enzyme) can produce long fragments of single-stranded DNA at the DSB in the cell. The long fragments of single-stranded DNA are conducive to the repair of the HDR pathway, but cannot be repaired by the NHEJ pathway, thereby reducing the repair efficiency of the NHEJ pathway and improving the HDR pathway. Repair efficiency.
根据本发明的实施例,所述T7核酸外切酶具有SEQ ID NO:1所示的氨基酸序列或者与其具有至少80%同源性的氨基酸序列。发明人通过大量试验发现,采用与T7核酸外切酶具有同源性的核酸外切酶,均能够降低NHEJ通路修复效率,同时提高了HDR通路修复效率。According to an embodiment of the present invention, the T7 exonuclease has the amino acid sequence shown in SEQ ID NO: 1 or an amino acid sequence having at least 80% homology therewith. The inventors have found through a large number of experiments that using an exonuclease homologous to T7 exonuclease can reduce the repair efficiency of the NHEJ pathway and improve the repair efficiency of the HDR pathway.
T7核酸外切酶的氨基酸序列如下所示(SEQ ID NO:1):
The amino acid sequence of T7 exonuclease is shown below (SEQ ID NO: 1):
基因编辑系统 gene editing system
在本发明的又一方面,本发明提出了一种基因编辑系统。根据本发明的实施例,该基因编辑系统包含:位点特异性核酸酶、5’→3’核酸外切酶和供体DNA;其中,所述5’→3’核酸外切酶与所述位点特异性核酸酶为非融合状态。In yet another aspect of the present invention, the present invention provides a gene editing system. According to an embodiment of the present invention, the gene editing system comprises: a site-specific nuclease, a 5'→3' exonuclease and a donor DNA; wherein, the 5'→3' exonuclease and the Site-specific nucleases are non-fused.
发明人通过试验发现,将上述位点特异性核酸酶、5’→3’核酸外切酶和供体DNA递送至细胞内后,位点特异性核酸酶于细胞内制造出DSB,然后5’→3’核酸外切酶在细胞系中发挥功能,产生长片段单链DNA,从而改变编辑结果中的HDR/NHEJ比例,降低NHEJ通路的修复效率,同时提高了HDR通路的修复效率。The inventors have found through experiments that after the above-mentioned site-specific nuclease, 5'→3' exonuclease and donor DNA are delivered into the cell, the site-specific nuclease produces a DSB in the cell, and then the 5' →The 3'exonuclease functions in the cell line to generate long fragments of single-stranded DNA, thereby changing the HDR/NHEJ ratio in the editing results, reducing the repair efficiency of the NHEJ pathway, and improving the repair efficiency of the HDR pathway.
需要说明的是,本发明的的递送方法不受限制,可根据具体的需求进行选择,具体可为,质粒过表达系统:包括质粒化学转染、质粒电穿孔等;病毒表达系统:腺相关病毒(AAV)侵染、慢病毒(lentivirus)侵染等;其他表达系统:包括蛋白质/RNA复合物电穿孔、mRMA电穿孔、mRMA化学转染等。It should be noted that the delivery method of the present invention is not limited, and can be selected according to specific needs, specifically, plasmid overexpression system: including plasmid chemical transfection, plasmid electroporation, etc.; viral expression system: adeno-associated virus (AAV) infection, lentivirus infection, etc.; other expression systems: including protein/RNA complex electroporation, mRMA electroporation, mRMA chemical transfection, etc.
在本文中,术语“供体DNA”包括待插入细胞DNA(如基因组DNA、线粒体DNA或病毒DNA)中的核酸序列。供体核酸序列可以通过细胞表达。供体核酸可以是外源的,对于细胞是外来的或在细胞内是非天然存在的。As used herein, the term "donor DNA" includes a nucleic acid sequence to be inserted into cellular DNA such as genomic DNA, mitochondrial DNA or viral DNA. The donor nucleic acid sequence can be expressed by the cell. The donor nucleic acid can be exogenous, foreign to the cell or non-naturally occurring within the cell.
供体DNA与可以被DNA结合结构域结合的序列相关。例如,供体DNA可以与DNA结合结构域的共有序列相邻,或者可以与共有序列存在于相同载体上,或者可以与共有序列存在于相同多核苷酸上。The donor DNA is associated with a sequence that can be bound by the DNA binding domain. For example, the donor DNA may be adjacent to the consensus sequence of the DNA binding domain, or may be present on the same vector as the consensus sequence, or may be present on the same polynucleotide as the consensus sequence.
根据本发明的实施例,所述位点特异性核酸酶的添加量为2~50ng。发明人经过大量实验得到上述较优添加量,由此,可进一步填提高的HDR通路的修复效率。According to an embodiment of the present invention, the added amount of the site-specific nuclease is 2-50 ng. The inventor obtained the above-mentioned optimal addition amount through a large number of experiments, thus, the repair efficiency of the improved HDR channel can be further filled.
根据本发明的实施例,所述5’→3’核酸外切酶的添加量为40~120ng。发明人经过大量实验得到上述较优添加量,由此,可进一步填提高的HDR通路的修复效率。并且,发明人发现,5’→3’核酸外切酶的浓度过高则会破坏NHEJ和HDR两种编辑的发生。According to an embodiment of the present invention, the added amount of the 5'→3' exonuclease is 40-120ng. The inventor obtained the above-mentioned optimal addition amount through a large number of experiments, thus, the repair efficiency of the improved HDR channel can be further filled. Moreover, the inventors found that an excessively high concentration of 5'→3' exonuclease would disrupt the occurrence of both NHEJ and HDR editing.
根据本发明的实施例,所述供体DNA的添加量为0.2~100ng。发明人经过大量实验得到上述较优添加量,由此,可进一步填提高的HDR通路的修复效率。According to an embodiment of the present invention, the added amount of the donor DNA is 0.2-100 ng. The inventor obtained the above-mentioned optimal addition amount through a large number of experiments, thus, the repair efficiency of the improved HDR channel can be further filled.
根据本发明的实施例,所述5’→3’核酸外切酶为T7核酸外切酶。由此,相对于其他5’→3’核酸外切酶,T7核酸外切酶可在细胞内DSB处制造出长片段的单链DNA,该长片段的单链DNA出现有利于HDR通路修复、抑制NHEJ通路修复,从而降低NHEJ通路,同时提高了HDR通路的修复效率。According to an embodiment of the present invention, the 5'→3' exonuclease is T7 exonuclease. Therefore, compared with other 5'→3' exonucleases, T7 exonuclease can produce long fragments of single-stranded DNA at DSBs in cells, and the appearance of this long fragment of single-stranded DNA is conducive to the repair of HDR pathway, Inhibit the repair of NHEJ pathway, thereby reducing the NHEJ pathway, while improving the repair efficiency of the HDR pathway.
根据本发明的实施例,所述T7核酸外切酶具有SEQ ID NO:1所示的氨基酸序列或者与其具有至少80%同源性的氨基酸序列。发明人通过大量试验发现,采用与T7核酸外切酶具有同源性的核酸外切酶,均能够降低NHEJ通路修复效率,同时提高了HDR通路修复效率。According to an embodiment of the present invention, the T7 exonuclease has the amino acid sequence shown in SEQ ID NO: 1 or an amino acid sequence having at least 80% homology therewith. The inventors have found through a large number of experiments that using an exonuclease homologous to T7 exonuclease can reduce the repair efficiency of the NHEJ pathway and improve the repair efficiency of the HDR pathway.
根据本发明的实施例,所述位点特异性核酸酶选自核酸内切酶,具体地,所述位点特异性核酸酶可选自成簇规律间隔短回文重复、转录激活样效应因子核酸酶、锌指核酸酶、归巢内切酶、限制性核酸内切酶中的至少之一。根据本发明的实施例,在基因编辑系统的应用中,可根据需求使用上述位点特异性核酸酶制造DSB,具体类型不受限制。According to an embodiment of the present invention, the site-specific nuclease is selected from endonucleases, specifically, the site-specific nuclease can be selected from clustered regularly interspaced short palindromic repeats, transcription activation-like effectors At least one of nuclease, zinc finger nuclease, homing endonuclease, and restriction endonuclease. According to an embodiment of the present invention, in the application of the gene editing system, the above-mentioned site-specific nucleases can be used to produce DSBs according to requirements, and the specific types are not limited.
需要说明的是,针对上述位点特异性核酸酶可选自成簇规律间隔短回文重复、转录激活样效应因子核酸酶、锌指核酸酶、归巢内切酶等核酸内切酶中的至少之一,也可为在上述任一位点特异性核酸酶的基础上改造的核酸酶(例如引入点突变),具体类型不受限制。It should be noted that, the specific nucleases for the above sites can be selected from endonucleases such as clustered regularly interspaced short palindromic repeats, transcription activator-like effector nucleases, zinc finger nucleases, and homing endonucleases. At least one of them may also be a nuclease modified on the basis of any of the above-mentioned site-specific nucleases (for example, introducing a point mutation), and the specific type is not limited.
根据本发明的实施例,所述基因编辑系统进一步包含:gRNA。若基因编辑系统为CRISPR系统时,通过gRNA与簇规律间隔短回文重复(CRISPR/Cas9,下文简称Cas9共同制造出DSB。According to an embodiment of the present invention, the gene editing system further comprises: gRNA. If the gene editing system is a CRISPR system, gRNA and cluster regularly interspaced short palindromic repeats (CRISPR/Cas9, hereinafter referred to as Cas9) are used to jointly produce DSB.
根据本发明的实施例,所述gRNA包括选自crRNA/tracrRNA、sgRNA、pegRNA中的至少之一。根据本发明的实施例,在该基因编辑系统中,只要满足该向导RNA是能够与位点特异性核酸酶形成复合物并由于与靶序列具有一定互补性而能够将所述复合物靶向靶序列即可,可采用上述向导RNA(本发明中的向导RNA包含传统的向导RNA以及对传统的向导RNA序列优化改进后的向导RNA)中的至少之一,其中,具体类型不受限制。According to an embodiment of the present invention, the gRNA includes at least one selected from crRNA/tracrRNA, sgRNA, and pegRNA. According to an embodiment of the present invention, in the gene editing system, as long as the guide RNA is capable of forming a complex with the site-specific nuclease and can target the complex to the target due to certain complementarity with the target sequence The sequence is enough, and at least one of the above-mentioned guide RNAs (the guide RNAs in the present invention include traditional guide RNAs and guide RNAs optimized and improved on traditional guide RNA sequences) can be used, wherein the specific type is not limited.
根据本发明的实施例,该基因编辑系统进一步包含:招募系统。发明人通过试验发现,通过采用招募系统,使5’→3’核酸外切酶大量富集,可进一步提高HDR通路的修复效率,且降低NHEJ通路的修复效率。According to an embodiment of the present invention, the gene editing system further includes: a recruitment system. The inventors found through experiments that by adopting the recruitment system, a large amount of 5'→3' exonuclease can be enriched, which can further improve the repair efficiency of the HDR pathway and reduce the repair efficiency of the NHEJ pathway.
需要说明的是,本发明中的招募系统只要满足能使5’→3’核酸外切酶大量富集即可,可为蛋白-小分子相互作用的招 募系统、核酸-蛋白相互作用的招募系统、蛋白-蛋白相互作用的招募系统,例如,可选择MS2/MCP系统、PP7-PCP系统、Suntag系统等作为招募系统,具体方式不受限制。It should be noted that as long as the recruitment system in the present invention can enrich 5'→3' exonuclease in a large amount, it can be a recruitment system for protein-small molecule interaction. Recruitment system, nucleic acid-protein interaction recruitment system, protein-protein interaction recruitment system, for example, MS2/MCP system, PP7-PCP system, Suntag system, etc. can be selected as the recruitment system, and the specific method is not limited.
根据本发明的实施例,若招募系统为MS2/MCP系统,gRNA为gRNA-MS2,5’→3’核酸外切酶为MCP-外切酶融合蛋白。由此,可使5’→3’核酸外切酶大量富集,进一步提高HDR通路的修复效率。According to an embodiment of the present invention, if the recruitment system is the MS2/MCP system, the gRNA is gRNA-MS2, and the 5'→3' exonuclease is an MCP-exonuclease fusion protein. In this way, the 5'→3' exonuclease can be enriched in large quantities, and the repair efficiency of the HDR pathway can be further improved.
根据本发明的实施例,所述供体DNA为采用硫代磷酸二酯键修饰的线性双链DNA和/或HMEJ形式的环状双链DNA。According to an embodiment of the present invention, the donor DNA is a linear double-stranded DNA modified with a phosphorothioate bond and/or a circular double-stranded DNA in the form of HMEJ.
发明人通过试验发现,通过引入末端保护手段保护供体DNA的末端,作为HDR通路的复制模板,在编辑开始前使供体DNA在细胞中稳定存在,进一步提高基因编辑的效率,并使编辑效率显著提高。根据本发明的实施例,相比于无修饰的线性双链DNA,本发明采用的上述供体DNA具有较高的HDR通路修复效率和HDR/NHEJ比例。The inventors found through experiments that by introducing end protection means to protect the end of the donor DNA as a replication template for the HDR pathway, the donor DNA can be stably present in the cell before editing begins, further improving the efficiency of gene editing, and making the editing efficiency Significantly increased. According to an embodiment of the present invention, compared with unmodified linear double-stranded DNA, the above-mentioned donor DNA used in the present invention has higher HDR pathway repair efficiency and HDR/NHEJ ratio.
根据本发明的实施例,所述供体DNA上的所述硫代磷酸二酯键的修饰个数大于2,优选为4-10。由此,可进一步提高HDR通路的修复效率和HDR/NHEJ比例。According to an embodiment of the present invention, the number of modifications of the phosphorothiodiester bonds on the donor DNA is greater than 2, preferably 4-10. As a result, the repair efficiency of the HDR pathway and the HDR/NHEJ ratio can be further improved.
根据本发明的实施例,所述基因编辑系统的技术路线包括寡核苷酸编辑模板介导的同源重组、单碱基编辑、引导编辑和双链长链核酸编辑模板介导的同源重组中的一种。According to an embodiment of the present invention, the technical route of the gene editing system includes homologous recombination mediated by an oligonucleotide editing template, single base editing, guided editing, and homologous recombination mediated by a double-stranded long-chain nucleic acid editing template One of.
本领域技术人员能够理解的是,前面针对5’→3’核酸外切酶在基因编辑系统中的用途所描述的特征和优点,同样适用于该基因编辑系统,在此不再赘述。Those skilled in the art can understand that the features and advantages described above for the use of the 5'→3' exonuclease in the gene editing system are also applicable to the gene editing system, and will not be repeated here.
对细胞进行基因编辑的方法Methods for gene editing cells
在本发明的另一方面,本发明提出了一种对细胞进行基因编辑的方法。根据本发明的实施例,该方法包括:将上述基因编辑系统引入细胞。发明人发现,将上述基因编辑系统引入细胞,能够提高细胞中HDR编辑效率以及提高HDR/NHEJ比例。In another aspect of the present invention, the present invention provides a method for gene editing of cells. According to an embodiment of the present invention, the method includes: introducing the above-mentioned gene editing system into cells. The inventors found that introducing the above-mentioned gene editing system into cells can improve the HDR editing efficiency and the HDR/NHEJ ratio in cells.
根据本发明的实施例,所述细胞为动物细胞,优选为哺乳动物细胞。发明人通过大量试验发现,通过添加5’→3’核酸外切酶提高HDR通路修复效率,适用于动物细胞的基因编辑系统中,尤其是适用于哺乳动物细胞中。According to an embodiment of the present invention, the cells are animal cells, preferably mammalian cells. The inventors have found through a large number of experiments that the repair efficiency of the HDR pathway can be improved by adding 5'→3' exonuclease, which is suitable for the gene editing system of animal cells, especially for mammalian cells.
根据本发明的实施例,所述细胞为终末分化原代细胞。发明人通过试验发现,T7核酸外切酶作为外源蛋白,其具有未明确受到细胞周期调控的特点,使得末端切割过程可以发生在细胞周期的各个时期,可诱导HDR通路在终末分化原代细胞等不分裂细胞发生。According to an embodiment of the present invention, the cells are terminally differentiated primary cells. The inventors found through experiments that T7 exonuclease, as an exogenous protein, has the characteristics of not being clearly regulated by the cell cycle, so that the end-cutting process can occur in various stages of the cell cycle, and can induce the HDR pathway in the terminal differentiation primary Cells such as non-dividing cells occur.
本领域技术人员能够理解的是,前面针对5’→3’核酸外切酶在基因编辑系统中的用途和基因编辑系统所描述的特征和优点,同样适用于该对细胞进行基因编辑的方法,在此不再赘述。Those skilled in the art can understand that the features and advantages described above for the use of 5'→3' exonuclease in the gene editing system and the gene editing system are also applicable to the method for gene editing of cells, I won't repeat them here.
下面将结合实施例对本发明的方案进行解释。本领域技术人员将会理解,下面的实施例仅用于说明本发明,而不应视为限定本发明的范围。实施例中未注明具体技术或条件的,按照本领域内的文献所描述的技术或条件或者按照产品说明书进行。所用试剂或仪器未注明生产厂商者,均为可以通过市购获得的常规产品。The solutions of the present invention will be explained below in conjunction with examples. Those skilled in the art will understand that the following examples are only for illustrating the present invention and should not be considered as limiting the scope of the present invention. If no specific technique or condition is indicated in the examples, it shall be carried out according to the technique or condition described in the literature in this field or according to the product specification. The reagents or instruments used were not indicated by the manufacturer, and they were all commercially available conventional products.
实施例1:不同5’→3’核酸外切酶在基因编辑系统对HDR通路的修复效率比较Example 1: Comparison of the repair efficiency of different 5'→3' exonucleases in the gene editing system for the HDR pathway
本实施例的基因编辑系统包含如下步骤:The gene editing system of this embodiment includes the following steps:
1、构建表达载体及供体DNA1. Construction of expression vector and donor DNA
sgRNA序列设计可通过benchling等设计网站完成。供体DNA建议使用700bp同源臂,构建为HMEJ供体形式。其余同源臂长度及供体DNA形式也可达成编辑目的。sgRNA sequence design can be done through design websites such as benchling. Donor DNA is recommended to use 700bp homology arms, constructed as HMEJ donor format. Other homology arm lengths and donor DNA forms can also be used for editing purposes.
通过CRISPR/Cas9系统制造DSB,通过MS2招募系统富集核酸外切酶,其中,核酸外切酶为T7噬菌体核酸外切酶、T5噬菌体核酸外切酶(氨基酸序列如SEQ ID NO:2所示)、lambda噬菌体核酸外切酶(氨基酸序列如SEQ ID NO:3所示)、大肠杆菌核酸外切酶DCLRE1B(氨基酸序列如SEQ ID NO:4所示)、大肠杆菌核酸外切酶RecE(氨基酸序列如SEQ ID NO:5所示)或人源核酸外切酶EXO1(氨基酸序列如SEQ ID NO:6所示),T7噬菌体核酸外切酶的氨基酸序列如SEQ ID NO:1所示。The DSB is produced by the CRISPR/Cas9 system, and the exonuclease is enriched by the MS2 recruitment system, wherein the exonuclease is T7 bacteriophage exonuclease, T5 phage exonuclease (the amino acid sequence is shown in SEQ ID NO: 2 ), lambda bacteriophage exonuclease (as shown in SEQ ID NO: 3), Escherichia coli exonuclease DCLRE1B (as shown in SEQ ID NO: 4), Escherichia coli exonuclease RecE (amino acid The sequence is shown in SEQ ID NO: 5) or human exonuclease EXO1 (amino acid sequence is shown in SEQ ID NO: 6), and the amino acid sequence of T7 bacteriophage exonuclease is shown in SEQ ID NO: 1.
T5噬菌体核酸外切酶的氨基酸序列:

Amino acid sequence of T5 bacteriophage exonuclease:

lambda噬菌体核酸外切酶的氨基酸序列:
Amino acid sequence of lambda bacteriophage exonuclease:
大肠杆菌核酸外切酶DCLRE1B的氨基酸序列:
Amino acid sequence of Escherichia coli exonuclease DCLRE1B:
大肠杆菌核酸外切酶RecE的氨基酸序列:
Amino acid sequence of Escherichia coli exonuclease RecE:
人源核酸外切酶EXO1的氨基酸序列:
Amino acid sequence of human exonuclease EXO1:
2、将编辑系统递送至目的细胞2. Delivery of editing system to target cells
通过质粒过表达体系实现,具体步骤如下:Through the plasmid overexpression system, the specific steps are as follows:
1)在96孔细胞培养板中接种293T细胞,于37℃,5%二氧化碳环境中培养过夜。1) Inoculate 293T cells in a 96-well cell culture plate and culture overnight at 37° C. in a 5% carbon dioxide environment.
2)通过瞬时转染的方法将含有编码Cas9蛋白和sgRNA-MS2的基因的表达载体(简称Cas9/sgRNA表达载体或Cas9/sgRNA)、以及含有编码MCP-外切酶融合蛋白的基因的表达载体(简称MCP-外切酶融合蛋白表达载体或MCP-外 切酶融合蛋白)与供体DNA一起导入细胞系,其中,转染的质粒添加量为:Cas9/sgRNA 50ng、MCP-外切酶融合蛋白100ng、供体DNA 50ng。若无特别说明,使用的供体DNA均为HMEJ供体,质粒总量200ng,转染试剂0.75μL。在细胞密度达到80%-90%时,通过PEI(1mg/mL)进行转染。或在细胞密度达到50%-60%时,通过lipo2000进行转染。2) The expression vector containing the gene encoding Cas9 protein and sgRNA-MS2 (abbreviated as Cas9/sgRNA expression vector or Cas9/sgRNA) and the expression vector containing the gene encoding MCP-exonuclease fusion protein were transferred by transient transfection (referred to as MCP-exonuclease fusion protein expression vector or MCP-exonuclease Dicer fusion protein) was introduced into the cell line together with donor DNA, wherein the amount of transfected plasmid added was: Cas9/sgRNA 50ng, MCP-exonuclease fusion protein 100ng, and donor DNA 50ng. Unless otherwise specified, the donor DNA used was HMEJ donor, the total amount of plasmid was 200ng, and the transfection reagent was 0.75 μL. Transfection was performed by PEI (1 mg/mL) when the cell density reached 80%-90%. Or transfect by lipo2000 when the cell density reaches 50%-60%.
其中,Cas9/sgRNA表达载体和MCP-外切酶融合蛋白表达载体可采用本领域常规技术构建获得,例如可参见“Gibson,D.G.et al.Enzymatic assembly of DNA molecules up to several hundred kilobases.Nat Methods 6,343-345,doi:10.1038/nmeth.1318(2009)”和“Potapov,V.et al.Comprehensive Profiling of Four Base Overhang Ligation Fidelity by T4DNA Ligase and Application to DNA Assembly.ACS Synth Biol 7,2665-2674,doi:10.1021/acssynbio.8b00333(2018)”。Among them, the Cas9/sgRNA expression vector and the MCP-exonuclease fusion protein expression vector can be constructed using conventional techniques in the art, for example, see "Gibson, D.G. et al. Enzymatic assembly of DNA molecules up to several hundred kilobases. Nat Methods 6,343 -345, doi:10.1038/nmeth.1318(2009)" and "Potapov, V. et al. Comprehensive Profiling of Four Base Overhang Ligation Fidelity by T4DNA Ligase and Application to DNA Assembly. ACS Synth Biol 7, 2665-2674, do i :10.1021/acssynbio.8b00333 (2018)".
Cas9蛋白的氨基酸序列为:
The amino acid sequence of the Cas9 protein is:
sgRNA-MS2的氨基酸序列如下所示,N表示为A、T、C、G中的任意一种,大写字母表示spacer区域,根据编辑位点设计,小写字母表示骨架区域:
The amino acid sequence of sgRNA-MS2 is as follows, N represents any one of A, T, C, and G, uppercase letters represent the spacer region, designed according to the editing site, and lowercase letters represent the backbone region:
MCP的氨基酸序列如下所示,MCP蛋白片段的C端和外切酶融合蛋白片段的N端相连:
The amino acid sequence of MCP is as follows, the C-terminal of the MCP protein fragment is connected to the N-terminal of the exonuclease fusion protein fragment:
本实施例中的蛋白片段相对应的核苷酸序列根据其氨基酸序列采用常规方法或常规的软件。The nucleotide sequences corresponding to the protein fragments in this example are based on their amino acid sequences using conventional methods or conventional software.
3)将转染后的细胞于37℃,5%二氧化碳环境中继续培养48h。3) Continue culturing the transfected cells for 48 hours at 37° C. in a 5% carbon dioxide environment.
4)根据实验目的,进行下游检测分析。4) According to the purpose of the experiment, carry out downstream detection and analysis.
3、检测编辑结果并进行下游应用3. Detect the editing results and apply them downstream
在细胞基因组插入TLR报告系统(如图1所示)进行检测,具体步骤如下:Insert the TLR reporter system (as shown in Figure 1) into the cell genome for detection, and the specific steps are as follows:
1)吸取孔中细胞培养基,缓慢加入50μL DPBS溶液清洗细胞。1) Aspirate the cell culture medium in the well, slowly add 50 μL DPBS solution to wash the cells.
2)吸去DPBS溶液,加入20μL的0.25%胰蛋白酶消化细胞1min。 2) The DPBS solution was sucked off, and 20 μL of 0.25% trypsin was added to digest the cells for 1 min.
3)加入80μL完全培养基,吹打重悬细胞。3) Add 80 μL of complete medium and resuspend the cells by pipetting.
4)将细胞转移至圆底96孔细胞培养板中,100g离心3min,弃去上清液。4) Transfer the cells to a round-bottom 96-well cell culture plate, centrifuge at 100 g for 3 min, and discard the supernatant.
5)使用100μL无血清培养基重悬细胞。5) Use 100 μL of serum-free medium to resuspend the cells.
6)使用流式细胞分析仪采集数据。mKate荧光信号对应NHEJ通路修复,EGFP荧光信号对应HDR通路修复。6) Collect data using a flow cytometer. The mKate fluorescence signal corresponds to the repair of the NHEJ pathway, and the EGFP fluorescence signal corresponds to the repair of the HDR pathway.
4、结果4. Results
HDR通路修复效率如图2所示,其中,横坐标表示核酸外切酶的物种来源,所有外切酶均与MS2形成融合蛋白,从左到右依次为:无外切酶阴性对照、T7噬菌体核酸外切酶、T5噬菌体核酸外切酶、lambda噬菌体核酸外切酶、大肠杆菌核酸外切酶DCLRE1B、大肠杆菌核酸外切酶RecE和人源核酸外切酶EXO1活性结构域;纵坐标表示修复效率。The repair efficiency of the HDR pathway is shown in Figure 2, where the abscissa indicates the species source of the exonuclease, and all the exonucleases form fusion proteins with MS2, from left to right: no exonuclease negative control, T7 phage Exonuclease, T5 bacteriophage exonuclease, lambda bacteriophage exonuclease, E. coli exonuclease DCLRE1B, E. coli exonuclease RecE and human exonuclease EXO1 active domain; ordinate indicates repair efficiency.
结果显示各来源的核酸外切酶均可在细胞系中发挥功能,改变编辑结果中的HDR/NHEJ比例。其中T7核酸外切酶最为显著的降低了NHEJ通路的修复效率,同时提高了HDR通路的修复效率。The results showed that exonucleases from each source were functional in cell lines, altering the HDR/NHEJ ratio in the edited results. Among them, T7 exonuclease most significantly reduced the repair efficiency of the NHEJ pathway, while improving the repair efficiency of the HDR pathway.
实施例2:不同供体DNA在基因编辑系统对HDR通路的修复效率比较Example 2: Comparison of the repair efficiency of different donor DNAs in the gene editing system for the HDR pathway
本实施例的基因编辑系统与实施例1的区别在于,核酸外切酶为T7核酸外切酶,本实施例选择不同的供体DNA进行实验,即为无修饰的线性双链DNA、硫代磷酸二酯键(PS)修饰的线性双链DNA(nPS指线性双链DNA的5’端有连续的n个硫代磷酸二酯键修饰)和HMEJ形式的环状双链DNA作为供体。The difference between the gene editing system in this example and Example 1 is that the exonuclease is T7 exonuclease. In this example, different donor DNAs are selected for experiments, that is, unmodified linear double-stranded DNA, sulfo Phosphodiester bond (PS)-modified linear double-stranded DNA (nPS refers to the 5' end of linear double-stranded DNA modified by n consecutive phosphorothioate bonds) and circular double-stranded DNA in the form of HMEJ were used as donors.
如图3所示,其中,横坐标表示供体DNA类型,PCR:线性双链DNA,nPS:5’末端有n个PS修饰的线性双链DNA,HMEJ:HMEJ形式环状双链DNA;纵坐标表示修复效率。As shown in Figure 3, where the abscissa indicates the type of donor DNA, PCR: linear double-stranded DNA, nPS: linear double-stranded DNA with n PS modifications at the 5' end, HMEJ: circular double-stranded DNA in the form of HMEJ; vertical Coordinates indicate repair efficiency.
结果显示在引入末端保护手段后,HDR编辑效率显著提高,其中HMEJ供体具有最高的HDR编辑效率和HDR/NHEJ比例。The results showed that after the introduction of end protection means, the HDR editing efficiency was significantly improved, and the HMEJ donor had the highest HDR editing efficiency and HDR/NHEJ ratio.
实施例3:对切割位点附近不同长度位点的编辑效率的研究Example 3: Research on the editing efficiency of sites of different lengths near the cleavage site
本实施例使用HMEJ双链DNA供体在距离切割位点100bp范围内的不同位置分别引入连续的三碱基突变(nBP指引入的突变位置与Cas9切割位点相距n个碱基对),通过二代测序分析表征基因编辑效率,其余步骤同实施例1。In this example, the HMEJ double-stranded DNA donor is used to introduce consecutive three-base mutations at different positions within 100 bp from the cutting site (nBP refers to the distance between the introduced mutation position and the Cas9 cutting site by n base pairs). The gene editing efficiency was characterized by next-generation sequencing analysis, and the rest of the steps were the same as in Example 1.
其中,二代测序分析表征基因编辑效率的具体步骤如下:Among them, the specific steps of next-generation sequencing analysis to characterize gene editing efficiency are as follows:
1.使用0.25%胰蛋白酶消化并收集细胞。1. Use 0.25% trypsin to digest and collect the cells.
2.500g离心沉淀细胞,去除上清液。2. Centrifuge at 500g to pellet the cells and remove the supernatant.
3.若瞬时转染的表达载体上带有荧光标签,可使用流式分选技术,根据荧光标签筛选出成功导入编辑系统的细胞,再进行下游步骤。该方法一般适用于转染效率较低的细胞系。3. If the transiently transfected expression vector has a fluorescent label, flow sorting technology can be used to screen out the cells successfully introduced into the editing system according to the fluorescent label, and then proceed to the downstream steps. This method is generally applicable to cell lines with low transfection efficiency.
4.使用Quick extract(Epicentre)试剂重悬细胞,65℃孵育15min,68℃孵育15min,95℃孵育10min,获取基因组片段溶液。4. Use Quick extract (Epicentre) reagent to resuspend the cells, incubate at 65°C for 15 minutes, at 68°C for 15 minutes, and at 95°C for 10 minutes to obtain the genome fragment solution.
5.使用编辑位点附近同源臂区域外的一段引物对编辑位点附近的基因组序列进行PCR扩增。扩增进行35个循环,用以消除供体DNA的干扰。5. Use a primer outside the homology arm region near the editing site to perform PCR amplification of the genomic sequence near the editing site. Amplification was performed for 35 cycles to eliminate interference from donor DNA.
6.将步骤5中的产物按1:10用超纯水稀释,取1μL作为模板,使用编辑位点附近的一对引物按照二代测序要求进行建库,该步骤扩增不超过20个循环。该步骤要求PCR产物全长150bp-200bp(不计二代测序接头部分)。6. Dilute the product in step 5 with ultrapure water at a ratio of 1:10, take 1 μL as a template, and use a pair of primers near the editing site to build a library according to the requirements of next-generation sequencing. This step does not exceed 20 cycles of amplification . This step requires the full length of the PCR product to be 150bp-200bp (excluding the part of the next-generation sequencing adapter).
7.使用CRISPResso2(具体参考文献Li X,Wang Y,Liu Y,et al.Base editing with a Cpf1-cytidine deaminase fusion[J].Nat Biotechnol,2018,36(4):324-327)对测序结果进行分析,统计NHEJ通路修复和HDR通路修复所占比例。7. Use CRISPResso2 (specific references Li X, Wang Y, Liu Y, et al. Base editing with a Cpf1-cytidine deaminase fusion [J]. Nat Biotechnol, 2018, 36(4): 324-327) to sequence the results Analyze and count the proportion of NHEJ pathway repair and HDR pathway repair.
如图4所示,其中,横坐标表示突变位点距离切割位点的距离,纵坐标表示修复效率。结果显示距切割位点距离更近的编辑位点具有更高的编辑效率,在距切割位点至少100bp长度处依旧可以完成有效编辑。As shown in Figure 4, where the abscissa represents the distance between the mutation site and the cleavage site, and the ordinate represents the repair efficiency. The results show that the editing site closer to the cutting site has higher editing efficiency, and effective editing can still be completed at a distance of at least 100 bp from the cutting site.
实施例4:对基因组的不同位点的编辑效率的研究Example 4: Research on the Editing Efficiency of Different Sites of the Genome
本实施例使用HMEJ双链DNA供体在细胞基因组的HEK3、DNMT1、RNF2、VEGFA四个内源位点分别进行编辑,通过二代测序(参考实施例3)分析表征基因编辑效率,其他步骤同实施例1。其中,本实施例中的质粒转染量为Cas9/sgRNA20ng、T7核酸外切酶80ng、供体DNA 100ng。 In this example, the HMEJ double-stranded DNA donor was used to edit the four endogenous sites of HEK3, DNMT1, RNF2, and VEGFA in the cell genome respectively, and the gene editing efficiency was analyzed and characterized by next-generation sequencing (refer to Example 3), and other steps were the same Example 1. Wherein, the amount of plasmid transfection in this embodiment is 20ng of Cas9/sgRNA, 80ng of T7 exonuclease, and 100ng of donor DNA.
如图5所示,其中,横坐标表示编辑位点所在基因名称或编辑位点名称,纵坐标表示修复效率,T7+为编辑过程中添加T7核酸外切酶,T7-为编辑过程中不添加T7核酸外切酶。结果显示,基因组的不同位点在编辑效率和修复通路选择上有所差异,但在所有位点均可完成高效率的HDR通路编辑。As shown in Figure 5, where the abscissa indicates the name of the gene where the editing site is located or the name of the editing site, and the ordinate indicates the repair efficiency, T7+ indicates the addition of T7 exonuclease during the editing process, and T7- indicates that T7 is not added during the editing process exonuclease. The results showed that different sites in the genome had differences in editing efficiency and repair pathway selection, but high-efficiency HDR pathway editing could be completed at all sites.
实施例5:基因编辑系统在细胞系中的蛋白标记作用Example 5: Protein labeling effect of gene editing system in cell lines
本实施例使用HMEJ双链DNA供体,在基因组的NPM1基因位点C端(图6)和FUS基因位点N端(图7)分别敲入FlAsH短肽标签(具体参考文献Griffin B A,Adams S R,Tsien R Y.Specific covalent labeling of recombinant protein molecules inside live cells[J].Science,1998,281(5374):269-72和Thorn K S,Naber N,Matuska M,et al.A novel method of affinity-purifying proteins using a bis-arsenical fluorescein[J].Protein Sci,2000,9(2):213-7),在活细胞中观察目的蛋白的细胞定位。其中,本实施例中的质粒转染量为Cas9 20ng、T7核酸外切酶80ng、供体DNA 100ng。In this example, the HMEJ double-stranded DNA donor was used to knock in FlAsH short peptide tags at the C-terminus of the NPM1 gene site (Figure 6) and the N-terminus of the FUS gene site (Figure 7) of the genome (specific references Griffin B A, Adams S R, Tsien R Y.Specific covalent labeling of recombinant protein molecules inside live cells[J].Science,1998,281(5374):269-72 and Thorn K S,Naber N,Matuska M,et al.A novel method of affinity-purifying proteins using a bis-arsenical fluorescein[J]. Protein Sci, 2000,9(2):213-7), to observe the cellular localization of the target protein in living cells. Wherein, the amount of plasmid transfection in this embodiment is Cas9 20ng, T7 exonuclease 80ng, donor DNA 100ng.
结果显示,该编辑工具可以非常高效的引入荧光标签,从而在活细胞中显示蛋白的细胞定位(NPM1定位:核仁,FUS定位:细胞核内核仁以外区域;左图:荧光标记内源蛋白,右图:hoechst标记染色质),从而可便捷实现蛋白内源表达水平的功能研究。The results show that this editing tool can introduce fluorescent tags very efficiently, thereby displaying the cellular localization of proteins in living cells (NPM1 localization: nucleolus, FUS localization: the area outside the nucleolus; left: fluorescently labeled endogenous proteins, right Figure: hoechst marks chromatin), which can facilitate the functional study of the endogenous expression level of the protein.
实施例6:T7核酸外切酶在PE编辑系统中编辑效率的研究Example 6: Research on Editing Efficiency of T7 Exonuclease in PE Editing System
本实施例对PE编辑系统进行改进,将Cas9切刻酶(Cas9n)突变恢复为野生型Cas9,延长引物结合区域并与T7核酸外切酶联用,通过二代测序(参考实施例3)分析表征基因编辑效率,转染时使用50ng含有编码Cas9-MMLV融合蛋白/pegRNA的基因的表达质粒,100ng含有编码T7核酸外切酶的基因的表达质粒作为实验组;50ng含有编码Cas9n-MMLV融合蛋白/pegRNA的基因的表达质粒,100ng pUC19质粒作为PE系统对照,其他步骤同实施例1。This example improves the PE editing system, restores the Cas9 nickase (Cas9n) mutation to wild-type Cas9, extends the primer binding region and uses it in conjunction with T7 exonuclease, and analyzes it by next-generation sequencing (reference example 3) To characterize gene editing efficiency, use 50ng of expression plasmids containing genes encoding Cas9-MMLV fusion protein/pegRNA during transfection, 100ng of expression plasmids containing genes encoding T7 exonuclease as the experimental group; 50ng containing expression plasmids encoding Cas9n-MMLV fusion proteins The expression plasmid of the gene of/pegRNA, 100ng pUC19 plasmid is used as PE system control, and other steps are with embodiment 1.
如图8和9所示,结果显示对于测试的多个基因组位点,改进型的PE-T7编辑系统(即为px330-mmlv/t7)均表现出显著高于PE系统的基因编辑效率。(图中cutting指在该基因组位点处造成的总编辑效率,ins/del指按照预期引入插入/缺失突变的效率)。As shown in Figures 8 and 9, the results showed that the improved PE-T7 editing system (ie, px330-mmlv/t7) showed significantly higher gene editing efficiency than the PE system for multiple genomic sites tested. (cutting in the figure refers to the total editing efficiency caused at the genomic site, and ins/del refers to the efficiency of introducing insertion/deletion mutations as expected).
实施例7:针对招募系统的有无的比较Example 7: Comparison with the presence or absence of the recruitment system
本实施例的基因编辑系统与实施例1的区别仅在于,核酸外切酶为T7核酸外切酶,通过使用无MS2-loop结构的sgRNA作为对照,对比了有/无MS2招募系统的HDR通路修复效率。The difference between the gene editing system in this example and Example 1 is that the exonuclease is T7 exonuclease, and the HDR pathway with/without MS2 recruitment system was compared by using sgRNA without MS2-loop structure as a control Repair efficiency.
如图10所示,结果显示在引入MS2招募系统后,T7核酸外切酶抑制NHEJ通路,提高HDR通路的功能得到了进一步加强。As shown in Figure 10, the results showed that after the introduction of the MS2 recruitment system, T7 exonuclease inhibited the NHEJ pathway and enhanced the function of the HDR pathway.
实施例8:T7核酸外切酶在细胞内DSB处制造的单链DNA的研究Example 8: Study of Single-Stranded DNA Manufactured at Intracellular DSBs by T7 Exonuclease
本实施例中未使用供体DNA,仅引入Cas9/sgRNA与T7核酸外切酶作为实验组(即为T7+),在转染后不同时间点进行采样,通过酶切-荧光定量PCR法对单链DNA进行检测,对照物使用MCP蛋白代替T7核酸外切酶(即为T7-)(本实施例中使用6孔细胞培养板,转染时Cas9/sgRNA,T7核酸外切酶的含量分别为500ng、1500ng,PEI用量为12μL)。Donor DNA was not used in this example, only Cas9/sgRNA and T7 exonuclease were introduced as the experimental group (i.e., T7+), and samples were taken at different time points after transfection, and single samples were analyzed by enzyme digestion-fluorescent quantitative PCR method. stranded DNA is detected, and the control uses MCP protein instead of T7 exonuclease (being T7-) (in this embodiment, a 6-well cell culture plate is used, and during transfection, the contents of Cas9/sgRNA and T7 exonuclease are respectively 500ng, 1500ng, PEI dosage is 12μL).
如图11所示,结果显示在转染后一段时间内,加入T7核酸外切酶的样品DSB附近的单链DNA总量明显高于对照组。As shown in FIG. 11 , the results showed that within a period of time after transfection, the total amount of single-stranded DNA near the DSB of the sample added with T7 exonuclease was significantly higher than that of the control group.
实施例9:T7核酸外切酶在不同细胞系编辑效率的研究Example 9: Research on editing efficiency of T7 exonuclease in different cell lines
本实施例使用HMEJ双链DNA供体,在多种细胞系上进行了基因编辑。实验通过流式细胞分选技术筛选出成功导入编辑工具的细胞,通过二代测序(参考实施例3)分析表征基因编辑效率,(本实施例中使用24孔细胞培养板,使用lipo3000试剂进行转染,转染试剂用量为1.5μL,转染时Cas9/sgRNA,T7核酸外切酶和donor DNA的含量分别为:HCT116,50ng,250ng,200ng;U2OS:250ng,350ng,300ng;Hela,50ng,250ng,200ng;mESC:200ng,200ng,150ng)其他步骤同实施例1,检测结果如图12-18所示。结果显示,在各种细胞系中,引入T7核酸外切酶均可抑制NHEJ通路的修复,并显著提高HDR通路的修复效率,因此,本发明的基因编辑系统的高效编辑性能不依赖细胞体系。In this example, gene editing was performed on various cell lines using the HMEJ double-stranded DNA donor. In the experiment, the cells that were successfully introduced into the editing tool were screened by flow cytometry technology, and the gene editing efficiency was analyzed and characterized by next-generation sequencing (refer to Example 3). Transfection, the amount of transfection reagent is 1.5μL, the contents of Cas9/sgRNA, T7 exonuclease and donor DNA during transfection are: HCT116, 50ng, 250ng, 200ng; U2OS: 250ng, 350ng, 300ng; Hela, 50ng, 250ng, 200ng; mESC: 200ng, 200ng, 150ng) Other steps are the same as in Example 1, and the test results are shown in Figures 12-18. The results show that in various cell lines, the introduction of T7 exonuclease can inhibit the repair of the NHEJ pathway and significantly improve the repair efficiency of the HDR pathway. Therefore, the efficient editing performance of the gene editing system of the present invention does not depend on the cell system.
实施例10:T7核酸外切酶在原代大鼠神经元编辑效率的研究Example 10: Research on Editing Efficiency of T7 Exonuclease in Primary Rat Neurons
本实施例使用HMEJ双链DNA供体,使用Lonza-4D电转仪通过电穿孔方式对E18大鼠胚胎的皮层神经细胞进行表 达载体递送,使用20μL电穿孔体系,Cas9/sgRNA,T7核酸外切酶和donor DNA的含量分别为:500ng,1000ng,500ng(具体操作方法参考https://bioscience.lonza.com/lonza_bs/DE/en/download/product/asset/21243),通过一代测序分析表征基因编辑效率,其他步骤同实施例1,分析结果见图19。结果显示,该编辑工具可在大鼠原代神经细胞完成有效的基因编辑。因此,本发明的基因编辑系统的可在终末分化原代细胞实现HDR编辑。In this example, the HMEJ double-stranded DNA donor was used to express the cortical nerve cells of E18 rat embryos by electroporation using a Lonza-4D electroporation instrument. For vector delivery, use 20μL electroporation system, the contents of Cas9/sgRNA, T7 exonuclease and donor DNA are: 500ng, 1000ng, 500ng respectively (refer to https://bioscience.lonza.com/lonza_bs/DE for specific operation methods /en/download/product/asset/21243), the efficiency of gene editing was characterized by next-generation sequencing analysis, other steps were the same as in Example 1, and the analysis results are shown in Figure 19. The results show that the editing tool can complete effective gene editing in rat primary nerve cells. Therefore, the gene editing system of the present invention can realize HDR editing in terminally differentiated primary cells.
实施例11:特异性核酸内切酶转染浓度对编辑结果影响Example 11: Effect of specific endonuclease transfection concentration on editing results
本实施例使用HMEJ双链DNA供体在细胞基因组的不同位点分别进行编辑,通过二代测序(参考实施例3)分析表征基因编辑效率,其他步骤同实施例1,分析结果见图20-24。并测试了编辑中使用不同转染量的Cas9/sgRNA表达质粒,使用pUC19质粒补全转染总量。结果显示,当内切酶浓度较低时,可以获得较高的HDR/NHEJ比例,当内切酶浓度较高时,可以获得较高的HDR修复效率。In this example, HMEJ double-stranded DNA donors are used to edit different sites in the cell genome, and the gene editing efficiency is analyzed and characterized by next-generation sequencing (refer to Example 3). Other steps are the same as in Example 1. The analysis results are shown in Figure 20- twenty four. And tested the Cas9/sgRNA expression plasmids with different transfection amounts in editing, and used the pUC19 plasmid to complete the total transfection amount. The results showed that a higher HDR/NHEJ ratio could be obtained when the endonuclease concentration was lower, and a higher HDR repair efficiency could be obtained when the endonuclease concentration was higher.
实施例12:T7核酸外切酶转染浓度对编辑结果影响Example 12: Effect of T7 exonuclease transfection concentration on editing results
本实施例使用HMEJ双链DNA供体在细胞基因组的不同位点分别进行编辑,通过二代测序(参考实施例3)分析表征基因编辑效率,Cas9/sgRNA含量为20ng,其他步骤同实施例1,分析结果见图25-30。并测试了编辑中使用不同转染量的T7核酸外切酶表达质粒,使用MCP蛋白表达质粒补全转染总量。结果显示,随T7核酸外切酶浓度提高,NHEJ效率呈现下降趋势,HDR效率呈现先上升后下降趋势。In this example, HMEJ double-stranded DNA donors were used to edit different sites in the cell genome, and the gene editing efficiency was analyzed and characterized by next-generation sequencing (refer to Example 3). The content of Cas9/sgRNA was 20ng, and other steps were the same as in Example 1. , the analysis results are shown in Figure 25-30. And tested the use of T7 exonuclease expression plasmids with different transfection amounts in editing, and used the MCP protein expression plasmid to complete the total amount of transfection. The results showed that as the concentration of T7 exonuclease increased, the efficiency of NHEJ showed a downward trend, and the efficiency of HDR showed a trend of first increasing and then decreasing.
实施例13:供体DNA转染浓度对编辑结果影响Example 13: Effect of Donor DNA Transfection Concentration on Editing Results
本实施例使用HMEJ双链DNA供体在细胞基因组的不同位点分别进行编辑,通过二代测序(参考实施例3)分析表征基因编辑效率,Cas9/sgRNA含量为20ng,T7核酸外切酶含量为80ng,其他步骤同实施例1,分析结果见图30-34。并测试了编辑中使用不同转染量的供体DNA质粒,使用pUC19质粒补全转染总量。结果显示,随供体DNA浓度上升,NHEJ通路效率下降,HDR通路效率上升。In this example, HMEJ double-stranded DNA donors were used to edit different sites in the cell genome, and the gene editing efficiency was analyzed and characterized by next-generation sequencing (reference example 3). The content of Cas9/sgRNA was 20ng, and the content of T7 exonuclease 80ng, the other steps are the same as in Example 1, and the analysis results are shown in Figures 30-34. And tested the use of different transfection amounts of donor DNA plasmids in editing, using the pUC19 plasmid to complete the total amount of transfection. The results showed that as the concentration of donor DNA increased, the efficiency of NHEJ pathway decreased and that of HDR pathway increased.
实施例14:对NPM1进行双荧光敲入编辑Example 14: Dual fluorescent knock-in editing of NPM1
本实施例为实施例5的延伸应用。实验中同时使用在NPM1蛋白C端引入split-sfGFP标签的供体DNA(即为NPM1WT)和在NPM1蛋白C端引入致病的+4突变和FlAsH短肽标签的供体DNA(即为NPM1mut)。Cas9/sgRNA含量为50ng,T7核酸外切酶含量为80ng,供体DNA为100ng。其他步骤同实施例5。通过流式细胞仪分选得到了荧光标记双阳性的细胞,并通过共聚焦显微镜对荧光进行了观察(图37,(a):DAPI;(b):NPM1WT;(c):NPM1mut)。对引入的荧光标签与NPM1蛋白野生型/突变体免疫荧光的共定位分别进行观察(图38,(a)NPM1WT DAPI(b)NPM1WT荧光标签(c)NPM1WT免疫荧光染色(d)NPM1mut DAPI(e)NPM1mut荧光标签(f)NPM1mut免疫荧光染色。结果表明,该方法可以同时对一个细胞中同一基因的不同等位基因引入不同的突变,高效的制造双敲入杂合体细胞。This embodiment is an extended application of Embodiment 5. In the experiment, the donor DNA with the split-sfGFP tag introduced at the C-terminal of the NPM1 protein (that is, NPM1 WT ) and the donor DNA with the pathogenic +4 mutation and the FlAsH short peptide tag introduced at the C-terminal of the NPM1 protein (that is, the NPM1 mut ). The Cas9/sgRNA content is 50ng, the T7 exonuclease content is 80ng, and the donor DNA is 100ng. Other steps are the same as in Example 5. Fluorescence-labeled double-positive cells were sorted by flow cytometry, and the fluorescence was observed by confocal microscopy (Figure 37, (a): DAPI; (b): NPM1 WT ; (c): NPM1 mut ) . The co-localization of the introduced fluorescent label and NPM1 protein wild-type/mutant immunofluorescence was observed separately (Figure 38, (a) NPM1 WT DAPI (b) NPM1 WT fluorescent label (c) NPM1 WT immunofluorescence staining (d) NPM1 mut DAPI (e) NPM1 mut fluorescent label (f) NPM1 mut immunofluorescent staining. The results show that this method can introduce different mutations to different alleles of the same gene in a cell at the same time, and efficiently create double knock-in hybrid cells .
实施例15:通过基因编辑手段对FUS蛋白疾病相关突变体进行研究Example 15: Research on FUS protein disease-associated mutants by means of gene editing
本实施例为实施例5的延伸应用。实验中使用实施例5中获得的在FUS蛋白敲入FlAsH标签的细胞系。本实验通过基因编辑方法在FUS蛋白引入目前已发现的疾病相关点突变(本实施例以C端NLS区域点突变为例)并通过荧光显微镜观察突变体蛋白在细胞中的行为变化。Cas9/sgRNA含量为50ng,T7核酸外切酶含量为80ng,供体DNA各50ng。其他步骤同实施例5。使用500μM亚砷酸钠刺激细胞1h,通过对引入不同点突变的细胞中FUS蛋白进入应激颗粒(SGs)的比例进行观察,筛选得到了一组导致FUS定位于SGs的突变体(图39),突变体与野生型FUS亚细胞定位有明显差别(图40,(a)P525L突变体;(b)野生型)。然后对筛选结果通过单细胞培养和免疫荧光染色验证其突变与表型的对应关系,结果参见图41(图41,(a)WT DAPI;(b)WT FUS(c);Q519X DAPI;(d);Q519X FUS;(e)P525L DAPI;(f)P525L FUS)。结果表明,该方法可以高通量的对各种蛋白的点突变进行筛选和初步的功能鉴定。This embodiment is an extended application of Embodiment 5. The cell line in which the FlAsH tag was knocked in the FUS protein obtained in Example 5 was used in the experiment. In this experiment, a currently discovered disease-associated point mutation was introduced into the FUS protein by a gene editing method (this example takes a point mutation in the C-terminal NLS region as an example), and the behavioral changes of the mutant protein in cells were observed by a fluorescence microscope. The Cas9/sgRNA content is 50ng, the T7 exonuclease content is 80ng, and the donor DNA is 50ng each. Other steps are the same as in Example 5. The cells were stimulated with 500 μM sodium arsenite for 1 h, and by observing the ratio of FUS protein entering the stress granules (SGs) in cells introduced with different point mutations, a group of mutants that caused FUS to localize to SGs were screened (Figure 39) , the subcellular localization of mutant and wild-type FUS was significantly different (Fig. 40, (a) P525L mutant; (b) wild-type). Then, the screening results were verified by single-cell culture and immunofluorescence staining to verify the corresponding relationship between the mutation and the phenotype, and the results are shown in Figure 41 (Figure 41, (a) WT DAPI; (b) WT FUS (c); Q519X DAPI; (d) ); Q519X FUS; (e) P525L DAPI; (f) P525L FUS). The results show that this method can be used for high-throughput screening and preliminary functional identification of point mutations in various proteins.
实施例16:通过基因编辑手段研究ATXN2蛋白polyQ结构长度对蛋白功能的影响Example 16: Study on the effect of polyQ structure length of ATXN2 protein on protein function by means of gene editing
ATXN2蛋白包含一个谷氨酰胺重复序列结构域(polyQ),野生型蛋白polyQ长度为22-23,其序列异常延长与多种疾病的发病相关。本实施例使用实施例5的方式在ATXN2蛋白引入FlAsH荧光标签,使用实施例15的方式在ATXN2蛋白polyQ区域引入突变,构建了具有不同polyQ长度的ATXN2基因编辑细胞系(nQ指该基因编辑细胞系中ATXN2 蛋白polyQ结构域包含n个连续的谷氨酰胺)。我们使用Western-bloting检测ATXN2降解条带分子量变化,并通过一代测序对编辑结果进行了验证(图42,(a)Western-bloting 70kD-80kD ATXN2条带;(b)44Q细胞系polyQ序列一代测序结果)。并据此研究polyQ长度对ATXN2在细胞应激与恢复过程中行为的影响,通过Western-bloting进行了探究和总结(图43,左:WT;中:30Q;右:44Q;每组内分别为:应激前,应激后恢复0h,应激后恢复2h,应激后恢复6h)。结果表明,通过该方法可以避免研究突变体蛋白时引入浓度变化,对实验结果产生干扰。The ATXN2 protein contains a glutamine repeat sequence domain (polyQ). The length of polyQ in the wild-type protein is 22-23. The abnormal extension of its sequence is related to the pathogenesis of various diseases. In this example, the FlAsH fluorescent label was introduced into the ATXN2 protein by the method of Example 5, and the mutation was introduced into the polyQ region of the ATXN2 protein by the method of Example 15, and ATXN2 gene editing cell lines with different polyQ lengths were constructed (nQ refers to the gene editing cell ATXN2 in the system The protein polyQ domain contains n consecutive glutamines). We used Western-blotting to detect the molecular weight change of the ATXN2 degradation band, and verified the editing results by first-generation sequencing (Figure 42, (a) Western-blotting 70kD-80kD ATXN2 band; (b) polyQ sequence generation sequencing of 44Q cell line result). Based on this, the effect of polyQ length on the behavior of ATXN2 in the process of cell stress and recovery was explored and summarized by Western-blotting (Figure 43, left: WT; middle: 30Q; right: 44Q; each group was : before stress, recovery 0h after stress, recovery 2h after stress, recovery 6h after stress). The results show that this method can avoid the introduction of concentration changes when studying mutant proteins, which will interfere with the experimental results.
实施例17:测试基因编辑工具引入突变的长度范围Example 17: Testing the length range of mutations introduced by gene editing tools
本实施例使用引入插入突变或替换突变的供体DNA,其余实验方法同实施例3。如图35(插入突变)和36(替换突变)所示,其中,横坐标表示引入插入/替换突变的长度,纵坐标表示修复效率。结果显示,该编辑工具可有效编辑至少长达200bp的DNA片段。In this example, the donor DNA with insertion mutation or substitution mutation was used, and the rest of the experimental methods were the same as in Example 3. As shown in Figures 35 (insertion mutation) and 36 (replacement mutation), the abscissa represents the length of the insertion/replacement mutation introduced, and the ordinate represents the repair efficiency. The results show that the editing tool can effectively edit at least 200bp DNA fragments.
在本说明书的描述中,参考术语“一个实施例”、“一些实施例”、“示例”、“具体示例”、或“一些示例”等的描述意指结合该实施例或示例描述的具体特征、结构、材料或者特点包含于本发明的至少一个实施例或示例中。在本说明书中,对上述术语的示意性表述不必须针对的是相同的实施例或示例。而且,描述的具体特征、结构、材料或者特点可以在任一个或多个实施例或示例中以合适的方式结合。此外,在不相互矛盾的情况下,本领域的技术人员可以将本说明书中描述的不同实施例或示例以及不同实施例或示例的特征进行结合和组合。In the description of this specification, descriptions referring to the terms "one embodiment", "some embodiments", "example", "specific examples", or "some examples" mean that specific features described in connection with the embodiment or example , structure, material or characteristic is included in at least one embodiment or example of the present invention. In this specification, the schematic representations of the above terms are not necessarily directed to the same embodiment or example. Furthermore, the described specific features, structures, materials or characteristics may be combined in any suitable manner in any one or more embodiments or examples. In addition, those skilled in the art can combine and combine different embodiments or examples and features of different embodiments or examples described in this specification without conflicting with each other.
尽管上面已经示出和描述了本发明的实施例,可以理解的是,上述实施例是示例性的,不能理解为对本发明的限制,本领域的普通技术人员在本发明的范围内可以对上述实施例进行变化、修改、替换和变型。 Although the embodiments of the present invention have been shown and described above, it can be understood that the above embodiments are exemplary and should not be construed as limiting the present invention, those skilled in the art can make the above-mentioned The embodiments are subject to changes, modifications, substitutions and variations.

Claims (24)

  1. 一种5’→3’核酸外切酶在基因编辑系统中的用途,其特征在于,所述5’→3’核酸外切酶与所述基因编辑系统中的位点特异性核酸酶为非融合状态。A use of a 5'→3' exonuclease in a gene editing system, characterized in that the 5'→3' exonuclease is non-specific to the site-specific nuclease in the gene editing system fusion state.
  2. 根据权利要求1所述的用途,其特征在于,所述基因编辑系统中采用的细胞为动物细胞。The use according to claim 1, characterized in that the cells used in the gene editing system are animal cells.
  3. 根据权利要求2所述的用途,其特征在于,所述基因编辑系统中采用的细胞为哺乳动物细胞。The use according to claim 2, characterized in that the cells used in the gene editing system are mammalian cells.
  4. 根据权利要求1所述的用途,其特征在于,所述述5’→3’核酸外切酶为T7核酸外切酶。purposes according to claim 1, is characterized in that, described 5 ' → 3 ' exonuclease is T7 exonuclease.
  5. 根据权利要求4所述的用途,其特征在于,所述T7核酸外切酶具有SEQ ID NO:1所示的氨基酸序列或者与其具有至少80%同源性的氨基酸序列。The use according to claim 4, wherein the T7 exonuclease has the amino acid sequence shown in SEQ ID NO: 1 or an amino acid sequence having at least 80% homology therewith.
  6. 一种基因编辑系统,其特征在于,包含:A gene editing system, characterized in that it comprises:
    位点特异性核酸酶、5’→3’核酸外切酶和供体DNA;Site-specific nucleases, 5'→3' exonucleases, and donor DNA;
    其中,所述5’→3’核酸外切酶与所述位点特异性核酸酶为非融合状态。Wherein, the 5'→3' exonuclease is in a non-fused state with the site-specific nuclease.
  7. 根据权利要求6所述的基因编辑系统,其特征在于,所述5’→3’核酸外切酶为T7核酸外切酶。The gene editing system according to claim 6, wherein the 5'→3' exonuclease is T7 exonuclease.
  8. 根据权利要求7所述的基因编辑系统,其特征在于,所述T7核酸外切酶具有SEQ ID NO:1所示的氨基酸序列或者与其具有至少80%同源性的氨基酸序列。The gene editing system according to claim 7, wherein the T7 exonuclease has the amino acid sequence shown in SEQ ID NO: 1 or an amino acid sequence having at least 80% homology therewith.
  9. 根据权利要求6~8任一项所述的基因编辑系统,其特征在于,所述位点特异性核酸酶选自成簇规律间隔短回文重复、转录激活样效应因子核酸酶、锌指核酸酶、归巢内切酶、限制性核酸内切酶中的至少之一。The gene editing system according to any one of claims 6 to 8, wherein the site-specific nuclease is selected from the group consisting of clustered regularly interspaced short palindromic repeats, transcriptional activation-like effector nucleases, and zinc finger nucleic acids At least one of enzymes, homing endonucleases, and restriction endonucleases.
  10. 根据权利要求6~8任一项所述的基因编辑系统,其特征在于,所述位点特异性核酸酶的添加量为2~50ng。The gene editing system according to any one of claims 6-8, wherein the added amount of the site-specific nuclease is 2-50 ng.
  11. 根据权利要求6~8任一项所述的基因编辑系统,其特征在于,所述5’→3’核酸外切酶的添加量为40~120ng。The gene editing system according to any one of claims 6-8, wherein the added amount of the 5'→3' exonuclease is 40-120ng.
  12. 根据权利要求6~8任一项所述的基因编辑系统,其特征在于,所述供体DNA的添加量为0.2~100ng。The gene editing system according to any one of claims 6-8, characterized in that the added amount of the donor DNA is 0.2-100 ng.
  13. 根据权利要求9所述的基因编辑系统,其特征在于,所述基因编辑系统进一步包含:gRNA。The gene editing system according to claim 9, wherein the gene editing system further comprises: gRNA.
  14. 根据权利要求13所述的基因编辑系统,其特征在于,所述gRNA包括选自sgRNA、pegRNA和crRNA/tracrRNA中的至少之一。The gene editing system according to claim 13, wherein the gRNA comprises at least one selected from sgRNA, pegRNA and crRNA/tracrRNA.
  15. 根据权利要求14所述的基因编辑系统,其特征在于,所述基因编辑系统进一步包含:招募系统。The gene editing system according to claim 14, wherein the gene editing system further comprises: a recruitment system.
  16. 根据权利要求15所述的基因编辑系统,其特征在于,若所述招募系统为MS2/MCP系统,所述gRNA为gRNA-MS2或pegRNA-MS2,所述5’→3’核酸外切酶为MCP-外切酶融合蛋白。The gene editing system according to claim 15, wherein if the recruitment system is the MS2/MCP system, the gRNA is gRNA-MS2 or pegRNA-MS2, and the 5'→3' exonuclease is MCP-exonuclease fusion protein.
  17. 根据权利要求6~8任一项所述的基因编辑系统,其特征在于,所述供体DNA为采用硫代磷酸二酯键修饰的线性双链DNA和/或HMEJ形式的环状双链DNA。The gene editing system according to any one of claims 6 to 8, wherein the donor DNA is a linear double-stranded DNA modified with a phosphorothioate bond and/or a circular double-stranded DNA in the form of HMEJ .
  18. 根据权利要求17所述的基因编辑系统,其特征在于,所述供体DNA上的所述硫代磷酸二酯键的修饰个数大于2。The gene editing system according to claim 17, wherein the number of modifications of the phosphorothioate bonds on the donor DNA is greater than 2.
  19. 根据权利要求18所述的基因编辑系统,其特征在于,所述供体DNA上的所述硫代磷酸二酯键的修饰个数为4-10。The gene editing system according to claim 18, wherein the number of modifications of the phosphorothioate bonds on the donor DNA is 4-10.
  20. 根据权利要求6~8任一项所述的基因编辑系统,其特征在于,所述基因编辑系统的技术路线包括寡核苷酸编辑模板介导的同源重组、单碱基编辑、引导编辑和双链长链核酸编辑模板介导的同源重组中的一种。The gene editing system according to any one of claims 6 to 8, wherein the technical route of the gene editing system includes homologous recombination mediated by oligonucleotide editing templates, single base editing, guided editing and A type of homologous recombination mediated by double-stranded long-chain nucleic acid editing templates.
  21. 一种对细胞进行基因编辑的方法,其特征在于,包括:A method for genetically editing cells, comprising:
    将权利要求6-20任一项所述的基因编辑系统引入细胞。Introducing the gene editing system according to any one of claims 6-20 into cells.
  22. 根据权利要求21所述的方法,其特征在于,所述细胞为动物细胞。The method of claim 21, wherein the cells are animal cells.
  23. 根据权利要求22所述的方法,其特征在于,所述细胞为哺乳动物细胞。The method of claim 22, wherein the cells are mammalian cells.
  24. 根据权利要求21~23任一项所述的方法,其特征在于,所述细胞为终末分化原代细胞。 The method according to any one of claims 21-23, wherein the cells are terminally differentiated primary cells.
PCT/CN2023/079632 2022-03-03 2023-03-03 Use of 5'→3' exonuclease in gene editing system, and gene editing system and gene editing method WO2023165613A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210204717 2022-03-03
CN202210204717.2 2022-03-03

Publications (1)

Publication Number Publication Date
WO2023165613A1 true WO2023165613A1 (en) 2023-09-07

Family

ID=86823639

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/079632 WO2023165613A1 (en) 2022-03-03 2023-03-03 Use of 5'→3' exonuclease in gene editing system, and gene editing system and gene editing method

Country Status (2)

Country Link
CN (1) CN116286741A (en)
WO (1) WO2023165613A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020236645A1 (en) * 2019-05-17 2020-11-26 Beth Israel Deaconess Medical Center, Inc. Compositions and methods for homology directed repair
US20200407754A1 (en) * 2019-06-25 2020-12-31 Inari Agriculture, Inc. Homology dependent repair genome editing
CN113025597A (en) * 2019-12-24 2021-06-25 中国科学院微生物研究所 Improved genome editing system
CN113481184A (en) * 2021-08-06 2021-10-08 北京大学 Fusion proteins and methods of use thereof
US20210403922A1 (en) * 2019-01-07 2021-12-30 Crisp-Hr Therapeutics, Inc. Non-toxic cas9 enzyme and application thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210403922A1 (en) * 2019-01-07 2021-12-30 Crisp-Hr Therapeutics, Inc. Non-toxic cas9 enzyme and application thereof
WO2020236645A1 (en) * 2019-05-17 2020-11-26 Beth Israel Deaconess Medical Center, Inc. Compositions and methods for homology directed repair
US20200407754A1 (en) * 2019-06-25 2020-12-31 Inari Agriculture, Inc. Homology dependent repair genome editing
CN113025597A (en) * 2019-12-24 2021-06-25 中国科学院微生物研究所 Improved genome editing system
CN113481184A (en) * 2021-08-06 2021-10-08 北京大学 Fusion proteins and methods of use thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
QIANWEI ZHANG; KANGQUAN YIN; GUANWEN LIU; SHENGNAN LI; MENGOU LI; JIN-LONG QIU: "Fusing T5 Exonuclease with Cas9 and Cas12a Increases the Frequency and Size of Deletion at Target Sites", SCIENCE CHINA LIFE SCIENCES, ZHONGGUO KEXUE ZAZHISHE, CHINA, vol. 63, no. 12, 6 May 2020 (2020-05-06), China , pages 1918 - 1927, XP009548348, ISSN: 1674-7305 *

Also Published As

Publication number Publication date
CN116286741A (en) 2023-06-23

Similar Documents

Publication Publication Date Title
US11555181B2 (en) Engineered cascade components and cascade complexes
US10501728B2 (en) Engineered nucleic-acid targeting nucleic acids
EP3523430B1 (en) Engineered nucleic acid-targeting nucleic acids
JP7093728B2 (en) Highly specific genome editing using chemically modified guide RNA
WO2022253185A1 (en) Cas12 protein, gene editing system containing cas12 protein, and application
JP6707133B2 (en) Engineered nucleic acid targeted nucleic acid
US20200354729A1 (en) Fusion proteins for improved precision in base editing
CN110959040A (en) Base editor with improved accuracy and specificity
WO2017161068A1 (en) Mutant cas proteins
Guo et al. Multiplexed genome regulation in vivo with hyper-efficient Cas12a
JP2023517041A (en) Class II type V CRISPR system
CN114634930A (en) Compositions and methods for improving genome engineering specificity using RNA-guided endonucleases
US20230212612A1 (en) Genome editing system and method
KR20230076820A (en) Synthetic Miniature CRISPR-CAS (CASMINI) System for Eukaryotic Genome Engineering
US20230340481A1 (en) Systems and methods for transposing cargo nucleotide sequences
CN113583999A (en) Cas9 protein, gene editing system containing Cas9 protein and application
WO2023165613A1 (en) Use of 5'→3' exonuclease in gene editing system, and gene editing system and gene editing method
CN113039276A (en) Nuclease-mediated modification of nucleic acids
JP2024501892A (en) Novel nucleic acid-guided nuclease
CN114901303A (en) Modified endonucleases and related methods
WO2020047531A1 (en) Scalable tagging of endogenous genes by homology-independent intron targeting
WO2023232109A1 (en) Novel crispr gene editing system
WO2024121790A2 (en) Cas12 protein, crispr-cas system and uses thereof
CN118165956A (en) CRISPR/Cas9 gene editing system based on Tsp2Cas9 protein and related application thereof
CN116615547A (en) System and method for transposing nucleotide sequences of cargo

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 23763011

Country of ref document: EP

Kind code of ref document: A1