WO2024078626A1 - Method for improving crispr/cas9-mediated genome knock-in editing efficiency - Google Patents

Method for improving crispr/cas9-mediated genome knock-in editing efficiency Download PDF

Info

Publication number
WO2024078626A1
WO2024078626A1 PCT/CN2023/124585 CN2023124585W WO2024078626A1 WO 2024078626 A1 WO2024078626 A1 WO 2024078626A1 CN 2023124585 W CN2023124585 W CN 2023124585W WO 2024078626 A1 WO2024078626 A1 WO 2024078626A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
small molecule
formula
compound
cell
Prior art date
Application number
PCT/CN2023/124585
Other languages
French (fr)
Chinese (zh)
Inventor
郭宜君
黄菲
叶露萌
李竑
Original Assignee
南京金斯瑞生物科技有限公司
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 南京金斯瑞生物科技有限公司 filed Critical 南京金斯瑞生物科技有限公司
Publication of WO2024078626A1 publication Critical patent/WO2024078626A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • 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
    • 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

Definitions

  • the present invention relates to a method for improving the efficiency of CRISPR/Cas9-mediated genome knock-in editing, in particular, a method for improving the efficiency of CRISPR/Cas9-mediated genome knock-in editing using a combination of small molecule protease inhibitors.
  • the present invention also relates to a kit including the above-mentioned small molecule protease inhibitors.
  • the CRISPR/Cas9 system is an RNA-guided genome editing tool that provides researchers with a simple method to quickly modify the genomes of various organisms. With the widespread use of the CRISPR/Cas9 system and the fact that precise gene editing is being or will soon be used clinically for a variety of diseases, the call to improve the efficiency of precise genome editing mediated by this system is growing.
  • Cas9 under the guidance of guide RNA (gRNA), locates to the target site of genomic DNA and cuts the targeted gene sequence, forming a DNA double-strand break (DSB), and then repairs the DSB through the intracellular repair mechanism.
  • gRNA guide RNA
  • NHEJ non-homologous end joining
  • HDR homologous recombination repair
  • the results of gene editing mainly depend on the choice between these two repair pathways.
  • the NHEJ pathway repairs DSB by directly connecting the ends. This repair does not require large fragments of homologous sequence recognition, but only requires 1-5bp base pairing for repair. It is an error-prone repair with poor accuracy.
  • the HDR pathway can repair DSB by inserting a piece of DNA into the editing site through homologous recombination in the presence of a highly homologous DNA template, thereby achieving the purpose of accurately inserting a DNA sequence into a specific genomic site.
  • the repair mechanism of mammalian cells will give priority to NHEJ rather than HDR: NHEJ is active throughout the cell cycle, while HDR is limited to the S/G2 phase; NHEJ is faster than HDR; and NHEJ also inhibits the HDR repair pathway. Therefore, there are more and more studies on whether and how to improve the efficiency of CRISPR/Cas9-mediated precise genome insertion editing by regulating cell repair pathways.
  • the present invention provides a method for enhancing the efficiency of gene knock-in mediated by a CRISPR gene editing system in a cell, comprising contacting the cell with a DNA-dependent protein kinase (DNA-PK) inhibitor and a cell division cycle protein 7 (Cdc7) inhibitor during the gene knock-in operation on the cell.
  • DNA-PK DNA-dependent protein kinase
  • Cdc7 cell division cycle protein 7
  • the CRISPR gene editing system includes a Cas9 protein.
  • the CRISPR gene editing system further comprises a guide RNA (gRNA) and a donor template for homologous recombination repair (HDR).
  • gRNA guide RNA
  • HDR homologous recombination repair
  • the donor template is a DNA molecule in single-stranded or double-stranded form.
  • the DNA-PK inhibitor is selected from a compound of formula (I), a compound of formula (II), and a combination thereof, wherein X in the compound of formula (I) is hydrogen or deuterium:
  • the compound of formula (I) is:
  • the Cdc7 inhibitor is a compound of formula (V) or its hydrochloride salt:
  • the compound of formula (I) is contacted with the cell at a concentration of not less than 1 ⁇ M; the compound of formula (II) is contacted with the cell at a concentration of not less than 0.5 ⁇ M; or the compound of formula (V) is contacted with the cell at a concentration of not less than 10 ⁇ M.
  • the compound of Formula (I) is contacted with the cell at a concentration of 1-30 ⁇ M, and the compound of Formula (V) is contacted with the cell at a concentration of 10-50 ⁇ M.
  • the compound of Formula (II) is contacted with the cell at a concentration of 0.5-5 ⁇ M, and the compound of Formula (V) is contacted with the cell at a concentration of 10-50 ⁇ M.
  • the compound of Formula (I) is contacted with the cell at a concentration of 5-20 ⁇ M, and the compound of Formula (V) is contacted with the cell at a concentration of 20-40 ⁇ M.
  • the compound of Formula (II) is contacted with the cell at a concentration of 2-4 ⁇ M, and the compound of Formula (V) is contacted with the cell at a concentration of 20-40 ⁇ M.
  • the compound of Formula (I) is contacted with the cell at a concentration of 5 ⁇ M, and the compound of Formula (V) is contacted with the cell at a concentration of 30 ⁇ M.
  • the compound of Formula (II) is contacted with the cell at a concentration of 2 ⁇ M, and the compound of Formula (V) is contacted with the cell at a concentration of 30 ⁇ M.
  • the cell is a HEK293T, Jurkat, SJCRH30, H1975, or A549 cell.
  • the cell is a T cell.
  • the cell is contacted with the compound of formula (I), the compound of formula (II), and the compound of formula (V) during a gene knock-in procedure.
  • the gene knock-in operation is performed by transfection, and the cell is contacted with the compound of formula (V) prior to the transfection.
  • the gene knock-in operation is performed by transfection, and the compound of formula (I) and/or the compound of formula (II) is contacted with the cells after the transfection step, and the compound of formula (V) is contacted with the cells both before and after the transfection step.
  • the present invention provides a kit for enhancing the gene knock-in efficiency mediated by the CRISPR gene editing system in cells, comprising a DNA-dependent protein kinase (DNA-PK) inhibitor and a cell division cycle protein 7 (Cdc7) inhibitor.
  • DNA-PK DNA-dependent protein kinase
  • Cdc7 cell division cycle protein 7
  • the kit further comprises a Cas9 protein or a nucleic acid molecule encoding the Cas9 protein.
  • the kit further comprises a guide RNA (gRNA) and/or a donor template for performing homologous recombination repair (HDR).
  • gRNA guide RNA
  • HDR homologous recombination repair
  • the donor template is a DNA molecule in single-stranded or double-stranded form.
  • the DNA-PK inhibitor is selected from a compound of formula (I), a compound of formula (II), and a combination thereof, wherein X in the compound of formula (I) is hydrogen or deuterium:
  • the compound of formula (I) is:
  • the Cdc7 inhibitor is a compound of formula (V) or its hydrochloride salt:
  • the cell is a HEK293T, Jurkat, SJCRH30, H1975, or A549 cell.
  • the cell is a T cell.
  • the method provided in this article uses a DNA-dependent protein kinase (DNA-PK) inhibitor and a cell division cycle protein 7 (Cdc7) inhibitor in combination, so that the gene knock-in efficiency of the CRISPR gene editing system can be significantly improved in different cell lines, different insertion sites, and different lengths of donor templates, and is superior to some commercial small molecule enhancers.
  • DNA-PK DNA-dependent protein kinase
  • Cdc7 cell division cycle protein 7
  • Figure 1 shows the cell viability (A) and knock-in efficiency (B) of small molecule 1 at different concentrations.
  • FIG2 shows the cell viability (A) and knock-in efficiency (B) of small molecule 2 at different concentrations.
  • FIG3 shows the cell viability (A) and knock-in efficiency (B) of small molecule 3 at different concentrations.
  • FIG4 shows the cell viability (A) and knock-in efficiency (B) of small molecule 4 at different concentrations.
  • FIG5 shows the cell viability (A) and knock-in efficiency (B) of small molecule 5 at different concentrations.
  • Figure 6 shows different incubation methods for small molecule 5.
  • Method 1 cells were cultured in a medium containing small molecule 5 for 24 hours before electroporation, and immediately after electroporation, the cells were placed in a medium without small molecule 5;
  • Method 2 cells were placed in a medium containing small molecule 5 for 24 hours immediately after electroporation;
  • Method 3 cells were placed in a medium containing small molecule 5 for 24 hours before and after electroporation. Small molecule 5 was not contained in the electroporation buffer.
  • FIG7 shows the cell viability (A) and knock-in efficiency (B) of small molecule 5 under different incubation modes.
  • Figure 8 shows the effective concentrations of small molecules and different combinations, where "+" indicates addition and “-" indicates no addition.
  • FIG. 9 shows the cell viability at 4 days under the action of different molecular combinations.
  • FIG. 10 shows the knock-in efficiency of cells at 7 days under the action of different molecular combinations.
  • FIG11 shows the design of the action concentration range of different small molecules and their combinations, where “+” indicates addition and “-” indicates no addition.
  • FIG. 12 shows the cell knock-in efficiency at 7 days under the concentration range of different small molecules and their combinations.
  • FIG. 13 shows the knock-in efficiency of cells at 7 days using two types of donor templates, dsDNA and ssDNA, under the action of different small molecule combinations.
  • Figure 14 shows the validation of cell viability at day 3 using dsDNA and plasmid donor templates in a T cell system.
  • FIG. 15 shows validation of cell knock-in efficiency at 7 days using dsDNA and plasmid donor templates in a T cell system.
  • FIG16 shows the restriction enzyme cleavage patterns of different cell lines at different sites 3 days after electroporation.
  • FIG. 17 shows a comparison of cell knock-in efficiency at 3 days after electroporation of different sites in different cell lines.
  • Figure 18 shows the comparison of cell knock-in efficiency at 7 days under the action of different small molecule combinations and IDT HDR enhancer.
  • Figure 19 shows the comparison of cell knock-in efficiency at 3 days under the action of small molecule combination and IDT HDR enhancer.
  • any value within the range of ⁇ 10% of the listed value is referred to at the same time. It will be understood by those skilled in the art that due to reasons such as instrument detection accuracy and errors introduced during operation, the values of the test results, operation time, etc. given may vary within a small range. For example, when it is mentioned that cells are cultured for 24 hours, it should be understood that culturing cells for 24 hours ⁇ 2.4 hours is also feasible. For another example, when it is mentioned that the detected knock-in efficiency is 20%, the actual knock-in efficiency may be 20% ⁇ 2%.
  • CRISPR Clustered Regularly Interspaced Short Palindromic Repeats
  • the CRISPR gene editing system used in this technology includes Cas nuclease and guide RNA (single-guide RNA, sgRNA or gRNA), Depending on the situation, double-stranded DNA (dsDNA) or single-stranded DNA (ssDNA) as a repair template may also be included.
  • dsDNA double-stranded DNA
  • ssDNA single-stranded DNA
  • ssDNA single-stranded DNA
  • a portion of the sequence of sgRNA can bind to the Cas nuclease, and another portion of the sequence (crRNA) can be complementary to a portion of the sequence of the target gene.
  • the Cas nuclease can form a single-stranded or double-stranded incision at a specific site of the target gene.
  • Cells usually repair DNA broken chains in two ways, namely homologous recombination repair mechanism (homology-directed repair, HDR) and non-homologous end joining repair mechanism (non-homologous end joining, NHEJ).
  • HDR homologous recombination repair mechanism
  • NHEJ non-homologous end joining
  • CRISPR gene editing technology can be used to knock out genes in cells. In this case, it is usually only necessary to consider destroying the normal coding function of the gene, such as causing frameshift mutations or gene fragment deletions, so that products with normal functions (such as proteins) cannot be produced.
  • CRISPR gene editing technology can be used to knock in cells, that is, to introduce a target gene into the cell genome so that the cell can produce a product with the target gene.
  • the donor template sequence may include the target gene sequence and homologous arm sequences on both sides of the target gene.
  • the purpose of introducing the target gene into the genome is achieved by homologous recombination of the homologous arm sequence and the gap site sequence generated by the Cas nuclease on the genome.
  • the cutting efficiency of Cas nuclease such as Cas9
  • the probability of repairing the double-stranded DNA of the cell genome by homologous recombination and the probability of recombination with the donor template will affect the final gene knock-in efficiency.
  • Cas9 protein is the key to the nuclease activity of the CRISPR/Cas9 system. It contains two domains with cutting activity: the HNH domain and the RuvC domain. HNH and RuvC can cut the two strands of DNA to form double-strand breaks.
  • the Cas9 protein can be a recombinant Cas9 protein SpCas9 derived from Streptococcus pyogenes.
  • the Cas9 protein can also be a variant of the Cas9 protein, such as the Cas9 protein variants eSpCas9 (1.0), eSpCas9 (1.1) and SpCas9-HF1 with significantly improved specificity obtained by directed modification of the Cas9 protein.
  • target sequence refers to a nucleotide fragment in the cell genome that is complementary to a portion of the sgRNA sequence (crRNA, about 20 bases).
  • proteins such as Cas9 can introduce nucleotide sequence changes in the genome at a relatively certain position to achieve the effect of gene knockout or gene knockin.
  • sgRNA targeting a specified sequence means that the target sequence of the sgRNA is the specified sequence.
  • donor template in this article refers to the exogenous DNA template used by the homologous recombination repair (HDR) pathway to mediate gene replacement or insertion after the double-strand break of the cell's DNA.
  • HDR homologous recombination repair
  • the cleavage site of the Cas9 protein is determined by the sgRNA sequence, and the sequences on both sides of the cleavage site are the homologous arm sequences in the donor template.
  • the target gene sequence that needs to be accurately inserted in the middle of the homologous arm is the sequence of the donor template.
  • the "donor template” can be double-stranded DNA (dsDNA) or single-stranded DNA (ssDNA), or it can be a single-stranded oligonucleotide (ssODN), or even a circular DNA (such as a plasmid).
  • the inserted target gene can be any gene sequence that you want to insert into the cell.
  • the target gene sequence may include, for example, genes (nucleotide sequences) encoding defective or missing proteins in the recipient individual or target cell; genes encoding proteins with desired biological or therapeutic effects (for example, antibacterial, antiviral or anti-tumor/anti-cancer functions); genes encoding inhibition or reduction
  • the donor template can be a DNA sequence of any length, such as from a few bp to several thousand bp.
  • gene knock-in operation refers to the purposeful introduction of a target gene into a cell by means of technical means such as CRISPR gene editing technology.
  • the target gene can be randomly integrated into the cell genome.
  • the target gene can be integrated into the cell genome at a relatively determined site (for example, by using the above-mentioned sgRNA with a specific sequence).
  • the target gene can then be expressed in the cell to generate products such as target proteins, which can be used to study the function of the target gene or for preparing protein products.
  • the gene knock-in operation process may, for example, include preparing cells to be knocked into genes, introducing Cas nucleases, gRNAs and donor templates into cells (for example, by electroporation), continuing to culture cells, and optionally screening positive clones and other steps.
  • knock-in efficiency or knock-in editing efficiency refers herein to the ratio of cells including a gene to be knocked-in (target gene) in its genome in a cell population after a gene knock-in operation relative to the total number of cells in the cell population.
  • the detection of gene knock-in efficiency can be carried out in a variety of ways.
  • the gene knock-in efficiency can be determined by detecting an expression product expressing the target gene, such as a fluorescent protein.
  • Improving knock-in efficiency refers to improving the gene knock-in operation process so as to obtain a higher knock-in efficiency.
  • a small molecule enzyme inhibitor is added during the gene knock-in operation process.
  • DNA-PK DNA-dependent protein kinase
  • Cdc7 cell division cycle protein 7
  • DNA-dependent protein kinase is a serine/threonine kinase and a member of the phosphatidylinositol 3-kinase-related kinase (PI3K) family. It is mainly located in the cell nucleus and can repair DNA double-strand breaks by non-homologous end joining. It plays an important role in maintaining genome stability and V(D)J recombination to produce antibody diversity. Its activity is regulated by DNA double-strand breaks.
  • DNA-PK and its components are involved in a variety of other physiological processes, including the regulation of chromatin structure, telomere maintenance, transcriptional regulation, and response to replication stress. Common DNA-PK inhibitors include AZD-7648, KU-57788Torin 2, NU5455, VX-984, Samotolisib, etc.
  • Cyclin 7 is a serine/threonine kinase that plays an important role in initiating DNA replication. From the late G1 phase to the S phase, Cdc7 forms a complex with Dbf4 (also known as ASK) and phosphorylates Cdc7 substrates to control the transition from G1 phase to S phase.
  • Dbf4 also known as ASK
  • Common Cdc7 inhibitors include XL413, XL413 hydrochloride, CAY10572 (PHA-767491), Cdc7-IN-1, Cdc7-IN-5, Cdc7-IN-17, LY3177833, etc.
  • cell transfection is a technique for introducing exogenous genes into eukaryotic cells.
  • Cell transfection pathways can be roughly divided into three categories: chemical mediation, biological mediation, and physical mediation.
  • chemical-mediated methods such as the classic calcium phosphate co-precipitation method, liposome transfection method, and various cationic substance-mediated techniques.
  • Biological-mediated methods include the more primitive protoplast transfection and the more common various virus-mediated transfection techniques.
  • Physical-mediated methods mainly include electroporation, microinjection, and gene guns.
  • electroporation refers to electroporation, which uses high pulse voltage to destroy cells.
  • the cell membrane potential allows DNA to be introduced through the small holes formed on the membrane for stable/transient transfection. This method is applicable to all types of cells, but the cell lethality is high and the electroporation experimental conditions need to be optimized according to different cell types.
  • This article provides a better method for improving the efficiency of CRISPR/Cas9-mediated genome knock-in editing.
  • the specific implementation of the present invention can be divided into the following steps:
  • the small molecule 1 or small molecule 2 and small molecule 5 with the determined effective concentration are used together to further improve the knock-in efficiency.
  • the incubation conditions are the same as those in the above step (2).
  • the cells when performing a gene knock-in operation, are contacted with small molecule 1 and small molecule 5.
  • the cells after transfection (such as electroporation transfection, referred to as electrotransfection), the cells are cultured in a culture medium containing small molecule 1 and small molecule 5 for a period of time; or before transfection, the cells are cultured in a culture medium containing small molecule 5 for a period of time, and after transfection (such as electrotransfection), the cells are cultured in a culture medium containing small molecule 1 and small molecule 5 for a period of time; or before transfection, the cells are cultured in a culture medium containing small molecule 1 and small molecule 5 for a period of time, and after transfection (such as electrotransfection), the cells are cultured in a culture medium containing small molecule 1 and small molecule 5 for a period of time.
  • the cells are cultured in a culture medium containing small molecule 5 for a period of time, and after transfection (such as electrotransfection), the cells are cultured in a culture medium containing small molecule 1 and small molecule 5 for a period of time.
  • the cell when performing a gene knock-in operation, the cell is contacted with small molecule 2 and small molecule 5.
  • the cell is cultured in a culture medium containing small molecule 2 and small molecule 5 for a period of time; or before transfection, the cell is cultured in a culture medium containing small molecule 5 for a period of time, and after transfection (such as electroporation), the cell is cultured in a culture medium containing small molecule 2 and small molecule 5 for a period of time; or before transfection, the cell is cultured in a culture medium containing small molecule 2 and small molecule 5 for a period of time, and after transfection (such as electroporation), the cell is cultured in a culture medium containing small molecule 2 and small molecule 5 for a period of time.
  • the cell is cultured in a culture medium containing small molecule 5 for a period of time before transfection, and the cell is cultured in a culture medium containing small molecule 2 and small molecule 5 for a period of time after transfection (such as electroporation).
  • the cell when performing a gene knock-in operation, the cell is contacted with small molecule 1, small molecule 2, and small molecule 5.
  • small molecule 1, small molecule 2, and small molecule 5 for example, after transfection (such as electroporation), the cell is cultured in a culture medium containing small molecule 1, small molecule 2, and small molecule 5 for a period of time; or before transfection, the cell is cultured in a culture medium containing small molecule 5 for a period of time, and after transfection (such as electroporation), the cell is cultured in a culture medium containing small molecule 1, small molecule 2, and small molecule 5 for a period of time; or before transfection, the cell is cultured in a culture medium containing small molecule 1, small molecule 2, and small molecule 5 for a period of time, and after transfection (such as electroporation), the cell is cultured in a culture medium containing small molecule 1, small molecule 2, and small molecule 5 for a period of time.
  • the cell is cultured in a culture medium containing small molecule 5 for a period of time, and after transfection (such as electroporation), the cell is cultured in a culture medium containing small molecule 1, small molecule 2, and small molecule 5 for a period of time.
  • “Cultivating for a period of time” used herein refers to cultivating a few minutes, a few hours, more than ten hours, or longer time, for example, 30min, 60min, 1h, 2h, 5h, 10h, 20h, 24h, 48h, 72h, and any duration between the listed durations.
  • “cultivating for a period of time” refers to cultivating 24-72h, for example 24h.
  • the medium when the medium contains small molecule 1, its concentration is not less than 1 ⁇ M, for example, so that the concentration of small molecule 1 in the medium is 1-30 ⁇ M, such as 5-20 ⁇ M. Preferably, the concentration of small molecule 1 in the medium is 5 ⁇ M.
  • the medium when the medium contains small molecule 2, its concentration is not less than 0.5 ⁇ M, for example, so that the concentration of small molecule 2 in the medium is 0.5-5 ⁇ M, such as 2-4 ⁇ M.
  • the concentration of small molecule 2 in the medium is 2 ⁇ M.
  • the medium when the medium contains small molecule 5, its concentration is not less than 10 ⁇ M, for example, so that the concentration of small molecule 5 in the medium is 10-50 ⁇ M, such as 20-40 ⁇ M. Preferably, the concentration of small molecule 5 in the medium is 30 ⁇ M.
  • kits for improving the efficiency of CRISPR/Cas9-mediated genome knock-in editing including small molecule 1, small molecule 2, small molecule 5, and any combination thereof.
  • the kit includes small molecule 1 and small molecule 5.
  • the kit includes small molecule 2 and small molecule 5.
  • the kit includes small molecule 1, small molecule 2, and small molecule 5.
  • the kit also optionally includes Cas9 protein or its encoding nucleic acid molecule.
  • the kit also optionally includes guide RNA (gRNA) and/or a donor template for homologous recombination repair (HDR).
  • gRNA guide RNA
  • HDR homologous recombination repair
  • DNA-PK DNA-dependent protein kinase
  • Cdc7 cell division cycle protein 7
  • Embodiment 1 is a diagrammatic representation of Embodiment 1:
  • RNP formation Add Cas9 protein (GenScript, Z03469), RAB11A-sgRNA (SEQ ID NO: 1) (GenScript, EasyEdit), RAB11A-dsDNA template (SEQ ID NO: 2) (GenScript) and buffer R (Thermo Fisher) as shown in Table 3 into a new, RNase-free centrifuge tube and incubate at room temperature for 5-15 min to form RNP.
  • Electroporation system Add 6 ⁇ l of cells resuspended in buffer R (HEK293T: 0.2M; Jurkat: 0.5M) to the above centrifuge tube containing RNPs, and mix by carefully pipetting with a pipette.
  • Electroporation Turn on the Neon electroporator (Thermo Fisher) and set the electroporation parameters (HEK293T cells: Voltage: 1200 V, Width: 10 ms, Pulse: 3 pulses; Jurkat cells: Voltage: 1600 V, Width: 10 ms, Pulse: 3 pulses).
  • the cells were cultured in a medium containing small molecules; after 72 hours, the cells were switched from a medium containing small molecules to a medium without small molecules.
  • small molecules 1, 2, and 5 significantly improved the gene knock-in efficiency, and their effective concentrations were: small molecule 1: ⁇ 1 ⁇ M; small molecule 2: ⁇ 0.5 ⁇ M; small molecule 5: ⁇ 10 ⁇ M. Moreover, these three small molecules had no significant effect on cell viability within the effective concentration.
  • Embodiment 2 is a diagrammatic representation of Embodiment 1:
  • GFP green fluorescent protein
  • the cells were passaged every 2-3 days, and the cells required for electroporation were plated 1 day before electroporation.
  • two cells were prepared, one of which was cultured without small molecule 5, and the other was cultured with small molecule 5 added to a final concentration of 30 ⁇ M 24 hours before electroporation.
  • small molecule 5 with a final concentration of 30 ⁇ M was added to the culture medium of some wells of the well plate, and then the well plate was placed in a 37° C. incubator for preheating.
  • RNP formation eSpCas9 protein (GenScript, Z03470-300), RAB11A-sgRNA (SEQ ID NO: 1) (GenScript, EasyEdit), RAB11A-dsDNA template (SEQ ID NO: 2) (GenScript) and buffer R (Thermo Fisher) were added to a new, RNase-free centrifuge tube as shown in Table 3 (Jurkat cells) and incubated at room temperature for 5-15 min to form RNP.
  • SEQ ID NO: 1 GenScript, EasyEdit
  • SEQ ID NO: 2 GenScript
  • buffer R Thermo Fisher
  • Electroporation system Add 6 ⁇ l of 0.5 M Jurkat cells resuspended in buffer R to the above centrifuge tube containing RNP (the sample group without small molecule 5 uses cells that have not been pre-treated with small molecule 5 in step 2), and the sample group with small molecule 5 uses cells that have been pre-treated with 30 ⁇ M small molecule 5 in step 2)), and mix carefully by pipetting with a pipette tip.
  • Electroporation Turn on the Neon electroporator (Thermo Fisher) and set the electroporation parameters: Voltage: 1700 V, Width: 20 ms, Pulse: 1 pulses.
  • the optimal incubation condition for small molecule 5 is incubation method 3: cells are cultured in the medium containing small molecule 5 for less than 24 h before electroporation; cells are cultured in the medium containing small molecule 5 after electroporation. The cells were cultured in a medium containing small molecule 5 for 24 h. After 24 h, the cells were switched from a medium containing small molecule 5 to a medium not containing small molecule 5.
  • Embodiment 3 is a diagrammatic representation of Embodiment 3
  • Small molecule 1, small molecule 2, small molecule 3, small molecule 4 after electroporation, the cells were cultured in a medium containing small molecules for 24 hours; after 24 hours, the cells were switched from the medium containing small molecules to a medium not containing small molecules.
  • Small molecule 5 Before electroporation, the cells are cultured in a medium containing small molecules for less than 24 hours; after electroporation, the cells are cultured in a medium containing small molecules for 24 hours. After 24 hours, the cells are switched from the medium containing small molecules to a medium without small molecules.
  • the cells were passaged every 2-3 days, and the cells required for electroporation were plated 1 day before electroporation.
  • cells were plated before electroporation two aliquots of cells were prepared, one of which was cultured without adding small molecules, and the other aliquot of cells was added with small molecules at a final concentration of 30 ⁇ M within 24 hours before electroporation.
  • RNP formation eSpCas9 protein (GenScript, Z03470-300), RAB11A-sgRNA (SEQ ID NO: 1) (GenScript, EasyEdit), RAB11A-dsDNA template (SEQ ID NO: 2) (GenScript) and buffer R (Thermo Fisher) were added to a new, RNase-free centrifuge tube as shown in Table 3 (Jurkat cells) and incubated at room temperature for 5-15 min to form RNP.
  • Electroporation system Add 6 ⁇ l of 0.5 M Jurkat cells resuspended in buffer R to the above centrifuge tube containing RNP (the sample group without small molecule 5 used cells not pre-treated with small molecule 5, and the sample group with small molecule 5 used cells pre-treated with 30 ⁇ M small molecule 5), and mix by carefully pipetting with a pipette tip.
  • Electroporation Turn on the Neon electroporator (Thermo Fisher) and set the electroporation parameters: Voltage: 1700 V, Width: 20 ms, Pulse: 1 pulses.
  • the knock-in efficiency was increased by 20.2% when small molecule 1 and small molecule 5 were used in combination; compared with the use of small molecule 2 alone, the knock-in efficiency was increased by 22.9% when small molecule 2 and small molecule 5 were used in combination.
  • the combined use of small molecule 1 and small molecule 5, as well as the combined use of small molecule 2 and small molecule 5, can maximize the knock-in efficiency while ensuring cell viability.
  • Embodiment 4 is a diagrammatic representation of Embodiment 4:
  • GFP green fluorescent protein
  • Small molecule 1 and small molecule 2 After electroporation, the cells were cultured in a medium containing small molecules for 24 hours; after 24 hours, the cells were switched from the medium containing small molecules to a medium not containing small molecules.
  • Small molecule 5 Before electroporation, the cells are cultured in a medium containing small molecules for less than 24 hours; after electroporation, the cells are cultured in a medium containing small molecules for 24 hours. After 24 hours, the cells are switched from the medium containing small molecules to a medium without small molecules.
  • the cells were passaged every 2-3 days, and the cells required for electroporation were plated one day before electroporation.
  • the cells required for electroporation were plated one day before electroporation.
  • four cells were prepared, one of which was cultured without small molecules, and the other three were added with small molecules at final concentrations of 20 ⁇ M, 30 ⁇ M, and 40 ⁇ M within 24h before electroporation.
  • RNP formation eSpCas9 protein (GenScript, Z03470-300), RAB11A-sgRNA (SEQ ID NO: 1) (GenScript, EasyEdit), RAB11A-dsDNA template (SEQ ID NO: 2) (GenScript) and buffer R (Thermo Fisher) were added to a new, RNase-free centrifuge tube as shown in Table 3 (Jurkat cells) and incubated at room temperature for 5-15 min to form RNP.
  • Electroporation system Add 6 ⁇ l of 0.5 M Jurkat cells resuspended in buffer R to the above centrifuge tube containing RNP (the sample group without small molecule 5 used cells not pre-treated with small molecule 5, the sample group with 20 ⁇ M small molecule 5 used cells pre-treated with 20 ⁇ M small molecule 5, the sample group with 30 ⁇ M small molecule 5 used cells pre-treated with 30 ⁇ M small molecule 5, and the sample group with 40 ⁇ M small molecule 5 used cells pre-treated with 40 ⁇ M small molecule 5), and mix by carefully pipetting with a pipette tip.
  • Electroporation Turn on the Neon electroporator (Thermo Fisher) and set the electroporation parameters: Voltage: 1600 V, Width: 10 ms, Pulse: 3 pulses.
  • Embodiment 5 is a diagrammatic representation of Embodiment 5:
  • GFP green fluorescent protein
  • Small molecule 1 and small molecule 2 After electroporation, the cells were cultured in a medium containing small molecules for 24 hours; after 24 hours, the cells were switched from the medium containing small molecules to a medium not containing small molecules.
  • Small molecule 5 Before electroporation, the cells are cultured in a medium containing small molecules for less than 24 hours; after electroporation, the cells are cultured in a medium containing small molecules for 24 hours. After 24 hours, the cells are switched from the medium containing small molecules to a medium without small molecules.
  • the cells were passaged every 2-3 days, and the cells required for electroporation were plated 1 day before electroporation.
  • cells were plated before electroporation two aliquots of cells were prepared, one of which was cultured without adding small molecules, and the other aliquot of cells was added with small molecules at a final concentration of 30 ⁇ M within 24 hours before electroporation.
  • RNP formation eSpCas9 protein (GenScript, Z03470-300), RAB11A-sgRNA (SEQ ID NO: 1) (GenScript, SafeEdit), RAB11A-dsDNA template or RAB11A-ssDNA template (SEQ ID NO: 2) (GenScript) and buffer R (Thermo Fisher) were added to a new, RNase-free centrifuge tube as shown in Table 4 and incubated at room temperature for 5-15 min to form RNP.
  • Electroporation system Add 6 ⁇ l of 0.2 M SJCRH30 cells resuspended in buffer R to the above centrifuge tube containing RNP (the sample group without small molecule 5 used cells not pre-treated with small molecule 5, and the sample group with small molecule 5 used cells pre-treated with 30 ⁇ M small molecule 5), and mix by carefully pipetting with a pipette tip.
  • Electroporation Turn on the Neon electroporator (Thermo Fisher) and set the electroporation parameters: Voltage: 1200 V, Width: 20 ms, Pulse: 3 pulses.
  • small molecule 1 or small molecule 2 and small molecule 5 can increase the knock-in efficiency by 68.7% (dsDNA template) and 46% (ssDNA template) compared to not adding small molecules.
  • Embodiment 6 is a diagrammatic representation of Embodiment 6
  • GFP green fluorescent protein
  • Small molecule 1 and small molecule 2 After electroporation, the cells were cultured in a medium containing small molecules for 24 hours; after 24 hours, the cells were switched from the medium containing small molecules to a medium not containing small molecules.
  • Small molecule 5 Before electroporation, the cells are cultured in a medium containing small molecules for less than 24 hours; after electroporation, the cells are cultured in a medium containing small molecules for 24 hours. After 24 hours, the cells are switched from the medium containing small molecules to a medium without small molecules.
  • T cells were isolated and purified from 100M PBMC (AllCells) (Thermo Fisher), and the purified T cells were activated using a T cell activation kit (Thermo Fisher).
  • the activated T cells were cultured in TexMACS TM Medium (Miltenyi Biotec) containing IL2, IL7, and IL15.
  • Two cells need to be prepared, one of which is cultured without adding small molecules, and the other is cultured with small molecule 5 added to it at a final concentration of 30 ⁇ M within 24 hours before electroporation.
  • RNP formation Add TrueCut HiFi Cas9 protein (Thermo Fisher), TRAC-sgRNA (SEQ ID NO: 3) (GenScript, SafeEdit), TRAC-dsDNA template (SEQ ID NO: 8) or TRAC plasmid template (SEQ ID NO: 9) (GenScript) and buffer R (Thermo Fisher) as shown in Table 5 into a new, RNase-free centrifuge tube and incubate at room temperature for 5-15 min to form RNP.
  • Electroporation system Add 6 ⁇ l of 0.5M T cells resuspended in buffer R to the above centrifuge tube containing RNP (the sample group without small molecule 5 uses cells not pre-treated with small molecule 5, and the sample group with small molecule 5 uses cells pre-treated with 30 ⁇ M small molecule 5), and mix by carefully pipetting with a pipette tip.
  • Electroporation Turn on the Neon electroporator (Thermo Fisher) and set the electroporation parameters: Voltage: 1700 V, Width: 10 ms, Pulse: 3 pulses.
  • small molecule 1 or small molecule 2 and small molecule 5 can increase the knock-in efficiency by 90%-150%, regardless of whether it is a dsDNA or plasmid type template.
  • Embodiment 7 is a diagrammatic representation of Embodiment 7:
  • a 6bp HindIII restriction endonuclease recognition sequence was inserted into the HPRT site of Jurkat cells and SJCRH30 cells; a 33bp HiBiT tag was inserted into the VEGFA site of H1975 cells and A549 cells.
  • the method of detecting knock-in efficiency by enzyme digestion, gel running, and Agilent 2100 Bioanalyzer System further verified that the combination of these two small molecules has a wide range of applicability in improving knock-in efficiency.
  • Small molecule 1 and small molecule 2 After electroporation, the cells were cultured in a medium containing small molecules for 24 hours; after 24 hours, the cells were switched from the medium containing small molecules to a medium not containing small molecules.
  • Small molecule 5 Before electroporation, the cells are cultured in a medium containing small molecules for less than 24 hours; after electroporation, the cells are cultured in a medium containing small molecules for 24 hours. After 24 hours, the cells are switched from the medium containing small molecules to a medium without small molecules.
  • Cells were passaged every 2-3 days, and cells required for electroporation were plated one day before electroporation. Two aliquots of each cell type were prepared before electroporation, one of which was cultured without small molecules, and the other was cultured with small molecules 5 added to a final concentration of 30 ⁇ M within 24 hours before electroporation.
  • RNP formation eSpCas9 protein (GenScript, Z03470-300), HPRT-sgRNA (SEQ ID NO: 10) or VEGFA-sgRNA (SEQ ID NO: 11) (GenScript, SafeEdit), HPRT-ssDNA template (SEQ ID NO: 12) or VEGFA-ssDNA template (SEQ ID NO: 13) (GenScript) and buffer R (Thermo Fisher) were added to a new, RNase-free centrifuge tube as shown in Table 7 and incubated at room temperature for 5-15 min to form RNP.
  • Electroporation system Add 6 ⁇ l of cells resuspended in buffer R (Jurkat: 0.5 M; SJCRH30: 0.2 M; H1975: 0.2 M; A549: 0.2 M) to the above centrifuge tube containing RNP (the sample group without small molecule 5 used cells not pre-treated with small molecule 5, and the sample group with small molecule 5 used cells pre-treated with 30 ⁇ M small molecule 5), and mix carefully by pipetting with a pipette tip.
  • buffer R Jurkat: 0.5 M; SJCRH30: 0.2 M; H1975: 0.2 M; A549: 0.2 M
  • Electroporation Turn on the Neon electroporator (Thermo Fisher) and set the electroporation parameters: Jurkat: Voltage: 1600V, Width: 10ms, Pulse: 3pulses; SJCRH30: Voltage: 1200V, Width: 20ms, Pulse: 3pulses; H1975: Voltage: 1400V, Width: 20ms, Pulse: 2pulses; A549: Voltage: 1200V, Width: 20ms, Pulse: 3pulses.
  • HPRT-F/R and VEGFA-F/R were synthesized at Nanjing GenScript Biotechnology Co., Ltd.
  • the concentration of the gel-recovered product was determined using Qubit 1X dsDNA BR (Broad Range) Assay Kits. 100 ng of the gel-recovered product was subjected to enzyme digestion (Jurkat and SJCRH30: HindIII (NEB); H1975 and A549: BsrBI (NEB)). The enzyme digestion system was as follows:
  • the enzyme digestion procedure is as follows:
  • the combined use of small molecules can increase the knock-in efficiency by 461% (small molecule 1 + small molecule 5) and 449% (small molecule 2 and small molecule 5); at the HPRT site in SJCRH30 cells, compared with not adding small molecules, the combined use of small molecules can increase the knock-in efficiency by 194% (small molecule 1 + small molecule 5) and 184% (small molecule 2 and small molecule 5); at the VEGFA site in H1975 cells, compared with not adding small molecules, the combined use of small molecules can increase the knock-in efficiency by 82.6% (small molecule 1 + small molecule 5) and 43.6% (small molecule 2 and small molecule 5); at the VEGFA site in A549 cells, compared with not adding small molecules, the combined use of small molecules can increase the knock-in efficiency by 65.3% (small molecule 1 + small molecule 5) and 63.0% (small molecule 2 and small molecule 5).
  • the two small molecules can greatly improve the efficiency of gene knock-in in different cell lines and different sites, and have wide applicability.
  • Embodiment 8 is a diagrammatic representation of Embodiment 8
  • GFP green fluorescent protein
  • IDT HDR enhancer V1 30 ⁇ M (recommended concentration on IDT official website).
  • Small molecule 1, small molecule 2, IDT HDR enhancer After electroporation, the cells were cultured in a medium containing small molecules for 24 hours. After 24 hours, the cells were switched from a medium containing small molecules to a medium without small molecules.
  • Small molecule 5 Before electroporation, the cells are cultured in a medium containing small molecules for less than 24 hours; after electroporation, the cells are cultured in a medium containing small molecules for 24 hours. After 24 hours, the cells are switched from the medium containing small molecules to a medium without small molecules.
  • the cells were passaged every 2-3 days, and the cells required for electroporation were plated 1 day before electroporation.
  • cells were plated before electroporation two aliquots of cells were prepared, one of which was cultured without adding small molecules, and the other aliquot of cells was added with small molecules at a final concentration of 30 ⁇ M within 24 hours before electroporation.
  • RNP formation eSpCas9 protein (GenScript, Z03470-300), RAB11A-sgRNA (SEQ ID NO: 1) (GenScript, EasyEdit), RAB11A-dsDNA template (SEQ ID NO: 2) (GenScript) and buffer R (Thermo Fisher) were added to a new, RNase-free centrifuge tube as shown in Table 3 (Jurkat cells) and incubated at room temperature for 5-15 min to form RNP.
  • Electroporation system Add 6 ⁇ l of 0.5 M Jurkat cells resuspended in buffer R to the above centrifuge tube containing RNP (the sample group without small molecule 5 used cells not pre-treated with small molecule 5, and the sample group with small molecule 5 used cells pre-treated with 30 ⁇ M small molecule 5), and mix by carefully pipetting with a pipette tip.
  • Electroporation Turn on the Neon electroporator (Thermo Fisher) and set the electroporation parameters: Voltage: 1700 V, Width: 20 ms, Pulse: 1 pulses.
  • the knock-in efficiency was increased by about 30% when small molecule 1 or 2 and small molecule 5 were used in combination; compared with IDT HDR enhancer, the knock-in efficiency was increased by about 50% when small molecule 1 or 2 and small molecule 5 were used in combination.
  • Embodiment 9 is a diagrammatic representation of Embodiment 9:
  • a 6 bp HindIII restriction endonuclease recognition sequence was inserted into the TRAC site of HEK293T cells, and the knock-in efficiency was detected using the Agilent 2100 Bioanalyzer System.
  • the small molecule combination (small molecule 1 + small molecule 5) was further compared with the commercial IDT The role of HDR enhancer V1 in improving knock-in efficiency.
  • the small molecule combination is: 5 ⁇ M small molecule 1 + 30 ⁇ M small molecule 5;
  • the concentration of HDR enhancerV1 is: 30 ⁇ M (recommended concentration on IDT official website).
  • IDT HDR enhancer V1 After electroporation, cells are cultured in a medium containing small molecules for 24 hours; after 24 hours, the cells are switched from a medium containing small molecules to a medium without small molecules.
  • Small molecule 5 Before electroporation, the cells are cultured in a medium containing small molecules for less than 24 hours; after electroporation, the cells are cultured in a medium containing small molecules for 24 hours. After 24 hours, the cells are switched from the medium containing small molecules to a medium without small molecules.
  • the cells were passaged every 2-3 days, and the cells required for electroporation were plated 1 day before electroporation.
  • cells were plated before electroporation two aliquots of cells were prepared, one of which was cultured without adding small molecules, and the other aliquot of cells was added with small molecules at a final concentration of 30 ⁇ M within 24 hours before electroporation.
  • RNP formation Add Cas9 protein (Thermo Fisher, A36499), TRAC-sgRNA (SEQ ID NO: 3) (GenScript, EasyEdit), TRAC-86bp dsODN template (annealing product of SEQ ID NO: 4 and 5) (GenScript) and buffer R (Thermo Fisher) as shown in Table 8 into a new, RNase-free centrifuge tube and incubate at room temperature for 5-15 min to form RNP.
  • Cas9 protein Thermo Fisher, A36499
  • TRAC-sgRNA SEQ ID NO: 3
  • TRAC-86bp dsODN template annealing product of SEQ ID NO: 4 and 5
  • buffer R Thermo Fisher
  • Electroporation system Add 6 ⁇ l of 0.2 M HEK293T cells resuspended in buffer R to the above centrifuge tube containing RNP (the sample group without small molecule 5 used cells not pre-treated with small molecule 5, and the sample group with small molecule 5 used cells pre-treated with 30 ⁇ M small molecule 5), and mix by carefully pipetting with a pipette tip.
  • Electroporation Turn on the Neon electroporator (Thermo Fisher) and set the electroporation parameters: Voltage: 1200 V, Width: 10 ms, Pulse: 3 pulses.
  • TRAC-F and TRAC-R were synthesized at Nanjing GenScript Biotechnology Co., Ltd.
  • the gel recovery product was measured for concentration using Nanodrop, and 200 ng of the gel recovery product was subjected to HindIII (NEB) digestion.
  • the digestion system was as follows:
  • the enzyme digestion procedure is as follows:
  • the knock-in efficiency was increased by 31.9% when small molecules 1 and 5 were used in combination.
  • the optimized small molecule combination was superior to the commercial IDT HDR enhancer in improving the knock-in efficiency.
  • the CRISPR/Cas9 system is a widely used genome editing tool that can achieve precise gene editing at specific sites.
  • the present invention uses two small molecule compounds together to provide a method with a wide range of applications that can significantly improve the efficiency of CRISPR/Cas9-mediated precision gene editing.
  • the present invention uses two small molecule compounds in combination to provide a method for better improving the efficiency of CRISPR/Cas9-mediated precise gene editing, the method comprising:
  • the small molecule 1 or small molecule 2 and small molecule 5 at the optimal concentration are used in combination to improve the knock-in efficiency at different sites in the above-mentioned different cells.
  • the present invention uses small molecule 1 or small molecule 2 and small molecule 5 in combination, which can better improve the knock-in efficiency in different cell lines, different sites and different donor lengths, and is superior to some commercial small molecule enhancers.

Landscapes

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

Abstract

Provided is a method for enhancing gene knock-in efficiency mediated by a CRISPR gene editing system in cells, comprising placing the cells in contact with a DNA-dependent protein kinase (DNA-PK) inhibitor and a cell division cyclin 7 (Cdc7) inhibitor during the process of performing a gene knock-in operation on the cells. Further provided is a kit for enhancing the gene knock-in efficiency mediated by the CRISPR gene editing system in cells. By means of the combined use of the DNA-PK inhibitor and the Cdc7 inhibitor, the method and kit provided can both significantly improve gene knock-in efficiency in different cell lines, different insertion sites, and different donor template lengths.

Description

一种提高CRISPR/Cas9介导的基因组敲入编辑效率的方法A method to improve the efficiency of CRISPR/Cas9-mediated genome knock-in editing
相关申请的交叉引用CROSS-REFERENCE TO RELATED APPLICATIONS
本申请要求申请号为CN202211263292.9、申请日为2022年10月14日的中国专利申请的优先权,该申请通过引用以其全文并入本文。This application claims priority to Chinese patent application number CN202211263292.9 and filing date October 14, 2022, which is incorporated herein by reference in its entirety.
技术领域Technical Field
本文涉及提高CRISPR/Cas9介导的基因组敲入编辑效率的方法,尤其是利用小分子蛋白酶抑制剂组合来提高CRISPR/Cas9介导的基因组敲入编辑效率的方法。本文还涉及包括上述小分子蛋白酶抑制剂的试剂盒。The present invention relates to a method for improving the efficiency of CRISPR/Cas9-mediated genome knock-in editing, in particular, a method for improving the efficiency of CRISPR/Cas9-mediated genome knock-in editing using a combination of small molecule protease inhibitors. The present invention also relates to a kit including the above-mentioned small molecule protease inhibitors.
背景技术Background technique
CRISPR/Cas9系统是一种由RNA引导的基因组编辑工具,为科研人员提供了一种简单的、能快速修饰各种生物基因组的方法。随着CRISPR/Cas9系统的广泛使用,以及精准的基因编辑正在或即将在临床上用于多种疾病,提高该系统介导的基因组精准编辑效率的呼声也不断高涨。The CRISPR/Cas9 system is an RNA-guided genome editing tool that provides researchers with a simple method to quickly modify the genomes of various organisms. With the widespread use of the CRISPR/Cas9 system and the fact that precise gene editing is being or will soon be used clinically for a variety of diseases, the call to improve the efficiency of precise genome editing mediated by this system is growing.
使用该系统过程中,Cas9在向导RNA(gRNA)的引导下定位到基因组DNA的靶位点并实现靶向基因序列的切割,形成DNA双链断裂(DSB),然后通过细胞内修复机制完成DSB的修复。在哺乳动物中,细胞修复DSB的途径主要有两种:非同源末端连接(NHEJ)和同源重组修复(HDR),基因编辑的结果主要取决于这两种修复途径之间的选择。NHEJ途径修复DSB是通过末端直接相连的方式进行的,这种修复不需要大片段的同源序列识别,只需要1-5bp的碱基配对就可以进行修复,属于一种易错修复,精确度很差。HDR途径能够在高度同源性DNA模板存在下,通过同源重组将一段DNA插入编辑位点来修复DSB,从而实现将一段DNA序列精准地插入特定基因组位点的目的。尽管HDR实现的精准插入颇具优势,但修复途径的选择在生物学背景下存在偏差。哺乳动物细胞的自身修复机制会优先使用NHEJ而不是HDR:NHEJ在整个细胞周期中均处于活跃状态,而HDR限于S/G2期;NHEJ比HDR快;且NHEJ也会抑制HDR修复途径。因此是否可以通过以及如何通过调控细胞修复途径来提高CRISPR/Cas9介导的基因组精准插入编辑效率的相关研究也越来越多。During the use of this system, Cas9, under the guidance of guide RNA (gRNA), locates to the target site of genomic DNA and cuts the targeted gene sequence, forming a DNA double-strand break (DSB), and then repairs the DSB through the intracellular repair mechanism. In mammals, there are two main ways for cells to repair DSB: non-homologous end joining (NHEJ) and homologous recombination repair (HDR). The results of gene editing mainly depend on the choice between these two repair pathways. The NHEJ pathway repairs DSB by directly connecting the ends. This repair does not require large fragments of homologous sequence recognition, but only requires 1-5bp base pairing for repair. It is an error-prone repair with poor accuracy. The HDR pathway can repair DSB by inserting a piece of DNA into the editing site through homologous recombination in the presence of a highly homologous DNA template, thereby achieving the purpose of accurately inserting a DNA sequence into a specific genomic site. Although the precise insertion achieved by HDR is quite advantageous, the choice of repair pathway is biased in the biological context. The repair mechanism of mammalian cells will give priority to NHEJ rather than HDR: NHEJ is active throughout the cell cycle, while HDR is limited to the S/G2 phase; NHEJ is faster than HDR; and NHEJ also inhibits the HDR repair pathway. Therefore, there are more and more studies on whether and how to improve the efficiency of CRISPR/Cas9-mediated precise genome insertion editing by regulating cell repair pathways.
许多团队针对CRISPR/Cas9介导的基因编辑条件下的NHEJ以及HDR修复途径的调控进行了深入研究,并通过大量实验筛选调控NHEJ和HDR修复途径的小分子化合物,发现并成功筛选了各种细胞类型中能提高CRISPR/Cas9介导的HDR效率的功能小分子。虽然通过筛选发现的小分子种类有很多,然而这些小分子具有一定的细胞类型特 异性以及上下文依赖性,且各作者描述的提高程度也存在一定的差异。因此亟需找到一种适用范围更广,且能够显著提高基因组敲入编辑效率的小分子增强剂。Many teams have conducted in-depth research on the regulation of NHEJ and HDR repair pathways under CRISPR/Cas9-mediated gene editing conditions, and have screened small molecule compounds that regulate NHEJ and HDR repair pathways through a large number of experiments. They have discovered and successfully screened functional small molecules that can improve the efficiency of CRISPR/Cas9-mediated HDR in various cell types. Although there are many types of small molecules discovered through screening, these small molecules have certain cell type characteristics. The enhancement degree described by different authors is also different. Therefore, it is urgent to find a small molecule enhancer with a wider range of applications and the ability to significantly improve the efficiency of genome knock-in editing.
发明内容Summary of the invention
在一方面,本文提供了在细胞内增强CRISPR基因编辑系统介导的基因敲入效率的方法,其包括在对所述细胞进行基因敲入操作过程中让所述细胞与DNA依赖性蛋白激酶(DNA-PK)抑制剂和细胞分裂周期蛋白7(Cdc7)抑制剂接触。In one aspect, the present invention provides a method for enhancing the efficiency of gene knock-in mediated by a CRISPR gene editing system in a cell, comprising contacting the cell with a DNA-dependent protein kinase (DNA-PK) inhibitor and a cell division cycle protein 7 (Cdc7) inhibitor during the gene knock-in operation on the cell.
在一些实施方案中,所述CRISPR基因编辑系统包括Cas9蛋白。In some embodiments, the CRISPR gene editing system includes a Cas9 protein.
在一些实施方案中,所述CRISPR基因编辑系统还包括向导RNA(gRNA)和用于进行同源重组修复(HDR)的供体模板。In some embodiments, the CRISPR gene editing system further comprises a guide RNA (gRNA) and a donor template for homologous recombination repair (HDR).
在一些实施方案中,所述供体模板为单链或双链形式的DNA分子。In some embodiments, the donor template is a DNA molecule in single-stranded or double-stranded form.
在一些实施方案中,所述DNA-PK抑制剂选自式(I)的化合物、式(II)的化合物及其组合,其中式(I)的化合物中X为氢或者氘:
In some embodiments, the DNA-PK inhibitor is selected from a compound of formula (I), a compound of formula (II), and a combination thereof, wherein X in the compound of formula (I) is hydrogen or deuterium:
其中优选地,式(I)的化合物为:
Preferably, the compound of formula (I) is:
在一些实施方案中,所述Cdc7抑制剂为式(V)的化合物或其盐酸盐:
In some embodiments, the Cdc7 inhibitor is a compound of formula (V) or its hydrochloride salt:
在一些实施方案中,所述式(I)的化合物以不低于1μM的浓度与所述细胞接触;所述式(II)的化合物以不低于0.5μM的浓度与所述细胞接触;或者,所述式(V)的化合物以不低于10μM的浓度与所述细胞接触。In some embodiments, the compound of formula (I) is contacted with the cell at a concentration of not less than 1 μM; the compound of formula (II) is contacted with the cell at a concentration of not less than 0.5 μM; or the compound of formula (V) is contacted with the cell at a concentration of not less than 10 μM.
在一些实施方案中,所述式(I)的化合物以1-30μM的浓度与所述细胞接触,并且所述式(V)的化合物以10-50μM的浓度与所述细胞接触。In some embodiments, the compound of Formula (I) is contacted with the cell at a concentration of 1-30 μM, and the compound of Formula (V) is contacted with the cell at a concentration of 10-50 μM.
在一些实施方案中,所述式(II)的化合物以0.5-5μM的浓度与所述细胞接触,并且所述式(V)的化合物以10-50μM的浓度与所述细胞接触。In some embodiments, the compound of Formula (II) is contacted with the cell at a concentration of 0.5-5 μM, and the compound of Formula (V) is contacted with the cell at a concentration of 10-50 μM.
在一些实施方案中,所述式(I)的化合物以5-20μM的浓度与所述细胞接触,并且所述式(V)的化合物以20-40μM的浓度与所述细胞接触。In some embodiments, the compound of Formula (I) is contacted with the cell at a concentration of 5-20 μM, and the compound of Formula (V) is contacted with the cell at a concentration of 20-40 μM.
在一些实施方案中,所述式(II)的化合物以2-4μM的浓度与所述细胞接触,并且所述式(V)的化合物以20-40μM的浓度与所述细胞接触。In some embodiments, the compound of Formula (II) is contacted with the cell at a concentration of 2-4 μM, and the compound of Formula (V) is contacted with the cell at a concentration of 20-40 μM.
在一些实施方案中,所述式(I)的化合物以5μM的浓度与所述细胞接触,并且所述式(V)的化合物以30μM的浓度与所述细胞接触。In some embodiments, the compound of Formula (I) is contacted with the cell at a concentration of 5 μM, and the compound of Formula (V) is contacted with the cell at a concentration of 30 μM.
在一些实施方案中,所述式(II)的化合物以2μM的浓度与所述细胞接触,并且所述式(V)的化合物以30μM的浓度与所述细胞接触。In some embodiments, the compound of Formula (II) is contacted with the cell at a concentration of 2 μM, and the compound of Formula (V) is contacted with the cell at a concentration of 30 μM.
在一些实施方案中,所述细胞为HEK293T、Jurkat、SJCRH30、H1975或A549细胞。In some embodiments, the cell is a HEK293T, Jurkat, SJCRH30, H1975, or A549 cell.
在一些实施方案中,所述细胞为T细胞。In some embodiments, the cell is a T cell.
在一些实施方案中,对所述细胞进行基因敲入操作过程中让所述细胞与所述式(I)的化合物、所述式(II)的化合物和所述式(V)的化合物接触。 In some embodiments, the cell is contacted with the compound of formula (I), the compound of formula (II), and the compound of formula (V) during a gene knock-in procedure.
在一些实施方案中,所述基因敲入操作通过转染进行,并在所述转染之前使所述细胞与所述式(V)的化合物接触。In some embodiments, the gene knock-in operation is performed by transfection, and the cell is contacted with the compound of formula (V) prior to the transfection.
在一些实施方案中,所述基因敲入操作通过转染进行,所述式(I)的化合物和/或所述式(II)的化合物在转染步骤后与所述细胞接触,所述式(V)的化合物在转染步骤前和转染步骤后都与所述细胞接触。In some embodiments, the gene knock-in operation is performed by transfection, and the compound of formula (I) and/or the compound of formula (II) is contacted with the cells after the transfection step, and the compound of formula (V) is contacted with the cells both before and after the transfection step.
另一方面,本文提供了在细胞内增强CRISPR基因编辑系统介导的基因敲入效率的试剂盒,包括DNA依赖性蛋白激酶(DNA-PK)抑制剂和细胞分裂周期蛋白7(Cdc7)抑制剂。On the other hand, the present invention provides a kit for enhancing the gene knock-in efficiency mediated by the CRISPR gene editing system in cells, comprising a DNA-dependent protein kinase (DNA-PK) inhibitor and a cell division cycle protein 7 (Cdc7) inhibitor.
在一些实施方案中,所述试剂盒还包括Cas9蛋白或其编码核酸分子。In some embodiments, the kit further comprises a Cas9 protein or a nucleic acid molecule encoding the Cas9 protein.
在一些实施方案中,所述试剂盒还包括向导RNA(gRNA)和/或用于进行同源重组修复(HDR)的供体模板。In some embodiments, the kit further comprises a guide RNA (gRNA) and/or a donor template for performing homologous recombination repair (HDR).
在一些实施方案中,所述供体模板为单链或双链形式的DNA分子。In some embodiments, the donor template is a DNA molecule in single-stranded or double-stranded form.
在一些实施方案中,所述DNA-PK抑制剂选自式(I)的化合物、式(II)的化合物及其组合,其中式(I)的化合物中X为氢或者氘:
In some embodiments, the DNA-PK inhibitor is selected from a compound of formula (I), a compound of formula (II), and a combination thereof, wherein X in the compound of formula (I) is hydrogen or deuterium:
其中优选地,式(I)的化合物为:
Preferably, the compound of formula (I) is:
在一些实施方案中,所述Cdc7抑制剂为式(V)的化合物或其盐酸盐:
In some embodiments, the Cdc7 inhibitor is a compound of formula (V) or its hydrochloride salt:
在一些实施方案中,所述细胞为HEK293T、Jurkat、SJCRH30、H1975或A549细胞。In some embodiments, the cell is a HEK293T, Jurkat, SJCRH30, H1975, or A549 cell.
在一些实施方案中,所述细胞为T细胞。In some embodiments, the cell is a T cell.
本文提供的方法通过DNA依赖性蛋白激酶(DNA-PK)抑制剂和细胞分裂周期蛋白7(Cdc7)抑制剂联合使用,使得在不同的细胞系、不同的插入位点以及不同长度的供体模板的情况下,都可以显著地提高CRISPR基因编辑系统的基因敲入效率,且优于某些商业化的小分子增强剂。The method provided in this article uses a DNA-dependent protein kinase (DNA-PK) inhibitor and a cell division cycle protein 7 (Cdc7) inhibitor in combination, so that the gene knock-in efficiency of the CRISPR gene editing system can be significantly improved in different cell lines, different insertion sites, and different lengths of donor templates, and is superior to some commercial small molecule enhancers.
附图说明BRIEF DESCRIPTION OF THE DRAWINGS
图1显示了小分子1在不同浓度下的细胞活率(A)和敲入效率(B)。Figure 1 shows the cell viability (A) and knock-in efficiency (B) of small molecule 1 at different concentrations.
图2显示了小分子2在不同浓度下的细胞活率(A)和敲入效率(B)。FIG2 shows the cell viability (A) and knock-in efficiency (B) of small molecule 2 at different concentrations.
图3显示了小分子3在不同浓度下的细胞活率(A)和敲入效率(B)。FIG3 shows the cell viability (A) and knock-in efficiency (B) of small molecule 3 at different concentrations.
图4显示了小分子4在不同浓度下的细胞活率(A)和敲入效率(B)。FIG4 shows the cell viability (A) and knock-in efficiency (B) of small molecule 4 at different concentrations.
图5显示了小分子5在不同浓度下的细胞活率(A)和敲入效率(B)。FIG5 shows the cell viability (A) and knock-in efficiency (B) of small molecule 5 at different concentrations.
图6显示了小分子5的不同孵育方式。方式①:电转前细胞在加入了小分子5的培养基中培养24小时,电转后立即将细胞置于不含小分子5的培养基中;方式②:电转后立即将细胞置于含有小分子5的培养基中培养24h;方式③:电转前后均将细胞置于含有小分子5的培养基中培养24h。电转缓冲液中不含小分子5。Figure 6 shows different incubation methods for small molecule 5. Method ①: cells were cultured in a medium containing small molecule 5 for 24 hours before electroporation, and immediately after electroporation, the cells were placed in a medium without small molecule 5; Method ②: cells were placed in a medium containing small molecule 5 for 24 hours immediately after electroporation; Method ③: cells were placed in a medium containing small molecule 5 for 24 hours before and after electroporation. Small molecule 5 was not contained in the electroporation buffer.
图7显示了小分子5在不同孵育方式下的细胞活率(A)和敲入效率(B)。 FIG7 shows the cell viability (A) and knock-in efficiency (B) of small molecule 5 under different incubation modes.
图8显示了小分子的作用浓度以及不同组合方式,其中“+”表示添加,“-”表示不添加。Figure 8 shows the effective concentrations of small molecules and different combinations, where "+" indicates addition and "-" indicates no addition.
图9显示了不同分子组合作用下4天时细胞活率。FIG. 9 shows the cell viability at 4 days under the action of different molecular combinations.
图10显示了不同分子组合作用下7天时细胞敲入效率。FIG. 10 shows the knock-in efficiency of cells at 7 days under the action of different molecular combinations.
图11显示了不同小分子及其组合的作用浓度范围设计,其中“+”表示添加,“-”表示不添加。FIG11 shows the design of the action concentration range of different small molecules and their combinations, where “+” indicates addition and “-” indicates no addition.
图12显示了不同小分子及其组合的作用浓度范围下7天时细胞敲入效率。FIG. 12 shows the cell knock-in efficiency at 7 days under the concentration range of different small molecules and their combinations.
图13显示了在不同小分子组合作用下使用dsDNA和ssDNA两种类型的供体模板在7天时细胞敲入效率。FIG. 13 shows the knock-in efficiency of cells at 7 days using two types of donor templates, dsDNA and ssDNA, under the action of different small molecule combinations.
图14显示了在T细胞体系中使用dsDNA和质粒供体模板验证3天时细胞活率。Figure 14 shows the validation of cell viability at day 3 using dsDNA and plasmid donor templates in a T cell system.
图15显示了在T细胞体系中使用dsDNA和质粒供体模板验证7天时细胞敲入效率。FIG. 15 shows validation of cell knock-in efficiency at 7 days using dsDNA and plasmid donor templates in a T cell system.
图16显示了不同细胞系不同位点电转后3天时酶切图。FIG16 shows the restriction enzyme cleavage patterns of different cell lines at different sites 3 days after electroporation.
图17显示了不同细胞系不同位点电转后3天时细胞敲入效率对比。FIG. 17 shows a comparison of cell knock-in efficiency at 3 days after electroporation of different sites in different cell lines.
图18显示了不同小分子组合作用与IDT HDR增强剂作用下7天时细胞敲入效率对比。Figure 18 shows the comparison of cell knock-in efficiency at 7 days under the action of different small molecule combinations and IDT HDR enhancer.
图19显示了小分子组合作用与IDT HDR增强剂作用下3天时细胞敲入效率对比。Figure 19 shows the comparison of cell knock-in efficiency at 3 days under the action of small molecule combination and IDT HDR enhancer.
具体实施方式Detailed ways
除非另有说明,本发明所用的技术和科学术语具有与本发明所属领域的普通技术员通常所理解的含义。Unless otherwise defined, technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
术语“或”是指列举的可选择要素中的单个要素,除非上下文明确地另外指出。The term "or" refers to a single element of the listed alternative elements unless the context clearly dictates otherwise.
术语“和/或”是指所列举的可选择要素中的任意一个、任意两个、任意三个、任意更多个或其全部。The term "and/or" means any one, any two, any three, any more or all of the listed optional elements.
术语“包含”或“包括”指包括所述的要素、整数或步骤,但是不排除任意其他要素、整数或步骤。当使用“包含”或“包括”时,除非另有指明,否则也涵盖由所述及的要素、整数或步骤组成的情形。例如,当提及某组合或组合物“包括A和B”时,也旨在涵盖由A和B组成的组合或组合物。The term "comprising" or "including" means including the stated elements, integers or steps, but does not exclude any other elements, integers or steps. When "comprising" or "including" is used, unless otherwise specified, the situation consisting of the stated elements, integers or steps is also covered. For example, when referring to a combination or composition "comprising A and B", it is also intended to cover the combination or composition consisting of A and B.
在提及具体数值时,除非上下文另有说明或暗示,通常可认为同时提及所列数值的±10%范围内的任何数值。本领域技术人员可理解,由于仪器检测精度、操作过程中引入的误差等原因,所给出的检测结果、操作时间等数值可以在小范围内变动。例如,当提到培养细胞24h时,应理解培养细胞24h±2.4h也是可行。又如,当提到检测到的敲入效率为20%时,实际敲入效率可能为20%±2%。When referring to specific values, unless otherwise specified or implied by the context, it is generally considered that any value within the range of ±10% of the listed value is referred to at the same time. It will be understood by those skilled in the art that due to reasons such as instrument detection accuracy and errors introduced during operation, the values of the test results, operation time, etc. given may vary within a small range. For example, when it is mentioned that cells are cultured for 24 hours, it should be understood that culturing cells for 24 hours ± 2.4 hours is also feasible. For another example, when it is mentioned that the detected knock-in efficiency is 20%, the actual knock-in efficiency may be 20% ± 2%.
“CRISPR(Clustered Regularly Interspaced Short Palindromic Repeats)基因编辑技术”是一种由RNA指导的通过Cas核酸酶对靶基因进行DNA编辑的技术。该技术所使用的CRISPR基因编辑系统包括Cas核酸酶和引导RNA(single-guide RNA,sgRNA或gRNA), 视情况可还包括作为修复模板的双链DNA(dsDNA)或单链DNA(ssDNA)。sgRNA的一部分序列可以与Cas核酸酶结合,另外部分序列(crRNA)可以与靶基因的部分序列互补,借助sgRNA的识别作用使得Cas核酸酶可以在靶基因特定位点形成单链或双链切口。细胞通常会通过两种方式对断裂链进行DNA修复,这两种方式分别是同源重组修复机制(homology-directed repair,HDR)和非同源末端连接修复机制(non-homologous end joining,NHEJ)。在一些应用中,可以将CRISPR基因编辑技术用来对细胞的基因进行基因敲除操作。这种情况下,通常只需要考虑破坏该基因的正常编码功能,例如引起移码突变或基因片段缺失,从而不能产生有正常功能的产物(如蛋白)。通常,可以在向细胞中引入Cas核酸酶(例如Cas9)和sgRNA后,再筛选出不表达待敲除基因的产物的细胞,即可达到基因敲除的目的。在另一些应用中,可以将CRISPR基因编辑技术用来对细胞进行基因敲入操作(knock in),即向细胞基因组中引入目的基因,使该细胞能够生成带有目的基因的产物。这种情况下,可通过在向细胞引入Cas核酸酶和引导RNA的同时提供带有目的基因的供体模板来实现。供体模板序列上可包括目的基因序列和位于目的基因两侧的同源臂序列。借助同源臂序列与Cas核酸酶在基因组上产生的缺口部位序列的同源重组,达到将目的基因引入基因组的目的。在这个过程中,Cas核酸酶(如Cas9)的切割效率、细胞基因组双链DNA断裂后以同源重组方式进行修复的概率、以供体模板重组的概率都会影响最终的基因敲入效率。"CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) gene editing technology" is a technology that uses Cas nuclease to edit the target gene DNA under the guidance of RNA. The CRISPR gene editing system used in this technology includes Cas nuclease and guide RNA (single-guide RNA, sgRNA or gRNA), Depending on the situation, double-stranded DNA (dsDNA) or single-stranded DNA (ssDNA) as a repair template may also be included. A portion of the sequence of sgRNA can bind to the Cas nuclease, and another portion of the sequence (crRNA) can be complementary to a portion of the sequence of the target gene. With the help of the recognition of sgRNA, the Cas nuclease can form a single-stranded or double-stranded incision at a specific site of the target gene. Cells usually repair DNA broken chains in two ways, namely homologous recombination repair mechanism (homology-directed repair, HDR) and non-homologous end joining repair mechanism (non-homologous end joining, NHEJ). In some applications, CRISPR gene editing technology can be used to knock out genes in cells. In this case, it is usually only necessary to consider destroying the normal coding function of the gene, such as causing frameshift mutations or gene fragment deletions, so that products with normal functions (such as proteins) cannot be produced. Usually, after introducing Cas nucleases (such as Cas9) and sgRNA into cells, cells that do not express the products of the gene to be knocked out can be screened out to achieve the purpose of gene knockout. In other applications, CRISPR gene editing technology can be used to knock in cells, that is, to introduce a target gene into the cell genome so that the cell can produce a product with the target gene. In this case, it can be achieved by providing a donor template with the target gene while introducing Cas nuclease and guide RNA into the cell. The donor template sequence may include the target gene sequence and homologous arm sequences on both sides of the target gene. The purpose of introducing the target gene into the genome is achieved by homologous recombination of the homologous arm sequence and the gap site sequence generated by the Cas nuclease on the genome. In this process, the cutting efficiency of Cas nuclease (such as Cas9), the probability of repairing the double-stranded DNA of the cell genome by homologous recombination, and the probability of recombination with the donor template will affect the final gene knock-in efficiency.
本文中“Cas9蛋白”是CRISPR/Cas9系统发挥核酸酶活性的关键,含有两个具有切割活性的结构域:HNH结构域和RuvC结构域,HNH和RuvC可分别剪切DNA的两条链形成双链断裂。Cas9蛋白可以是来源于Streptococcuspyogenes的重组Cas9蛋白SpCas9。Cas9蛋白也可以是Cas9蛋白的变体,如对Cas9蛋白定向改造得到的特异性显著提高的Cas9蛋白变体eSpCas9(1.0)、eSpCas9(1.1)和SpCas9-HF1等。In this article, "Cas9 protein" is the key to the nuclease activity of the CRISPR/Cas9 system. It contains two domains with cutting activity: the HNH domain and the RuvC domain. HNH and RuvC can cut the two strands of DNA to form double-strand breaks. The Cas9 protein can be a recombinant Cas9 protein SpCas9 derived from Streptococcus pyogenes. The Cas9 protein can also be a variant of the Cas9 protein, such as the Cas9 protein variants eSpCas9 (1.0), eSpCas9 (1.1) and SpCas9-HF1 with significantly improved specificity obtained by directed modification of the Cas9 protein.
当提及sgRNA(或gRNA)时,术语“靶序列”指细胞基因组中与sgRNA的部分序列(crRNA,约20个碱基)互补的核苷酸片段。借助于sgRNA中与靶序列互补的这部分序列,让Cas9等蛋白可以在相对确定的位置在基因组中引入核苷酸序列改变,达到基因敲除或基因敲入的效果。相应地,在本文中,“靶向某指定序列的sgRNA”指该sgRNA的靶序列为该指定序列。When referring to sgRNA (or gRNA), the term "target sequence" refers to a nucleotide fragment in the cell genome that is complementary to a portion of the sgRNA sequence (crRNA, about 20 bases). With the help of this portion of the sgRNA that is complementary to the target sequence, proteins such as Cas9 can introduce nucleotide sequence changes in the genome at a relatively certain position to achieve the effect of gene knockout or gene knockin. Accordingly, in this article, "sgRNA targeting a specified sequence" means that the target sequence of the sgRNA is the specified sequence.
术语“供体模板”在本文中是细胞DNA双链断裂后,同源重组修复(HDR)途径介导基因替换或插入所借助的外源DNA模板。通过sgRNA序列确定Cas9蛋白的切割位点,切割位点两侧的序列为供体模板中的同源臂序列,在同源臂中间加入需要精准插入的目的基因序列即为供体模板的序列。“供体模板”可以是双链DNA(dsDNA)或单链DNA(ssDNA),也可以是单链寡核苷酸(ssODN),甚至可以是环形DNA(如质粒)。其中插入的目的基因可以是任意希望插入到细胞内的基因序列。目的基因序列可包括例如编码受体个体或靶细胞中有缺陷或缺少的蛋白质的基因(核苷酸序列);编码具有所需生物或治疗效果(例如,抗细菌、抗病毒或抗肿瘤/抗癌功能)的蛋白质的基因;编码抑制或减 少有害或另外不希望有的蛋白质产生的RNA的核苷酸序列(例如,编码RNA干扰剂的核苷酸序列);和/或编码抗原蛋白的核苷酸序列。供体模板可以是任意长度的DNA序列,如从几个bp到几千bp。The term "donor template" in this article refers to the exogenous DNA template used by the homologous recombination repair (HDR) pathway to mediate gene replacement or insertion after the double-strand break of the cell's DNA. The cleavage site of the Cas9 protein is determined by the sgRNA sequence, and the sequences on both sides of the cleavage site are the homologous arm sequences in the donor template. The target gene sequence that needs to be accurately inserted in the middle of the homologous arm is the sequence of the donor template. The "donor template" can be double-stranded DNA (dsDNA) or single-stranded DNA (ssDNA), or it can be a single-stranded oligonucleotide (ssODN), or even a circular DNA (such as a plasmid). The inserted target gene can be any gene sequence that you want to insert into the cell. The target gene sequence may include, for example, genes (nucleotide sequences) encoding defective or missing proteins in the recipient individual or target cell; genes encoding proteins with desired biological or therapeutic effects (for example, antibacterial, antiviral or anti-tumor/anti-cancer functions); genes encoding inhibition or reduction The donor template can be a DNA sequence of any length, such as from a few bp to several thousand bp.
术语“基因敲入操作”在本文中指有目的地通过诸如CRISPR基因编辑技术的技术手段向细胞中引入目的基因。在一些情况下,该目的基因可随机整合在细胞基因组中。在另一些情况下,该目的基因可在相对确定的位点整合在细胞基因组中(例如通过采用具有特定序列的上述sgRNA)。通常,该目的基因随后可以在细胞中进行表达,生成诸如目的蛋白的产物,可用于研究目的基因的功能或者用于制备蛋白产物。在采用上述CRISPR基因编辑系统进行基因敲入操作时,该基因敲入操作过程例如可包括准备待敲入基因的细胞,将Cas核酸酶、gRNA和供体模板引入细胞(例如通过电转),继续培养细胞,以及任选地筛选阳性克隆等步骤。The term "gene knock-in operation" refers to the purposeful introduction of a target gene into a cell by means of technical means such as CRISPR gene editing technology. In some cases, the target gene can be randomly integrated into the cell genome. In other cases, the target gene can be integrated into the cell genome at a relatively determined site (for example, by using the above-mentioned sgRNA with a specific sequence). Typically, the target gene can then be expressed in the cell to generate products such as target proteins, which can be used to study the function of the target gene or for preparing protein products. When the above-mentioned CRISPR gene editing system is used for gene knock-in operation, the gene knock-in operation process may, for example, include preparing cells to be knocked into genes, introducing Cas nucleases, gRNAs and donor templates into cells (for example, by electroporation), continuing to culture cells, and optionally screening positive clones and other steps.
术语“敲入效率或敲入编辑效率”在本文中指进行基因敲入操作后细胞群体中在其基因组中包括待敲入基因(目的基因)的细胞相对于细胞群体总细胞数量的比例。基因敲入效率的检测可通过多种方式进行。例如,可通过检测表达目的基因的表达产物,例如荧光蛋白,来确定基因敲入效率。“提高敲入效率”指通过对基因敲入操作过程进行改进,以便获得更高的敲入效率。在本文提供的提高敲入效率的实施方案中,采用在基因敲入操作过程中添加小分子酶抑制剂。如下文所描述的,通过添加DNA依赖性蛋白激酶(DNA-PK)抑制剂和/或细胞分裂周期蛋白7(Cdc7)抑制剂,可显著提高基因敲入效率。The term "knock-in efficiency or knock-in editing efficiency" refers herein to the ratio of cells including a gene to be knocked-in (target gene) in its genome in a cell population after a gene knock-in operation relative to the total number of cells in the cell population. The detection of gene knock-in efficiency can be carried out in a variety of ways. For example, the gene knock-in efficiency can be determined by detecting an expression product expressing the target gene, such as a fluorescent protein. "Improving knock-in efficiency" refers to improving the gene knock-in operation process so as to obtain a higher knock-in efficiency. In the embodiment of improving knock-in efficiency provided herein, a small molecule enzyme inhibitor is added during the gene knock-in operation process. As described below, by adding a DNA-dependent protein kinase (DNA-PK) inhibitor and/or a cell division cycle protein 7 (Cdc7) inhibitor, the gene knock-in efficiency can be significantly improved.
DNA依赖性蛋白激酶(DNA-dependentprotein kinase,DNA-PK)是丝氨酸/苏氨酸激酶,为磷脂酰肌醇3-激酶相关激酶(PI3K)家族成员之一,具有主要定位于细胞核,可通过非同源未端连接的方式进行DNA双链断裂的修复,在维持基因组稳定以及产生抗体多样性的V(D)J重组中发挥着重要作用,其活性受DNA双链断裂的调控。另外,DNA-PK和其组分参与多种其它生理过程,包括染色质结构的调节、端粒维持、转录调节和对复制应激的反应等。常见的DNA-PK抑制剂包括AZD-7648、KU-57788Torin 2、NU5455、VX-984、Samotolisib等。DNA-dependent protein kinase (DNA-PK) is a serine/threonine kinase and a member of the phosphatidylinositol 3-kinase-related kinase (PI3K) family. It is mainly located in the cell nucleus and can repair DNA double-strand breaks by non-homologous end joining. It plays an important role in maintaining genome stability and V(D)J recombination to produce antibody diversity. Its activity is regulated by DNA double-strand breaks. In addition, DNA-PK and its components are involved in a variety of other physiological processes, including the regulation of chromatin structure, telomere maintenance, transcriptional regulation, and response to replication stress. Common DNA-PK inhibitors include AZD-7648, KU-57788Torin 2, NU5455, VX-984, Samotolisib, etc.
细胞分裂周期蛋白7(Cdc7)是丝氨酸/苏氨酸激酶,在起始DNA复制方面起重要作用。从G1期的后期到S期,Cdc7与Dbf4(也称为ASK)形成复合物,并且将Cdc7底物磷酸化,以控制从G1期到S期的转变。常见的Cdc7抑制剂包括XL413、XL413盐酸盐、CAY10572(PHA-767491)、Cdc7-IN-1、Cdc7-IN-5、Cdc7-IN-17、LY3177833等。Cyclin 7 (Cdc7) is a serine/threonine kinase that plays an important role in initiating DNA replication. From the late G1 phase to the S phase, Cdc7 forms a complex with Dbf4 (also known as ASK) and phosphorylates Cdc7 substrates to control the transition from G1 phase to S phase. Common Cdc7 inhibitors include XL413, XL413 hydrochloride, CAY10572 (PHA-767491), Cdc7-IN-1, Cdc7-IN-5, Cdc7-IN-17, LY3177833, etc.
术语“细胞转染”是将外源性基因导入真核细胞的一种技术。细胞转染途径大致可分为三大类:化学介导、生物介导和物理介导。化学介导的方法很多,如经典的磷酸钙共沉淀法、脂质体转染方法和多种阳离子物质介导的技术。生物介导方法,有较为原始的原生质体转染和现在比较多见的各种病毒介导的转染技术。物理介导方法主要有电穿孔法、显微注射和基因枪。在本文中“电转”指电穿孔法,此法利用高脉冲电压破坏细 胞膜电位,使得DNA通过膜上形成的小孔导入稳定/瞬时性转染,该方法适用于所有类型的细胞,但细胞致死率高,且需根据不同细胞类型优化电穿孔实验条件。The term "cell transfection" is a technique for introducing exogenous genes into eukaryotic cells. Cell transfection pathways can be roughly divided into three categories: chemical mediation, biological mediation, and physical mediation. There are many chemical-mediated methods, such as the classic calcium phosphate co-precipitation method, liposome transfection method, and various cationic substance-mediated techniques. Biological-mediated methods include the more primitive protoplast transfection and the more common various virus-mediated transfection techniques. Physical-mediated methods mainly include electroporation, microinjection, and gene guns. In this article, "electroporation" refers to electroporation, which uses high pulse voltage to destroy cells. The cell membrane potential allows DNA to be introduced through the small holes formed on the membrane for stable/transient transfection. This method is applicable to all types of cells, but the cell lethality is high and the electroporation experimental conditions need to be optimized according to different cell types.
本文提供了更好的提高CRISPR/Cas9介导的基因组敲入编辑效率的方法。在一些实施方案中,本发明的具体实施方式可以分为以下几步:This article provides a better method for improving the efficiency of CRISPR/Cas9-mediated genome knock-in editing. In some embodiments, the specific implementation of the present invention can be divided into the following steps:
(1)根据目的细胞以及目的位点,设计并合成对应的sgRNA;通过sgRNA序列确定供体DNA两端的同源臂序列,在同源臂中间加入需要精准插入的DNA序列即为供体DNA的序列,设计并合成供体DNA模板。(1) Design and synthesize the corresponding sgRNA according to the target cells and target sites; determine the homology arm sequences at both ends of the donor DNA through the sgRNA sequence, add the DNA sequence that needs to be accurately inserted in the middle of the homology arm, which is the sequence of the donor DNA, and design and synthesize the donor DNA template.
(2)分别设置小分子1、小分子2、小分子5(结构式见下文)这三种小分子的测试浓度梯度,并在转染前后和细胞进行一定时间的孵育。小分子1、小分子2这两种小分子的孵育条件为:转染后细胞在含有小分子的培养基中培养24h以上;小分子5的孵育条件为:转染前细胞在含有小分子的培养基中培养24h以内,转染后细胞在含有小分子的培养基中培养24h以上。(2) Set the test concentration gradients of small molecules 1, 2, and 5 (structural formulas are shown below) respectively, and incubate the cells for a certain period of time before and after transfection. The incubation conditions of small molecules 1 and 2 are: the cells are cultured in the medium containing the small molecules for more than 24 hours after transfection; the incubation conditions of small molecule 5 are: the cells are cultured in the medium containing the small molecules for less than 24 hours before transfection, and the cells are cultured in the medium containing the small molecules for more than 24 hours after transfection.
(3)转染后的48-96h进行细胞取样,根据供体DNA的序列,选择相应的方式,比如NGS、FACS、酶标仪、Agilent 2100Bioanalyzer System等,去检测3种小分子不同作用浓度下的敲入效率,从而确定三种小分子各自的作用浓度。(3) Cell samples are collected 48-96 hours after transfection. According to the sequence of the donor DNA, the corresponding method is selected, such as NGS, FACS, microplate reader, Agilent 2100 Bioanalyzer System, etc., to detect the knock-in efficiency of the three small molecules at different concentrations, thereby determining the effective concentrations of each of the three small molecules.
(4)最终在CRISPR/Cas9介导的基因组精准编辑的实验中,将确定作用浓度的小分子1或小分子2和小分子5联合使用,来更大程度的提高敲入效率。孵育条件同上述步骤(2)。(4) Finally, in the CRISPR/Cas9-mediated genome precision editing experiment, the small molecule 1 or small molecule 2 and small molecule 5 with the determined effective concentration are used together to further improve the knock-in efficiency. The incubation conditions are the same as those in the above step (2).
在一些实施方案中,在进行基因敲入操作时,包括让细胞与小分子1和小分子5接触。例如,在转染(如电穿孔转染,简称电转)后将细胞在含有小分子1和小分子5的培养基中培养一段时间;或者在转染前将细胞在含有小分子5的培养基中培养一段时间,并且在转染(如电转)后将细胞在含有小分子1和小分子5的培养基中培养一段时间;或者在转染前将细胞在含有小分子1和小分子5的培养基中培养一段时间,并且在转染(如电转)后将细胞在含有小分子1和小分子5的培养基中培养一段时间。优选地,在转染前将细胞在含有小分子5的培养基中培养一段时间,并且在转染(如电转)后将细胞在含有小分子1和小分子5的培养基中培养一段时间。In some embodiments, when performing a gene knock-in operation, the cells are contacted with small molecule 1 and small molecule 5. For example, after transfection (such as electroporation transfection, referred to as electrotransfection), the cells are cultured in a culture medium containing small molecule 1 and small molecule 5 for a period of time; or before transfection, the cells are cultured in a culture medium containing small molecule 5 for a period of time, and after transfection (such as electrotransfection), the cells are cultured in a culture medium containing small molecule 1 and small molecule 5 for a period of time; or before transfection, the cells are cultured in a culture medium containing small molecule 1 and small molecule 5 for a period of time, and after transfection (such as electrotransfection), the cells are cultured in a culture medium containing small molecule 1 and small molecule 5 for a period of time. Preferably, before transfection, the cells are cultured in a culture medium containing small molecule 5 for a period of time, and after transfection (such as electrotransfection), the cells are cultured in a culture medium containing small molecule 1 and small molecule 5 for a period of time.
在一些实施方案中,在进行基因敲入操作时,包括让细胞与小分子2和小分子5接触。例如,在转染(如电转)后将细胞在含有小分子2和小分子5的培养基中培养一段时间;或者在转染前将细胞在含有小分子5的培养基中培养一段时间,并且在转染(如电转)后将细胞在含有小分子2和小分子5的培养基中培养一段时间;或者在转染前将细胞在含有小分子2和小分子5的培养基中培养一段时间,并且在转染(如电转)后将细胞在含有小分子2和小分子5的培养基中培养一段时间。优选地,在转染前将细胞在含有小分子5的培养基中培养一段时间,并且在转染(如电转)后将细胞在含有小分子2和小分子5的培养基中培养一段时间。 In some embodiments, when performing a gene knock-in operation, the cell is contacted with small molecule 2 and small molecule 5. For example, after transfection (such as electroporation), the cell is cultured in a culture medium containing small molecule 2 and small molecule 5 for a period of time; or before transfection, the cell is cultured in a culture medium containing small molecule 5 for a period of time, and after transfection (such as electroporation), the cell is cultured in a culture medium containing small molecule 2 and small molecule 5 for a period of time; or before transfection, the cell is cultured in a culture medium containing small molecule 2 and small molecule 5 for a period of time, and after transfection (such as electroporation), the cell is cultured in a culture medium containing small molecule 2 and small molecule 5 for a period of time. Preferably, the cell is cultured in a culture medium containing small molecule 5 for a period of time before transfection, and the cell is cultured in a culture medium containing small molecule 2 and small molecule 5 for a period of time after transfection (such as electroporation).
在一些实施方案中,在进行基因敲入操作时,包括让细胞与小分子1、小分子2和小分子5接触。例如,在转染(如电转)后将细胞在含有小分子1、小分子2和小分子5的培养基中培养一段时间;或者在转染前将细胞在含有小分子5的培养基中培养一段时间,并且在转染(如电转)后将细胞在含有小分子1、小分子2和小分子5的培养基中培养一段时间;或者在转染前将细胞在含有小分子1、小分子2和小分子5的培养基中培养一段时间,并且在转染(如电转)后将细胞在含有小分子1、小分子2和小分子5的培养基中培养一段时间。优选地,在转染前将细胞在含有小分子5的培养基中培养一段时间,并且在转染(如电转)后将细胞在含有小分子1、小分子2和小分子5的培养基中培养一段时间。In some embodiments, when performing a gene knock-in operation, the cell is contacted with small molecule 1, small molecule 2, and small molecule 5. For example, after transfection (such as electroporation), the cell is cultured in a culture medium containing small molecule 1, small molecule 2, and small molecule 5 for a period of time; or before transfection, the cell is cultured in a culture medium containing small molecule 5 for a period of time, and after transfection (such as electroporation), the cell is cultured in a culture medium containing small molecule 1, small molecule 2, and small molecule 5 for a period of time; or before transfection, the cell is cultured in a culture medium containing small molecule 1, small molecule 2, and small molecule 5 for a period of time, and after transfection (such as electroporation), the cell is cultured in a culture medium containing small molecule 1, small molecule 2, and small molecule 5 for a period of time. Preferably, before transfection, the cell is cultured in a culture medium containing small molecule 5 for a period of time, and after transfection (such as electroporation), the cell is cultured in a culture medium containing small molecule 1, small molecule 2, and small molecule 5 for a period of time.
这里所用的“培养一段时间”指培养数分钟、数个小时,十几个小时,或者更长时间,例如,30min、60min、1h、2h、5h、10h、20h、24h、48h、72h,以及介于所列出时长之间的任何时长。优选地,“培养一段时间”指培养24-72h,例如24h。"Cultivating for a period of time" used herein refers to cultivating a few minutes, a few hours, more than ten hours, or longer time, for example, 30min, 60min, 1h, 2h, 5h, 10h, 20h, 24h, 48h, 72h, and any duration between the listed durations. Preferably, "cultivating for a period of time" refers to cultivating 24-72h, for example 24h.
在一些实施方案中,当培养基中含有小分子1时,其使用浓度不低于1μM,例如使得小分子1在培养基中的浓度为1-30μM,例如5-20μM。优选地,小分子1在培养基中的浓度为5μM。In some embodiments, when the medium contains small molecule 1, its concentration is not less than 1 μM, for example, so that the concentration of small molecule 1 in the medium is 1-30 μM, such as 5-20 μM. Preferably, the concentration of small molecule 1 in the medium is 5 μM.
在一些实施方案中,当培养基中含有小分子2时,其使用浓度不低于0.5μM,例如使得小分子2在培养基中的浓度为0.5-5μM,例如2-4μM。优选地,小分子2在培养基中的浓度为2μM。In some embodiments, when the medium contains small molecule 2, its concentration is not less than 0.5 μM, for example, so that the concentration of small molecule 2 in the medium is 0.5-5 μM, such as 2-4 μM. Preferably, the concentration of small molecule 2 in the medium is 2 μM.
在一些实施方案中,当培养基中含有小分子5时,其使用浓度不低于10μM,例如使得小分子5在培养基中的浓度为10-50μM,例如20-40μM。优选地,小分子5在培养基中的浓度为30μM。In some embodiments, when the medium contains small molecule 5, its concentration is not less than 10 μM, for example, so that the concentration of small molecule 5 in the medium is 10-50 μM, such as 20-40 μM. Preferably, the concentration of small molecule 5 in the medium is 30 μM.
本文还提供了用于提高CRISPR/Cas9介导的基因组敲入编辑效率的试剂盒,其中包括小分子1、小分子2、小分子5及其任意组合。在一些实施方案中,该试剂盒包括小分子1和小分子5。在另一些实施方案中,该试剂盒包括小分子2和小分子5。在另一些实施方案中,该试剂盒包括小分子1、小分子2和小分子5。在一些实施方案中,该试剂盒还任选地包括Cas9蛋白或其编码核酸分子。在一些实施方案中,该试剂盒还任选地包括向导RNA(gRNA)和/或用于进行同源重组修复(HDR)的供体模板。Also provided herein is a kit for improving the efficiency of CRISPR/Cas9-mediated genome knock-in editing, including small molecule 1, small molecule 2, small molecule 5, and any combination thereof. In some embodiments, the kit includes small molecule 1 and small molecule 5. In other embodiments, the kit includes small molecule 2 and small molecule 5. In other embodiments, the kit includes small molecule 1, small molecule 2, and small molecule 5. In some embodiments, the kit also optionally includes Cas9 protein or its encoding nucleic acid molecule. In some embodiments, the kit also optionally includes guide RNA (gRNA) and/or a donor template for homologous recombination repair (HDR).
本文通过将DNA依赖性蛋白激酶(DNA-PK)抑制剂和细胞分裂周期蛋白7(Cdc7)抑制剂联合使用,在不同细胞系、不同位点以及不同供体模板长度的情况下,可以更好的提高基因敲入效率,且优于某些商业化的小分子增强剂。In this paper, by combining DNA-dependent protein kinase (DNA-PK) inhibitors and cell division cycle protein 7 (Cdc7) inhibitors, the gene knock-in efficiency can be better improved in different cell lines, different sites and different donor template lengths, which is better than some commercial small molecule enhancers.
下面通过实施例,并结合附图,对本发明的技术方案作进一步详细的说明。除非另有说明,下文描述的实施例的方法和材料均为可以通过市场购买获得的常规产品。本发明所属领域技术员将会理解,下文描述的方法和材料,仅是示例性的,而不应视为限定本发明的范围。 The technical scheme of the present invention is further described in detail below by examples and in conjunction with the accompanying drawings. Unless otherwise specified, the methods and materials of the embodiments described below are conventional products that can be purchased from the market. It will be understood by those skilled in the art that the present invention belongs to that the methods and materials described below are only exemplary and should not be considered as limiting the scope of the present invention.
本发明的实施方式并不限于上述实施例所述,在不偏离本发明的精神和范围的情况下,本领域普通技术人员可以在形式和细节上对本发明做出各种改变和改进,而这些均被认为落入了本发明的保护范围。The implementation of the present invention is not limited to the above embodiments. Without departing from the spirit and scope of the present invention, ordinary technicians in this field can make various changes and improvements to the present invention in form and detail, and these are considered to fall within the protection scope of the present invention.
实施例1:Embodiment 1:
方案:plan:
根据相关文献,选择5种小分子(小分子的结构式如下所示),分别测试5种小分子的作用浓度范围。在HEK293T或者Jurkat细胞中,以绿色荧光蛋白(GFP)作为报告基因,用流式细胞仪进行检测,验证每种小分子不同作用浓度下RAB11A位点的敲入效率,从而确定每一种小分子的作用浓度。According to relevant literature, 5 small molecules were selected (the structural formula of the small molecules is shown below) and the concentration ranges of the 5 small molecules were tested respectively. In HEK293T or Jurkat cells, green fluorescent protein (GFP) was used as a reporter gene and flow cytometry was used to verify the knock-in efficiency of the RAB11A site at different concentrations of each small molecule, thereby determining the concentration of each small molecule.
方法:method:
1)细胞培养1) Cell culture
细胞每2-3天传代一次,且电转所需的细胞在电转前1-2天进行铺板,尽量确保电转当天细胞汇合度达到70-90%。Cells were passaged every 2-3 days, and cells required for electroporation were plated 1-2 days before electroporation, and the cell confluence was ensured to reach 70-90% on the day of electroporation.
2)电转2) Electroporation
2.1)向孔板中加入一定量的培养基(如表1所示)2.1) Add a certain amount of culture medium to the well plate (as shown in Table 1)
表1
Table 1
2.2)向孔板的培养基中加入不同量小分子(培养基中小分子终浓度如表2所示,小分子的结构式如以下所示),然后将孔板放到37℃培养箱中预热。2.2) Add different amounts of small molecules to the culture medium of the well plate (the final concentration of the small molecules in the culture medium is shown in Table 2, and the structural formula of the small molecules is shown below), and then place the well plate in a 37° C. incubator for preheating.
表2
Table 2
小分子结构式Small molecule structure
小分子1:
Small molecule 1:
小分子2:
Small molecule 2:
小分子3:
Small molecule 3:
小分子4:
Small molecule 4:
小分子5:
Small molecule 5:
2.3)RNP形成:将Cas9蛋白(GenScript,Z03469)、RAB11A-sgRNA(SEQ ID NO:1)(GenScript,EasyEdit)、RAB11A-dsDNA模板(SEQ ID NO:2)(GenScript)和缓冲液R(Thermo Fisher)按照表3所示的加入到一个新的、无RNase的离心管中,室温孵育5-15min形成RNP。2.3) RNP formation: Add Cas9 protein (GenScript, Z03469), RAB11A-sgRNA (SEQ ID NO: 1) (GenScript, EasyEdit), RAB11A-dsDNA template (SEQ ID NO: 2) (GenScript) and buffer R (Thermo Fisher) as shown in Table 3 into a new, RNase-free centrifuge tube and incubate at room temperature for 5-15 min to form RNP.
表3
table 3
2.4)电转体系:向上述有RNP的离心管中加入6μl用缓冲液R重悬的细胞(HEK293T:0.2M;Jurkat:0.5M),并用枪头小心吹吸混匀。2.4) Electroporation system: Add 6 μl of cells resuspended in buffer R (HEK293T: 0.2M; Jurkat: 0.5M) to the above centrifuge tube containing RNPs, and mix by carefully pipetting with a pipette.
2.5)电转:打开Neon电转仪(Thermo Fisher),设置电转参数(HEK293T细胞:Voltage:1200V,Width:10ms,Pulse:3pulses;Jurkat细胞:Voltage:1600V,Width:10ms,Pulse:3pulses)。2.5) Electroporation: Turn on the Neon electroporator (Thermo Fisher) and set the electroporation parameters (HEK293T cells: Voltage: 1200 V, Width: 10 ms, Pulse: 3 pulses; Jurkat cells: Voltage: 1600 V, Width: 10 ms, Pulse: 3 pulses).
2.6)用10μl Neon电转枪头(Thermo Fisher)吸取离心管中的细胞和RNP混合物,注意枪头中一定不能有气泡。然后将Neon电转枪头插入到加了3ml缓冲液E(Thermo Fisher)的Neon电转管中,点击Neon电转仪屏幕上的“Start键”。2.6) Use a 10μl Neon electroporation pipette tip (Thermo Fisher) to aspirate the cell and RNP mixture in the centrifuge tube. Be careful not to have bubbles in the tip. Then insert the Neon electroporation pipette tip into the Neon electroporation tube with 3ml buffer E (Thermo Fisher) added, and click the "Start" button on the Neon electroporation instrument screen.
2.7)将电转样品立即转移到上述含有不同小分子浓度的孔板的培养基中,电转结束后将孔板放入37℃/5%CO2培养箱中继续培养。2.7) Immediately transfer the electroporated samples to the culture medium of the well plate containing different small molecule concentrations. After the electroporation, place the well plate in a 37°C/5% CO2 incubator for further culture.
3)小分子作用方式3) Small molecule mode of action
Neon电转后的72h内,细胞在含有小分子的培养基中培养;72h之后,将细胞从含有小分子的培养基中转换到不含有小分子的培养基中。Within 72 hours after Neon electroporation, the cells were cultured in a medium containing small molecules; after 72 hours, the cells were switched from a medium containing small molecules to a medium without small molecules.
4)敲入效率的检测方法:用流式细胞仪(Beckman)检测电转后3天时的细胞活率,以及电转后7天时的GFP+细胞百分比(即敲入效率)。4) Method for detecting knock-in efficiency: Flow cytometry (Beckman) was used to detect the cell viability 3 days after electroporation and the percentage of GFP+ cells (i.e., knock-in efficiency) 7 days after electroporation.
实验结论:Experimental results:
如图1-5所示,小分子1、小分子2、小分子5对于基因敲入效率有较明显的提高,且其有效作用浓度为:小分子1:≥1μM;小分子2:≥0.5μM;小分子5:≥10μM。且这3种小分子在有效作用浓度内,对细胞活率无明显影响。 As shown in Figures 1-5, small molecules 1, 2, and 5 significantly improved the gene knock-in efficiency, and their effective concentrations were: small molecule 1: ≥1μM; small molecule 2: ≥0.5μM; small molecule 5: ≥10μM. Moreover, these three small molecules had no significant effect on cell viability within the effective concentration.
实施例2:Embodiment 2:
方案:plan:
在Jurkat细胞中,以绿色荧光蛋白(GFP)作为报告基因,用流式细胞仪检测RAB11A位点的敲入效率,优化小分子5的孵育条件。In Jurkat cells, green fluorescent protein (GFP) was used as a reporter gene, and the knock-in efficiency of the RAB11A locus was detected by flow cytometry to optimize the incubation conditions of small molecule 5.
方法:method:
1)小分子5的不同孵育方式如图6所示。1) Different incubation methods of small molecule 5 are shown in FIG6 .
2)细胞培养2) Cell culture
细胞每2-3天传代一次,且电转所需的细胞在电转前1天进行铺板。细胞在进行电转前铺板时,需要准备两份细胞,其中一份细胞不加小分子5进行培养,另外一份细胞在电转前24h向其中加入终浓度为30μM的小分子5。The cells were passaged every 2-3 days, and the cells required for electroporation were plated 1 day before electroporation. When cells were plated before electroporation, two cells were prepared, one of which was cultured without small molecule 5, and the other was cultured with small molecule 5 added to a final concentration of 30 μM 24 hours before electroporation.
3)电转3) Electroporation
3.1)向24孔板中加入0.5mL培养基(含有10%FBS的RPMI 1640)。3.1) Add 0.5 mL of culture medium (RPMI 1640 containing 10% FBS) to a 24-well plate.
3.2)根据图6小分子的作用浓度和方式,向孔板的部分孔的培养基中加入终浓度为30μM的小分子5,然后将孔板放到37℃培养箱中预热。3.2) According to the concentration and mode of action of the small molecule in FIG6 , small molecule 5 with a final concentration of 30 μM was added to the culture medium of some wells of the well plate, and then the well plate was placed in a 37° C. incubator for preheating.
3.3)RNP形成:将eSpCas9蛋白(GenScript,Z03470-300)、RAB11A-sgRNA(SEQ ID NO:1)(GenScript,EasyEdit)、RAB11A-dsDNA模板(SEQ ID NO:2)(GenScript)和缓冲液R(Thermo Fisher)按照表3(Jurkat细胞)所示的加入到一个新的、无RNase的离心管中,室温孵育5-15min形成RNP。3.3) RNP formation: eSpCas9 protein (GenScript, Z03470-300), RAB11A-sgRNA (SEQ ID NO: 1) (GenScript, EasyEdit), RAB11A-dsDNA template (SEQ ID NO: 2) (GenScript) and buffer R (Thermo Fisher) were added to a new, RNase-free centrifuge tube as shown in Table 3 (Jurkat cells) and incubated at room temperature for 5-15 min to form RNP.
3.4)电转体系:向上述有RNP的离心管中加入6μl用缓冲液R重悬的0.5M的Jurkat细胞(没加小分子5的样品组用的是步骤2)中未用小分子5提前处理的细胞,加小分子5的样品组用的是步骤2)中30μM小分子5提前处理的细胞),并用枪头小心吹吸混匀。3.4) Electroporation system: Add 6 μl of 0.5 M Jurkat cells resuspended in buffer R to the above centrifuge tube containing RNP (the sample group without small molecule 5 uses cells that have not been pre-treated with small molecule 5 in step 2), and the sample group with small molecule 5 uses cells that have been pre-treated with 30 μM small molecule 5 in step 2)), and mix carefully by pipetting with a pipette tip.
3.5)电转:打开Neon电转仪(Thermo Fisher),设置电转参数:Voltage:1700V,Width:20ms,Pulse:1pulses。3.5) Electroporation: Turn on the Neon electroporator (Thermo Fisher) and set the electroporation parameters: Voltage: 1700 V, Width: 20 ms, Pulse: 1 pulses.
3.6)用10μl Neon电转枪头(Thermo Fisher)吸取离心管中的细胞和RNP混合物,注意枪头中一定不能有气泡。然后将Neon电转枪头插入到加了3ml缓冲液E(Thermo Fisher)的Neon电转管中,点击Neon电转仪屏幕上的“Start键”。3.6) Use a 10μl Neon electroporation pipette tip (Thermo Fisher) to aspirate the cell and RNP mixture in the centrifuge tube. Be careful not to have bubbles in the tip. Then insert the Neon electroporation pipette tip into the Neon electroporation tube with 3ml buffer E (Thermo Fisher) added, and click the "Start" button on the Neon electroporation instrument screen.
3.7)将电转样品立即转移到相应含小分子的培养基孔中,电转结束后将孔板放入37℃/5%CO2培养箱中继续培养24h后,再将细胞转移至不含小分子的培养基中培养。3.7) Immediately transfer the electroporated samples to the corresponding medium wells containing small molecules. After electroporation, place the well plate in a 37°C/5% CO2 incubator for further culturing for 24 hours, and then transfer the cells to medium without small molecules for culturing.
4)敲入效率的检测方法:用流式细胞仪(Beckman)检测电转后3天时的细胞活率,以及电转后6天时的GFP+细胞百分比(即敲入效率)(如图7所示)。4) Method for detecting knock-in efficiency: Flow cytometry (Beckman) was used to detect the cell viability 3 days after electroporation and the percentage of GFP+ cells (ie, knock-in efficiency) 6 days after electroporation (as shown in FIG. 7 ).
实验结论:Experimental results:
如图7所示,在对细胞活率无明显影响的前提下,小分子5的最优孵育条件为孵育方式③:电转前细胞在含有小分子5的培养基中培养24h以内;电转后细胞在含有小分 子5的培养基中培养24h,24h之后,将细胞从含有小分子5的培养基中转换到不含有小分子5的培养基中。As shown in Figure 7, under the premise of no significant effect on cell viability, the optimal incubation condition for small molecule 5 is incubation method ③: cells are cultured in the medium containing small molecule 5 for less than 24 h before electroporation; cells are cultured in the medium containing small molecule 5 after electroporation. The cells were cultured in a medium containing small molecule 5 for 24 h. After 24 h, the cells were switched from a medium containing small molecule 5 to a medium not containing small molecule 5.
实施例3:Embodiment 3:
方案:plan:
在Jurkat细胞中,以绿色荧光蛋白(GFP)作为报告基因,将5种小分子组合使用,用流式细胞仪检测RAB11A位点的敲入效率,确认较优的小分子组合方式。In Jurkat cells, five small molecules were used in combination using green fluorescent protein (GFP) as a reporter gene, and the knock-in efficiency of the RAB11A site was detected by flow cytometry to confirm the optimal small molecule combination.
方法:method:
1)小分子的作用浓度以及不同组合方式(如图8所示),其中“+”表示添加,“-”表示不添加。1) The concentration of small molecules and different combinations (as shown in FIG8 ), where “+” indicates addition and “-” indicates no addition.
2)小分子孵育时间2) Small molecule incubation time
小分子1、小分子2、小分子3、小分子4:电转后细胞在含有小分子的培养基中培养24h;24h之后,将细胞从含有小分子的培养基中转换到不含有小分子的培养基中。Small molecule 1, small molecule 2, small molecule 3, small molecule 4: after electroporation, the cells were cultured in a medium containing small molecules for 24 hours; after 24 hours, the cells were switched from the medium containing small molecules to a medium not containing small molecules.
小分子5:电转前细胞在含有小分子的培养基中培养24h以内;电转后细胞在含有小分子的培养基中培养24h,24h之后,将细胞从含有小分子的培养基中转换到不含有小分子的培养基中。Small molecule 5: Before electroporation, the cells are cultured in a medium containing small molecules for less than 24 hours; after electroporation, the cells are cultured in a medium containing small molecules for 24 hours. After 24 hours, the cells are switched from the medium containing small molecules to a medium without small molecules.
3)细胞培养3) Cell culture
细胞每2-3天传代一次,且电转所需的细胞在电转前1天进行铺板。细胞在进行电转前铺板时,需要准备两份细胞,其中一份细胞不加小分子进行培养,另外一份细胞在电转前24h内向其中加入终浓度为30μM的小分子5。The cells were passaged every 2-3 days, and the cells required for electroporation were plated 1 day before electroporation. When cells were plated before electroporation, two aliquots of cells were prepared, one of which was cultured without adding small molecules, and the other aliquot of cells was added with small molecules at a final concentration of 30 μM within 24 hours before electroporation.
4)电转4) Electroporation
4.1)向24孔板中加入0.5mL培养基(含有10%FBS的RPMI 1640)。4.1) Add 0.5 mL of culture medium (RPMI 1640 containing 10% FBS) to a 24-well plate.
4.2)根据小分子的作用浓度以及不同组合方式,向孔板的部分孔的培养基中分别加入不同的小分子,然后将孔板放到37℃培养箱中预热。4.2) According to the concentration of small molecules and different combinations, different small molecules are added to the culture medium of some wells of the well plate, and then the well plate is placed in a 37° C. incubator for preheating.
4.3)RNP形成:将eSpCas9蛋白(GenScript,Z03470-300)、RAB11A-sgRNA(SEQ ID NO:1)(GenScript,EasyEdit)、RAB11A-dsDNA模板(SEQ ID NO:2)(GenScript)和缓冲液R(Thermo Fisher)按照表3(Jurkat细胞)所示的加入到一个新的、无RNase的离心管中,室温孵育5-15min形成RNP。4.3) RNP formation: eSpCas9 protein (GenScript, Z03470-300), RAB11A-sgRNA (SEQ ID NO: 1) (GenScript, EasyEdit), RAB11A-dsDNA template (SEQ ID NO: 2) (GenScript) and buffer R (Thermo Fisher) were added to a new, RNase-free centrifuge tube as shown in Table 3 (Jurkat cells) and incubated at room temperature for 5-15 min to form RNP.
4.4)电转体系:向上述有RNP的离心管中加入6μl用缓冲液R重悬的0.5M的Jurkat细胞(没加小分子5的样品组用的是未用小分子5提前处理的细胞,加小分子5的样品组用的是30μM小分子5提前处理的细胞),并用枪头小心吹吸混匀。4.4) Electroporation system: Add 6 μl of 0.5 M Jurkat cells resuspended in buffer R to the above centrifuge tube containing RNP (the sample group without small molecule 5 used cells not pre-treated with small molecule 5, and the sample group with small molecule 5 used cells pre-treated with 30 μM small molecule 5), and mix by carefully pipetting with a pipette tip.
4.5)电转:打开Neon电转仪(Thermo Fisher),设置电转参数:Voltage:1700V,Width:20ms,Pulse:1pulses。 4.5) Electroporation: Turn on the Neon electroporator (Thermo Fisher) and set the electroporation parameters: Voltage: 1700 V, Width: 20 ms, Pulse: 1 pulses.
4.6)用10μl Neon电转枪头(Thermo Fisher)吸取离心管中的细胞和RNP混合物,注意枪头中一定不能有气泡。然后将Neon电转枪头插入到加了3ml缓冲液E(Thermo Fisher)的Neon电转管中,点击Neon电转仪屏幕上的“Start键”。4.6) Use a 10μl Neon electroporation pipette tip (Thermo Fisher) to aspirate the cell and RNP mixture in the centrifuge tube. Be careful not to have bubbles in the tip. Then insert the Neon electroporation pipette tip into the Neon electroporation tube with 3ml buffer E (Thermo Fisher) added, and click the "Start" button on the Neon electroporation instrument screen.
4.7)将电转样品立即转移到相应含小分子的培养基孔中,电转结束后将孔板放入37℃/5%CO2培养箱中继续培养24h,再将细胞转移至不含小分子的培养基中培养。4.7) Immediately transfer the electroporated samples to the corresponding medium wells containing small molecules. After electroporation, place the well plate in a 37°C/5% CO2 incubator for further 24 hours, and then transfer the cells to medium without small molecules for culturing.
5)敲入效率的检测方法:用流式细胞仪(Beckman)检测电转后4天时的细胞活率(如图9所示),以及电转后7天时的GFP+细胞百分比(即敲入效率,如图10所示)。5) Method for detecting knock-in efficiency: Flow cytometry (Beckman) was used to detect the cell viability 4 days after electroporation (as shown in FIG9 ), and the percentage of GFP+ cells 7 days after electroporation (i.e., knock-in efficiency, as shown in FIG10 ).
实验结论:Experimental results:
如图10所示,相比于单独使用小分子1,小分子1和小分子5联合使用,敲入效率提高了20.2%;相比于单独使用小分子2,小分子2和小分子5联合使用,敲入效率提高了22.9%。As shown in FIG10 , compared with the use of small molecule 1 alone, the knock-in efficiency was increased by 20.2% when small molecule 1 and small molecule 5 were used in combination; compared with the use of small molecule 2 alone, the knock-in efficiency was increased by 22.9% when small molecule 2 and small molecule 5 were used in combination.
小分子1和小分子5联合使用,以及小分子2和小分子5联合使用,能够在保证细胞活率的基础上最大程度的提高敲入效率。The combined use of small molecule 1 and small molecule 5, as well as the combined use of small molecule 2 and small molecule 5, can maximize the knock-in efficiency while ensuring cell viability.
实施例4:Embodiment 4:
方案:plan:
在Jurkat细胞中,以绿色荧光蛋白(GFP)作为报告基因,用流式细胞仪检测RAB11A位点的敲入效率,进一步验证实施例3中的两种最优小分子组合有较广的适用浓度范围。In Jurkat cells, green fluorescent protein (GFP) was used as a reporter gene, and flow cytometry was used to detect the knock-in efficiency of the RAB11A site, further verifying that the two optimal small molecule combinations in Example 3 had a wider applicable concentration range.
方法:method:
1)小分子的作用浓度范围如图11所示,其中“+”表示添加,“-”表示不添加。1) The effective concentration range of small molecules is shown in FIG11 , where “+” indicates addition and “-” indicates no addition.
2)小分子孵育时间2) Small molecule incubation time
小分子1、小分子2:电转后细胞在含有小分子的培养基中培养24h;24h之后,将细胞从含有小分子的培养基中转换到不含有小分子的培养基中。Small molecule 1 and small molecule 2: After electroporation, the cells were cultured in a medium containing small molecules for 24 hours; after 24 hours, the cells were switched from the medium containing small molecules to a medium not containing small molecules.
小分子5:电转前细胞在含有小分子的培养基中培养24h以内;电转后细胞在含有小分子的培养基中培养24h,24h之后,将细胞从含有小分子的培养基中转换到不含有小分子的培养基中。Small molecule 5: Before electroporation, the cells are cultured in a medium containing small molecules for less than 24 hours; after electroporation, the cells are cultured in a medium containing small molecules for 24 hours. After 24 hours, the cells are switched from the medium containing small molecules to a medium without small molecules.
3)细胞培养3) Cell culture
细胞每2-3天传代一次,且电转所需的细胞在电转前1天进行铺板。细胞在进行电转前铺板时,需要准备四份细胞,其中一份细胞不加小分子进行培养,另外三份细胞在电转前24h内分别向其中加入终浓度为20μM、30μM、40μM的小分子5。The cells were passaged every 2-3 days, and the cells required for electroporation were plated one day before electroporation. When cells were plated before electroporation, four cells were prepared, one of which was cultured without small molecules, and the other three were added with small molecules at final concentrations of 20μM, 30μM, and 40μM within 24h before electroporation.
4)电转4) Electroporation
4.1)向24孔板中加入0.5mL培养基(含有10%FBS的RPMI 1640)。4.1) Add 0.5 mL of culture medium (RPMI 1640 containing 10% FBS) to a 24-well plate.
4.2)根据小分子的作用浓度以及不同组合方式,向孔板的部分孔的培养基中分别加入不同的小分子,然后将孔板放到37℃培养箱中预热。 4.2) According to the concentration of small molecules and different combinations, different small molecules are added to the culture medium of some wells of the well plate, and then the well plate is placed in a 37° C. incubator for preheating.
4.3)RNP形成:将eSpCas9蛋白(GenScript,Z03470-300)、RAB11A-sgRNA(SEQ ID NO:1)(GenScript,EasyEdit)、RAB11A-dsDNA模板(SEQ ID NO:2)(GenScript)和缓冲液R(Thermo Fisher)按照表3(Jurkat细胞)所示的加入到一个新的、无RNase的离心管中,室温孵育5-15min形成RNP。4.3) RNP formation: eSpCas9 protein (GenScript, Z03470-300), RAB11A-sgRNA (SEQ ID NO: 1) (GenScript, EasyEdit), RAB11A-dsDNA template (SEQ ID NO: 2) (GenScript) and buffer R (Thermo Fisher) were added to a new, RNase-free centrifuge tube as shown in Table 3 (Jurkat cells) and incubated at room temperature for 5-15 min to form RNP.
4.4)电转体系:向上述有RNP的离心管中加入6μl用缓冲液R重悬的0.5M的Jurkat细胞(没加小分子5的样品组用的是未用小分子5提前处理的细胞,加20μM小分子5的样品组用的是20μM小分子5提前处理的细胞,加30μM小分子5的样品组用的是30μM小分子5提前处理的细胞,加40μM小分子5的样品组用的是40μM小分子5提前处理的细胞),并用枪头小心吹吸混匀。4.4) Electroporation system: Add 6 μl of 0.5 M Jurkat cells resuspended in buffer R to the above centrifuge tube containing RNP (the sample group without small molecule 5 used cells not pre-treated with small molecule 5, the sample group with 20 μM small molecule 5 used cells pre-treated with 20 μM small molecule 5, the sample group with 30 μM small molecule 5 used cells pre-treated with 30 μM small molecule 5, and the sample group with 40 μM small molecule 5 used cells pre-treated with 40 μM small molecule 5), and mix by carefully pipetting with a pipette tip.
4.5)电转:打开Neon电转仪(Thermo Fisher),设置电转参数:Voltage:1600V,Width:10ms,Pulse:3pulses。4.5) Electroporation: Turn on the Neon electroporator (Thermo Fisher) and set the electroporation parameters: Voltage: 1600 V, Width: 10 ms, Pulse: 3 pulses.
4.6)用10μl Neon电转枪头(Thermo Fisher)吸取离心管中的细胞和RNP混合物,注意枪头中一定不能有气泡。然后将Neon电转枪头插入到加了3ml缓冲液E(Thermo Fisher)的Neon电转管中,点击Neon电转仪屏幕上的“Start键”。4.6) Use a 10μl Neon electroporation pipette tip (Thermo Fisher) to aspirate the cell and RNP mixture in the centrifuge tube. Be careful not to have bubbles in the tip. Then insert the Neon electroporation pipette tip into the Neon electroporation tube with 3ml buffer E (Thermo Fisher) added, and click the "Start" button on the Neon electroporation instrument screen.
4.7)将电转样品立即转移到相应含小分子的培养基孔中,电转结束后将孔板放入37℃/5%CO2培养箱中继续培养24h,再将细胞转移至不含小分子的培养基中培养。4.7) Immediately transfer the electroporated samples to the corresponding medium wells containing small molecules. After electroporation, place the well plate in a 37°C/5% CO2 incubator for further culturing for 24 hours, and then transfer the cells to medium without small molecules for culturing.
5)敲入效率的检测方法:用流式细胞仪(Beckman)检测电转后7天时的GFP+细胞百分比(即敲入效率,如图12所示)。5) Method for detecting knock-in efficiency: Flow cytometer (Beckman) was used to detect the percentage of GFP+ cells 7 days after electroporation (ie, knock-in efficiency, as shown in FIG. 12 ).
实验结论Experimental results
如图12所示,相比于单独使用5μM小分子1,5μM小分子1和小分子5(20-40μM)联合使用,敲入效率提高了31.0%;相比于单独使用20μM小分子1,20μM小分子1和小分子5(20-40μM)联合使用,敲入效率提高了21.3%。As shown in Figure 12, compared with the use of 5μM small molecule 1 alone, the combined use of 5μM small molecule 1 and small molecule 5 (20-40μM) increased the knock-in efficiency by 31.0%; compared with the use of 20μM small molecule 1 alone, the combined use of 20μM small molecule 1 and small molecule 5 (20-40μM) increased the knock-in efficiency by 21.3%.
相比于单独使用2μM小分子2,2μM小分子2和小分子5(20-40μM)联合使用,敲入效率提高了33.4%;相比于单独使用4μM小分子2,4μM小分子2和小分子5(20-40μM)联合使用,敲入效率提高了21.5%。Compared with the use of 2μM small molecule 2 alone, the combined use of 2μM small molecule 2 and small molecule 5 (20-40μM) increased the knock-in efficiency by 33.4%; compared with the use of 4μM small molecule 2 alone, the combined use of 4μM small molecule 2 and small molecule 5 (20-40μM) increased the knock-in efficiency by 21.5%.
小分子1和小分子5联合使用,以及小分子2和小分子5联合使用,这两种最优的小分子组合有较广的适用浓度范围。The combination of small molecule 1 and small molecule 5, and the combination of small molecule 2 and small molecule 5, these two optimal small molecule combinations have a wider applicable concentration range.
实施例5:Embodiment 5:
方案:plan:
在SJCRH30细胞中,以绿色荧光蛋白(GFP)作为报告基因,用流式细胞仪检测RAB11A位点的敲入效率,使用dsDNA和ssDNA这两种类型的模板,进一步验证这两种小分子组合有较广的适用性。In SJCRH30 cells, green fluorescent protein (GFP) was used as a reporter gene, and flow cytometry was used to detect the knock-in efficiency of the RAB11A site. Two types of templates, dsDNA and ssDNA, were used to further verify that the combination of these two small molecules had a wide range of applicability.
方法:method:
1)小分子作用方案 1) Small molecule action plan
①无小分子;②5μM小分子1;③2μM小分子2;④30μM小分子5;⑤5μM小分子1+30μM小分子5;⑥2μM小分子2+30μM小分子5。① No small molecule; ② 5μM small molecule 1; ③ 2μM small molecule 2; ④ 30μM small molecule 5; ⑤ 5μM small molecule 1 + 30μM small molecule 5; ⑥ 2μM small molecule 2 + 30μM small molecule 5.
2)小分子孵育时间2) Small molecule incubation time
小分子1、小分子2:电转后细胞在含有小分子的培养基中培养24h;24h之后,将细胞从含有小分子的培养基中转换到不含有小分子的培养基中。Small molecule 1 and small molecule 2: After electroporation, the cells were cultured in a medium containing small molecules for 24 hours; after 24 hours, the cells were switched from the medium containing small molecules to a medium not containing small molecules.
小分子5:电转前细胞在含有小分子的培养基中培养24h以内;电转后细胞在含有小分子的培养基中培养24h,24h之后,将细胞从含有小分子的培养基中转换到不含有小分子的培养基中。Small molecule 5: Before electroporation, the cells are cultured in a medium containing small molecules for less than 24 hours; after electroporation, the cells are cultured in a medium containing small molecules for 24 hours. After 24 hours, the cells are switched from the medium containing small molecules to a medium without small molecules.
3)细胞培养3) Cell culture
细胞每2-3天传代一次,且电转所需的细胞在电转前1天进行铺板。细胞在进行电转前铺板时,需要准备两份细胞,其中一份细胞不加小分子进行培养,另外一份细胞在电转前24h内向其中加入终浓度为30μM的小分子5。The cells were passaged every 2-3 days, and the cells required for electroporation were plated 1 day before electroporation. When cells were plated before electroporation, two aliquots of cells were prepared, one of which was cultured without adding small molecules, and the other aliquot of cells was added with small molecules at a final concentration of 30 μM within 24 hours before electroporation.
4)电转4) Electroporation
4.1)向12孔板中加入1mL培养基(含有10%FBS的RPMI 1640)。4.1) Add 1 mL of culture medium (RPMI 1640 containing 10% FBS) to a 12-well plate.
4.2)根据小分子的作用浓度以及不同组合方式,向孔板的部分孔的培养基中分别加入不同的小分子,然后将孔板放到37℃培养箱中预热。4.2) According to the concentration of small molecules and different combinations, different small molecules are added to the culture medium of some wells of the well plate, and then the well plate is placed in a 37° C. incubator for preheating.
4.3)RNP形成:将eSpCas9蛋白(GenScript,Z03470-300)、RAB11A-sgRNA(SEQ ID NO:1)(GenScript,SafeEdit)、RAB11A-dsDNA模板或者RAB11A-ssDNA模板(SEQ ID NO:2)(GenScript)和缓冲液R(Thermo Fisher)按照表4所示的加入到一个新的、无RNase的离心管中,室温孵育5-15min形成RNP。4.3) RNP formation: eSpCas9 protein (GenScript, Z03470-300), RAB11A-sgRNA (SEQ ID NO: 1) (GenScript, SafeEdit), RAB11A-dsDNA template or RAB11A-ssDNA template (SEQ ID NO: 2) (GenScript) and buffer R (Thermo Fisher) were added to a new, RNase-free centrifuge tube as shown in Table 4 and incubated at room temperature for 5-15 min to form RNP.
表4
Table 4
4.4)电转体系:向上述有RNP的离心管中加入6μl用缓冲液R重悬的0.2M的SJCRH30细胞(没加小分子5的样品组用的是未用小分子5提前处理的细胞,加小分子5的样品组用的是30μM小分子5提前处理的细胞),并用枪头小心吹吸混匀。4.4) Electroporation system: Add 6 μl of 0.2 M SJCRH30 cells resuspended in buffer R to the above centrifuge tube containing RNP (the sample group without small molecule 5 used cells not pre-treated with small molecule 5, and the sample group with small molecule 5 used cells pre-treated with 30 μM small molecule 5), and mix by carefully pipetting with a pipette tip.
4.5)电转:打开Neon电转仪(Thermo Fisher),设置电转参数:Voltage:1200V,Width:20ms,Pulse:3pulses。4.5) Electroporation: Turn on the Neon electroporator (Thermo Fisher) and set the electroporation parameters: Voltage: 1200 V, Width: 20 ms, Pulse: 3 pulses.
4.6)用10μl Neon电转枪头(Thermo Fisher)吸取离心管中的细胞和RNP混合物,注意枪头中一定不能有气泡。然后将Neon电转枪头插入到加了3ml缓冲液E(Thermo Fisher)的Neon电转管中,点击Neon电转仪屏幕上的“Start键”。4.6) Use a 10μl Neon electroporation pipette tip (Thermo Fisher) to aspirate the cell and RNP mixture in the centrifuge tube. Be careful not to have bubbles in the tip. Then insert the Neon electroporation pipette tip into the Neon electroporation tube with 3ml buffer E (Thermo Fisher) added, and click the "Start" button on the Neon electroporation instrument screen.
4.7)将电转样品立即转移到相应含小分子孔的培养基中,电转结束后将孔板放入37℃/5%CO2培养箱中继续培养24h,再将细胞转移至不含小分子的培养基中培养。 4.7) Immediately transfer the electroporated samples to the culture medium of the corresponding wells containing small molecules. After electroporation, place the well plate in a 37°C/5% CO2 incubator for further 24 hours, and then transfer the cells to the culture medium without small molecules for culturing.
5)敲入效率的检测方法:用流式细胞仪(Beckman)检测电转后7天时的GFP+细胞百分比(即敲入效率,如图13所示)。5) Method for detecting knock-in efficiency: Flow cytometer (Beckman) was used to detect the percentage of GFP+ cells 7 days after electroporation (ie, knock-in efficiency, as shown in FIG. 13 ).
实验结论Experimental results
如图13所示,对于dsDNA模板体系,相比于单独使用小分子1或2,小分子1或2和小分子5联合使用,敲入效率分别提高了9.85%、10.9%;对于ssDNA模板体系,相比于单独使用小分子1或2,小分子1或2和小分子5联合使用,敲入效率分别提高了23.2%、21.4%。As shown in Figure 13, for the dsDNA template system, compared with using small molecule 1 or 2 alone, the knock-in efficiency was increased by 9.85% and 10.9% when small molecule 1 or 2 and small molecule 5 were used in combination, respectively; for the ssDNA template system, compared with using small molecule 1 or 2 alone, the knock-in efficiency was increased by 23.2% and 21.4% when small molecule 1 or 2 and small molecule 5 were used in combination, respectively.
在SJCRH30细胞中的RAB11A位点,小分子1或小分子2和小分子5联合使用,相比于不添加小分子,能够将敲入效率提高了68.7%(dsDNA模板)和46%(ssDNA模板)。At the RAB11A site in SJCRH30 cells, the combined use of small molecule 1 or small molecule 2 and small molecule 5 can increase the knock-in efficiency by 68.7% (dsDNA template) and 46% (ssDNA template) compared to not adding small molecules.
实施例6:Embodiment 6:
方案:plan:
在T细胞中,以绿色荧光蛋白(GFP)作为报告基因,用流式细胞仪检测TRAC位点的敲入效率,使用dsDNA和质粒这两种类型的模板,进一步验证这两种小分子组合有较广的适用性。In T cells, green fluorescent protein (GFP) was used as a reporter gene, and flow cytometry was used to detect the knock-in efficiency of the TRAC site. Two types of templates, dsDNA and plasmid, were used to further verify that the combination of these two small molecules had a wide range of applicability.
方法:method:
1)小分子作用方案1) Small molecule action plan
①无小分子;②5μM小分子1+30μM小分子5;③2μM小分子2+30μM小分子5。① No small molecule; ② 5μM small molecule 1 + 30μM small molecule 5; ③ 2μM small molecule 2 + 30μM small molecule 5.
2)小分子孵育时间2) Small molecule incubation time
小分子1、小分子2:电转后细胞在含有小分子的培养基中培养24h;24h之后,将细胞从含有小分子的培养基中转换到不含有小分子的培养基中。Small molecule 1 and small molecule 2: After electroporation, the cells were cultured in a medium containing small molecules for 24 hours; after 24 hours, the cells were switched from the medium containing small molecules to a medium not containing small molecules.
小分子5:电转前细胞在含有小分子的培养基中培养24h以内;电转后细胞在含有小分子的培养基中培养24h,24h之后,将细胞从含有小分子的培养基中转换到不含有小分子的培养基中。Small molecule 5: Before electroporation, the cells are cultured in a medium containing small molecules for less than 24 hours; after electroporation, the cells are cultured in a medium containing small molecules for 24 hours. After 24 hours, the cells are switched from the medium containing small molecules to a medium without small molecules.
3)细胞的分离纯化、激活和培养3) Cell isolation, purification, activation and culture
从100M PBMC(AllCells)中分离纯化T细胞(Thermo Fisher),纯化后的T细胞用T细胞激活试剂盒(Thermo Fisher)进行激活,激活后的T细胞在含有IL2、IL7、IL15的TexMACSTMMedium(Miltenyi Biotec)中进行培养。T cells were isolated and purified from 100M PBMC (AllCells) (Thermo Fisher), and the purified T cells were activated using a T cell activation kit (Thermo Fisher). The activated T cells were cultured in TexMACS Medium (Miltenyi Biotec) containing IL2, IL7, and IL15.
需要准备两份细胞,其中一份细胞不加小分子进行培养,另外一份细胞在电转前24h内向其中加入终浓度为30μM的小分子5。Two cells need to be prepared, one of which is cultured without adding small molecules, and the other is cultured with small molecule 5 added to it at a final concentration of 30 μM within 24 hours before electroporation.
4)电转4) Electroporation
4.1)向48孔板中加入700μL培养基(含有5%FBS、IL2、IL7、IL15的TexMACS)。 4.1) Add 700 μL of culture medium (TexMACS containing 5% FBS, IL2, IL7, IL15) to a 48-well plate.
4.2)根据小分子的作用浓度以及不同组合方式,向孔板的部分孔的培养基中分别加入不同的小分子,然后将孔板放到37℃培养箱中预热。4.2) According to the concentration of small molecules and different combinations, different small molecules are added to the culture medium of some wells of the well plate, and then the well plate is placed in a 37° C. incubator for preheating.
4.3)RNP形成:将TrueCut HiFi Cas9蛋白(Thermo Fisher)、TRAC-sgRNA(SEQ ID NO:3)(GenScript,SafeEdit)、TRAC-dsDNA模板(SEQ ID NO:8)或者TRAC质粒模板(SEQ ID NO:9)(GenScript)和缓冲液R(Thermo Fisher)按照表5所示的加入到一个新的、无RNase的离心管中,室温孵育5-15min形成RNP。4.3) RNP formation: Add TrueCut HiFi Cas9 protein (Thermo Fisher), TRAC-sgRNA (SEQ ID NO: 3) (GenScript, SafeEdit), TRAC-dsDNA template (SEQ ID NO: 8) or TRAC plasmid template (SEQ ID NO: 9) (GenScript) and buffer R (Thermo Fisher) as shown in Table 5 into a new, RNase-free centrifuge tube and incubate at room temperature for 5-15 min to form RNP.
表5
table 5
4.4)电转体系:向上述有RNP的离心管中加入6μl用缓冲液R重悬的0.5M的T细胞(没加小分子5的样品组用的是未用小分子5提前处理的细胞,加小分子5的样品组用的是30μM小分子5提前处理的细胞),并用枪头小心吹吸混匀。4.4) Electroporation system: Add 6 μl of 0.5M T cells resuspended in buffer R to the above centrifuge tube containing RNP (the sample group without small molecule 5 uses cells not pre-treated with small molecule 5, and the sample group with small molecule 5 uses cells pre-treated with 30 μM small molecule 5), and mix by carefully pipetting with a pipette tip.
4.5)电转:打开Neon电转仪(Thermo Fisher),设置电转参数:Voltage:1700V,Width:10ms,Pulse:3pulses。4.5) Electroporation: Turn on the Neon electroporator (Thermo Fisher) and set the electroporation parameters: Voltage: 1700 V, Width: 10 ms, Pulse: 3 pulses.
4.6)用10μl Neon电转枪头(Thermo Fisher)吸取离心管中的细胞和RNP混合物,注意枪头中一定不能有气泡。然后将Neon电转枪头插入到加了3ml缓冲液E(Thermo Fisher)的Neon电转管中,点击Neon电转仪屏幕上的“Start键”。4.6) Use a 10μl Neon electroporation pipette tip (Thermo Fisher) to aspirate the cell and RNP mixture in the centrifuge tube. Be careful not to have bubbles in the tip. Then insert the Neon electroporation pipette tip into the Neon electroporation tube with 3ml buffer E (Thermo Fisher) added, and click the "Start" button on the Neon electroporation instrument screen.
4.7)将电转样品立即转移到相应含小分子的培养基孔中,电转结束后将孔板放入37℃/5%CO2培养箱中继续培养24h,再将细胞转移至不含小分子的培养基中培养。4.7) Immediately transfer the electroporated samples to the corresponding medium wells containing small molecules. After electroporation, place the well plate in a 37°C/5% CO2 incubator for further 24 hours, and then transfer the cells to medium without small molecules for culturing.
5)敲入效率的检测方法:用流式细胞仪(Beckman)检测电转后3天时的细胞活率(如图14所示),以及电转后7天时的GFP+细胞百分比(即敲入效率,如图15所示)。5) Method for detecting knock-in efficiency: Flow cytometer (Beckman) was used to detect the cell viability 3 days after electroporation (as shown in FIG. 14 ), and the percentage of GFP+ cells 7 days after electroporation (i.e., knock-in efficiency, as shown in FIG. 15 ).
实验结论Experimental results
如图15所示,在T细胞中的TRAC位点,相比于不添加小分子,小分子1或小分子2和小分子5联合使用,不管是对于dsDNA还是质粒类型的模板,都能够将敲入效率提高90%-150%。As shown in Figure 15, at the TRAC site in T cells, compared with not adding small molecules, the combined use of small molecule 1 or small molecule 2 and small molecule 5 can increase the knock-in efficiency by 90%-150%, regardless of whether it is a dsDNA or plasmid type template.
实施例7:Embodiment 7:
方案:plan:
在Jurkat细胞和SJCRH30细胞的HPRT位点,插入6bp HindIII限制性内切酶的识别序列;在H1975细胞和A549细胞的VEGFA位点,插入33bp HiBiT标签。用酶切、跑胶、Agilent 2100Bioanalyzer System检测敲入效率的方法,进一步验证这两种小分子组合在提高敲入效率方面有较广的适用性。 A 6bp HindIII restriction endonuclease recognition sequence was inserted into the HPRT site of Jurkat cells and SJCRH30 cells; a 33bp HiBiT tag was inserted into the VEGFA site of H1975 cells and A549 cells. The method of detecting knock-in efficiency by enzyme digestion, gel running, and Agilent 2100 Bioanalyzer System further verified that the combination of these two small molecules has a wide range of applicability in improving knock-in efficiency.
方法:method:
1)小分子作用方案1) Small molecule action plan
①无小分子;②5μM小分子1+30μM小分子5;③2μM小分子2+30μM小分子5。① No small molecule; ② 5μM small molecule 1 + 30μM small molecule 5; ③ 2μM small molecule 2 + 30μM small molecule 5.
2)小分子孵育时间2) Small molecule incubation time
小分子1、小分子2:电转后细胞在含有小分子的培养基中培养24h;24h之后,将细胞从含有小分子的培养基中转换到不含有小分子的培养基中。Small molecule 1 and small molecule 2: After electroporation, the cells were cultured in a medium containing small molecules for 24 hours; after 24 hours, the cells were switched from the medium containing small molecules to a medium not containing small molecules.
小分子5:电转前细胞在含有小分子的培养基中培养24h以内;电转后细胞在含有小分子的培养基中培养24h,24h之后,将细胞从含有小分子的培养基中转换到不含有小分子的培养基中。Small molecule 5: Before electroporation, the cells are cultured in a medium containing small molecules for less than 24 hours; after electroporation, the cells are cultured in a medium containing small molecules for 24 hours. After 24 hours, the cells are switched from the medium containing small molecules to a medium without small molecules.
3)细胞培养3) Cell culture
细胞每2-3天传代一次,且电转所需的细胞在电转前1天进行铺板。每种细胞在进行电转前铺板时,需要准备两份细胞,其中一份细胞不加小分子进行培养,另外一份细胞在电转前24h内向其中加入终浓度为30μM的小分子5。Cells were passaged every 2-3 days, and cells required for electroporation were plated one day before electroporation. Two aliquots of each cell type were prepared before electroporation, one of which was cultured without small molecules, and the other was cultured with small molecules 5 added to a final concentration of 30 μM within 24 hours before electroporation.
4)电转4) Electroporation
4.1)向孔板中加入一定量的培养基(如表6所示):4.1) Add a certain amount of culture medium to the well plate (as shown in Table 6):
表6
Table 6
4.2)根据小分子的作用浓度以及不同组合方式,向孔板的部分孔的培养基中分别加入不同的小分子,然后将孔板放到37℃培养箱中预热。4.2) According to the concentration of small molecules and different combinations, different small molecules are added to the culture medium of some wells of the well plate, and then the well plate is placed in a 37° C. incubator for preheating.
4.3)RNP形成:将eSpCas9蛋白(GenScript,Z03470-300)、HPRT-sgRNA(SEQ ID NO:10)或VEGFA-sgRNA(SEQ ID NO:11)(GenScript,SafeEdit)、HPRT-ssDNA模板(SEQ ID NO:12)或VEGFA-ssDNA模板(SEQ ID NO:13)(GenScript)和缓冲液R(Thermo Fisher)按照表7所示的加入到一个新的、无RNase的离心管中,室温孵育5-15min形成RNP。4.3) RNP formation: eSpCas9 protein (GenScript, Z03470-300), HPRT-sgRNA (SEQ ID NO: 10) or VEGFA-sgRNA (SEQ ID NO: 11) (GenScript, SafeEdit), HPRT-ssDNA template (SEQ ID NO: 12) or VEGFA-ssDNA template (SEQ ID NO: 13) (GenScript) and buffer R (Thermo Fisher) were added to a new, RNase-free centrifuge tube as shown in Table 7 and incubated at room temperature for 5-15 min to form RNP.
表7
Table 7
4.4)电转体系:向上述有RNP的离心管中加入6μl用缓冲液R重悬的细胞(Jurkat:0.5M;SJCRH30:0.2M;H1975:0.2M;A549:0.2M)(没加小分子5的样品组用的是未用小分子5提前处理的细胞,加小分子5的样品组用的是30μM小分子5提前处理的细胞),并用枪头小心吹吸混匀。4.4) Electroporation system: Add 6 μl of cells resuspended in buffer R (Jurkat: 0.5 M; SJCRH30: 0.2 M; H1975: 0.2 M; A549: 0.2 M) to the above centrifuge tube containing RNP (the sample group without small molecule 5 used cells not pre-treated with small molecule 5, and the sample group with small molecule 5 used cells pre-treated with 30 μM small molecule 5), and mix carefully by pipetting with a pipette tip.
4.5)电转:打开Neon电转仪(Thermo Fisher),设置电转参数:Jurkat为Voltage:1600V,Width:10ms,Pulse:3pulses、SJCRH30为Voltage:1200V,Width:20ms,Pulse:3pulses、H1975为Voltage:1400V,Width:20ms,Pulse:2pulses、A549为Voltage:1200V,Width:20ms,Pulse:3pulses。4.5) Electroporation: Turn on the Neon electroporator (Thermo Fisher) and set the electroporation parameters: Jurkat: Voltage: 1600V, Width: 10ms, Pulse: 3pulses; SJCRH30: Voltage: 1200V, Width: 20ms, Pulse: 3pulses; H1975: Voltage: 1400V, Width: 20ms, Pulse: 2pulses; A549: Voltage: 1200V, Width: 20ms, Pulse: 3pulses.
4.6)用10μl Neon电转枪头(Thermo Fisher)吸取离心管中的细胞和RNP混合物,注意枪头中一定不能有气泡。然后将Neon电转枪头插入到加了3ml缓冲液E(Thermo Fisher)的Neon电转管中,点击Neon电转仪屏幕上的“Start键”。4.6) Use a 10μl Neon electroporation pipette tip (Thermo Fisher) to aspirate the cell and RNP mixture in the centrifuge tube. Be careful not to have bubbles in the tip. Then insert the Neon electroporation pipette tip into the Neon electroporation tube with 3ml buffer E (Thermo Fisher) added, and click the "Start" button on the Neon electroporation instrument screen.
4.7)将电转样品立即转移到相应含小分子的培养基孔中,电转结束后将孔板放入37℃/5%CO2培养箱中继续培养24h,再将细胞转移至不含小分子的培养基中培养。4.7) Immediately transfer the electroporated samples to the corresponding medium wells containing small molecules. After electroporation, place the well plate in a 37°C/5% CO2 incubator for further culturing for 24 hours, and then transfer the cells to medium without small molecules for culturing.
5)敲入效率的检测方法5) Methods for detecting knock-in efficiency
5.1)电转后3天时取样,样品用PBS缓冲液清洗两遍,尽量去除细胞中的PBS缓冲液;5.1) Samples were taken 3 days after electroporation and washed twice with PBS buffer to remove as much PBS buffer as possible from the cells;
5.2)向细胞中加入50μl细胞裂解液(Lucigen),用枪头吹吸混匀,转移到PCR管中,PCR程序如下:
5.2) Add 50 μl of cell lysis buffer (Lucigen) to the cells, mix well by pipetting with a pipette tip, and transfer to a PCR tube. The PCR program is as follows:
5.3)取4μl上述反应产物为模板,在50μl体系中进行PCR扩增,基于KOD FX Polymerase(TOYOBO)的PCR体系如下:

5.3) Take 4 μl of the above reaction product as a template and perform PCR amplification in a 50 μl system. The PCR system based on KOD FX Polymerase (TOYOBO) is as follows:

HPRT-F/R和VEGFA-F/R均在南京金斯瑞生物科技有限公司合成。HPRT-F/R and VEGFA-F/R were synthesized at Nanjing GenScript Biotechnology Co., Ltd.
PCR程序:
PCR procedure:
5.4)上述PCR产物进行胶回收(天根)5.4) The above PCR products were recovered by gel (Tian Gen)
5.5)胶回收产物用Qubit 1X dsDNA BR(Broad Range)Assay Kits进行浓度测定,取100ng胶回收产物进行酶切(Jurkat和SJCRH30:HindIII(NEB);H1975和A549:BsrBI(NEB)),酶切体系如下:
5.5) The concentration of the gel-recovered product was determined using Qubit 1X dsDNA BR (Broad Range) Assay Kits. 100 ng of the gel-recovered product was subjected to enzyme digestion (Jurkat and SJCRH30: HindIII (NEB); H1975 and A549: BsrBI (NEB)). The enzyme digestion system was as follows:
酶切程序如下:
The enzyme digestion procedure is as follows:
5.6)取4μl上述酶切产物,用1%琼脂糖凝胶跑胶,胶图如图16所示,酶切片段1和酶切片段2与理论值一致。另取1μl上述酶切产物,用Agilent 2100Bioanalyzer System检测酶切产物中每种长度片段的浓度,敲入效率的计算公式为:(Conc.酶切片段1+Conc.酶切片段2)/(Conc.酶切片段1+Conc.酶切片段2+Conc.全长)(如图17所示)。5.6) Take 4 μl of the above digestion product and run it on 1% agarose gel. The gel map is shown in Figure 16. The digestion fragments 1 and 2 are consistent with the theoretical values. Take another 1 μl of the above digestion product and use the Agilent 2100 Bioanalyzer System to detect the concentration of each length fragment in the digestion product. The calculation formula for the knock-in efficiency is: (Conc. digestion fragment 1 + Conc. digestion fragment 2) / (Conc. digestion fragment 1 + Conc. digestion fragment 2 + Conc. full length) (as shown in Figure 17).
实验结论Experimental results
如图17所示,在Jurkat细胞中的HPRT位点,相比于不添加小分子,小分子联合使用能够将敲入效率提高461%(小分子1+小分子5)和449%(小分子2和小分子5);在SJCRH30细胞中的HPRT位点,相比于不添加小分子,小分子联合使用能够将敲入效率提高194%(小分子1+小分子5)和184%(小分子2和小分子5);在H1975细胞中的VEGFA位点,相比于不添加小分子,小分子联合使用能够将敲入效率提高82.6%(小分子1+小分子5)和43.6%(小分子2和小分子5);在A549细胞中的VEGFA位点,相比于不添加小分子,小分子联合使用能够将敲入效率提高65.3%(小分子1+小分子5)和63.0%(小分子2和小分子5)。As shown in Figure 17, at the HPRT site in Jurkat cells, compared with not adding small molecules, the combined use of small molecules can increase the knock-in efficiency by 461% (small molecule 1 + small molecule 5) and 449% (small molecule 2 and small molecule 5); at the HPRT site in SJCRH30 cells, compared with not adding small molecules, the combined use of small molecules can increase the knock-in efficiency by 194% (small molecule 1 + small molecule 5) and 184% (small molecule 2 and small molecule 5); at the VEGFA site in H1975 cells, compared with not adding small molecules, the combined use of small molecules can increase the knock-in efficiency by 82.6% (small molecule 1 + small molecule 5) and 43.6% (small molecule 2 and small molecule 5); at the VEGFA site in A549 cells, compared with not adding small molecules, the combined use of small molecules can increase the knock-in efficiency by 65.3% (small molecule 1 + small molecule 5) and 63.0% (small molecule 2 and small molecule 5).
总的来说,两种小分子在不同细胞系、不同位点均能够很大程度上提高基因敲入效率,有较广的适用性。In general, the two small molecules can greatly improve the efficiency of gene knock-in in different cell lines and different sites, and have wide applicability.
实施例8:Embodiment 8:
方案:plan:
在Jurkat细胞中,以绿色荧光蛋白(GFP)作为报告基因,用流式细胞仪检测RAB11A位点的敲入效率,将优化出的敲入效率提升较高的几种小分子方案和商业化的HDR EnhancerV1(IDT)进行对比。In Jurkat cells, green fluorescent protein (GFP) was used as a reporter gene, and the knock-in efficiency of the RAB11A locus was detected by flow cytometry. The optimized knock-in efficiency was improved by several small molecule solutions and commercial HDR EnhancerV1(IDT) for comparison.
方法:method:
1)小分子作用方案1) Small molecule action plan
优化出的敲入效率提升较高的几种小分子方案:①5μM小分子1;②2μM小分子2;③5μM小分子1+30μM小分子5;④2μM小分子2+30μM小分子5。Several optimized small molecule solutions with higher knock-in efficiency are as follows: ①5μM small molecule 1; ②2μM small molecule 2; ③5μM small molecule 1+30μM small molecule 5; ④2μM small molecule 2+30μM small molecule 5.
商业化的IDTHDR enhancer V1:30μM(IDT官网推荐浓度)。Commercialization of IDT HDR enhancer V1: 30μM (recommended concentration on IDT official website).
2)小分子孵育时间 2) Small molecule incubation time
小分子1、小分子2、IDT HDR enhancer:电转后细胞在含有小分子的培养基中培养24h;24h之后,将细胞从含有小分子的培养基中转换到不含有小分子的培养基中。Small molecule 1, small molecule 2, IDT HDR enhancer: After electroporation, the cells were cultured in a medium containing small molecules for 24 hours. After 24 hours, the cells were switched from a medium containing small molecules to a medium without small molecules.
小分子5:电转前细胞在含有小分子的培养基中培养24h以内;电转后细胞在含有小分子的培养基中培养24h,24h之后,将细胞从含有小分子的培养基中转换到不含有小分子的培养基中。Small molecule 5: Before electroporation, the cells are cultured in a medium containing small molecules for less than 24 hours; after electroporation, the cells are cultured in a medium containing small molecules for 24 hours. After 24 hours, the cells are switched from the medium containing small molecules to a medium without small molecules.
3)细胞培养3) Cell culture
细胞每2-3天传代一次,且电转所需的细胞在电转前1天进行铺板。细胞在进行电转前铺板时,需要准备两份细胞,其中一份细胞不加小分子进行培养,另外一份细胞在电转前24h内向其中加入终浓度为30μM的小分子5。The cells were passaged every 2-3 days, and the cells required for electroporation were plated 1 day before electroporation. When cells were plated before electroporation, two aliquots of cells were prepared, one of which was cultured without adding small molecules, and the other aliquot of cells was added with small molecules at a final concentration of 30 μM within 24 hours before electroporation.
4)电转4) Electroporation
4.1)向24孔板中加入0.5mL培养基(含有10%FBS的RPMI 1640)。4.1) Add 0.5 mL of culture medium (RPMI 1640 containing 10% FBS) to a 24-well plate.
4.2)根据小分子的作用浓度以及不同组合方式,向孔板的部分孔的培养基中分别加入不同的小分子,然后将孔板放到37℃培养箱中预热。4.2) According to the concentration of small molecules and different combinations, different small molecules are added to the culture medium of some wells of the well plate, and then the well plate is placed in a 37° C. incubator for preheating.
4.3)RNP形成:将eSpCas9蛋白(GenScript,Z03470-300)、RAB11A-sgRNA(SEQ ID NO:1)(GenScript,EasyEdit)、RAB11A-dsDNA模板(SEQ ID NO:2)(GenScript)和缓冲液R(Thermo Fisher)按照表3(Jurkat细胞)所示的加入到一个新的、无RNase的离心管中,室温孵育5-15min形成RNP。4.3) RNP formation: eSpCas9 protein (GenScript, Z03470-300), RAB11A-sgRNA (SEQ ID NO: 1) (GenScript, EasyEdit), RAB11A-dsDNA template (SEQ ID NO: 2) (GenScript) and buffer R (Thermo Fisher) were added to a new, RNase-free centrifuge tube as shown in Table 3 (Jurkat cells) and incubated at room temperature for 5-15 min to form RNP.
4.4)电转体系:向上述有RNP的离心管中加入6μl用缓冲液R重悬的0.5M的Jurkat细胞(没加小分子5的样品组用的是未用小分子5提前处理的细胞,加小分子5的样品组用的是30μM小分子5提前处理的细胞),并用枪头小心吹吸混匀。4.4) Electroporation system: Add 6 μl of 0.5 M Jurkat cells resuspended in buffer R to the above centrifuge tube containing RNP (the sample group without small molecule 5 used cells not pre-treated with small molecule 5, and the sample group with small molecule 5 used cells pre-treated with 30 μM small molecule 5), and mix by carefully pipetting with a pipette tip.
4.5)电转:打开Neon电转仪(Thermo Fisher),设置电转参数:Voltage:1700V,Width:20ms,Pulse:1pulses。4.5) Electroporation: Turn on the Neon electroporator (Thermo Fisher) and set the electroporation parameters: Voltage: 1700 V, Width: 20 ms, Pulse: 1 pulses.
4.6)用10μl Neon电转枪头(Thermo Fisher)吸取离心管中的细胞和RNP混合物,注意枪头中一定不能有气泡。然后将Neon电转枪头插入到加了3ml缓冲液E(Thermo Fisher)的Neon电转管中,点击Neon电转仪屏幕上的“Start键”。4.6) Use a 10μl Neon electroporation pipette tip (Thermo Fisher) to aspirate the cell and RNP mixture in the centrifuge tube. Be careful not to have bubbles in the tip. Then insert the Neon electroporation pipette tip into the Neon electroporation tube with 3ml buffer E (Thermo Fisher) added, and click the "Start" button on the Neon electroporation instrument screen.
4.7)将电转样品立即转移到相应孔的培养基中,电转结束后将孔板放入37℃/5%CO2培养箱中继续培养24h,再将细胞转移至不含小分子的培养基中培养。4.7) Immediately transfer the electroporated samples to the culture medium of the corresponding wells. After electroporation, place the well plate in a 37°C/5% CO2 incubator for further culturing for 24 hours, and then transfer the cells to a culture medium without small molecules for culturing.
5)敲入效率的检测方法:用流式细胞仪(Beckman)检测电转后7天时的GFP+细胞百分比(即敲入效率,如图18所示)。5) Method for detecting knock-in efficiency: Flow cytometer (Beckman) was used to detect the percentage of GFP+ cells 7 days after electroporation (ie, knock-in efficiency, as shown in FIG. 18 ).
实验结论Experimental results
如图18所示,相比于单独使用小分子1或小分子2,小分子1或2和小分子5联合使用,敲入效率提高约30%;相比于IDT HDR增强剂,小分子1或2和小分子5联合使用,敲入效率提高约50%。As shown in Figure 18, compared with the use of small molecule 1 or small molecule 2 alone, the knock-in efficiency was increased by about 30% when small molecule 1 or 2 and small molecule 5 were used in combination; compared with IDT HDR enhancer, the knock-in efficiency was increased by about 50% when small molecule 1 or 2 and small molecule 5 were used in combination.
总的来说,优化的小分子组合,在提高基因敲入效率方面的作用优于商业化的IDT HDR增强剂。 Overall, the optimized small molecule combination performed better than the commercial IDT HDR enhancer in improving gene knock-in efficiency.
实施例9:Embodiment 9:
方案:plan:
在HEK293T细胞的TRAC位点,插入6bp HindIII限制性内切酶的识别序列,用Agilent 2100Bioanalyzer System检测敲入效率,进一步对比小分子组合(小分子1+小分子5)和商业化的IDTHDR enhancer V1在提高敲入效率方面的作用。A 6 bp HindIII restriction endonuclease recognition sequence was inserted into the TRAC site of HEK293T cells, and the knock-in efficiency was detected using the Agilent 2100 Bioanalyzer System. The small molecule combination (small molecule 1 + small molecule 5) was further compared with the commercial IDT The role of HDR enhancer V1 in improving knock-in efficiency.
方法:method:
1)小分子作用方案1) Small molecule action plan
小分子组合为:5μM小分子1+30μM小分子5;The small molecule combination is: 5 μM small molecule 1 + 30 μM small molecule 5;
IDTHDR enhancerV1浓度为:30μM(IDT官网推荐浓度)。IDT The concentration of HDR enhancerV1 is: 30μM (recommended concentration on IDT official website).
2)小分子孵育时间2) Small molecule incubation time
小分子1、IDTHDR enhancer V1:电转后细胞在含有小分子的培养基中培养24h;24h之后,将细胞从含有小分子的培养基中转换到不含有小分子的培养基中。Small molecules 1. IDT HDR enhancer V1: After electroporation, cells are cultured in a medium containing small molecules for 24 hours; after 24 hours, the cells are switched from a medium containing small molecules to a medium without small molecules.
小分子5:电转前细胞在含有小分子的培养基中培养24h以内;电转后细胞在含有小分子的培养基中培养24h,24h之后,将细胞从含有小分子的培养基中转换到不含有小分子的培养基中。Small molecule 5: Before electroporation, the cells are cultured in a medium containing small molecules for less than 24 hours; after electroporation, the cells are cultured in a medium containing small molecules for 24 hours. After 24 hours, the cells are switched from the medium containing small molecules to a medium without small molecules.
3)细胞培养3) Cell culture
细胞每2-3天传代一次,且电转所需的细胞在电转前1天进行铺板。细胞在进行电转前铺板时,需要准备两份细胞,其中一份细胞不加小分子进行培养,另外一份细胞在电转前24h内向其中加入终浓度为30μM的小分子5。The cells were passaged every 2-3 days, and the cells required for electroporation were plated 1 day before electroporation. When cells were plated before electroporation, two aliquots of cells were prepared, one of which was cultured without adding small molecules, and the other aliquot of cells was added with small molecules at a final concentration of 30 μM within 24 hours before electroporation.
4)电转4) Electroporation
4.1)向12孔板中加入1mL培养基(含有10%FBS的DMEM)。4.1) Add 1 mL of culture medium (DMEM containing 10% FBS) to a 12-well plate.
4.2)根据小分子的作用浓度以及不同组合方式,向孔板的部分孔的培养基中分别加入不同的小分子,然后将孔板放到37℃培养箱中预热。4.2) According to the concentration of small molecules and different combinations, different small molecules are added to the culture medium of some wells of the well plate, and then the well plate is placed in a 37° C. incubator for preheating.
4.3)RNP形成:将Cas9蛋白(Thermo Fisher,A36499)、TRAC-sgRNA(SEQ ID NO:3)(GenScript,EasyEdit)、TRAC-86bp dsODN模板(SEQ ID NO:4和5的退火产物)(GenScript)和缓冲液R(Thermo Fisher)按照表8所示的加入到一个新的、无RNase的离心管中,室温孵育5-15min形成RNP。4.3) RNP formation: Add Cas9 protein (Thermo Fisher, A36499), TRAC-sgRNA (SEQ ID NO: 3) (GenScript, EasyEdit), TRAC-86bp dsODN template (annealing product of SEQ ID NO: 4 and 5) (GenScript) and buffer R (Thermo Fisher) as shown in Table 8 into a new, RNase-free centrifuge tube and incubate at room temperature for 5-15 min to form RNP.
表8
Table 8
4.4)电转体系:向上述有RNP的离心管中加入6μl用缓冲液R重悬的0.2M的HEK293T细胞(没加小分子5的样品组用的是未用小分子5提前处理的细胞,加小分子5的样品组用的是30μM小分子5提前处理的细胞),并用枪头小心吹吸混匀。 4.4) Electroporation system: Add 6 μl of 0.2 M HEK293T cells resuspended in buffer R to the above centrifuge tube containing RNP (the sample group without small molecule 5 used cells not pre-treated with small molecule 5, and the sample group with small molecule 5 used cells pre-treated with 30 μM small molecule 5), and mix by carefully pipetting with a pipette tip.
4.5)电转:打开Neon电转仪(Thermo Fisher),设置电转参数:Voltage:1200V,Width:10ms,Pulse:3pulses。4.5) Electroporation: Turn on the Neon electroporator (Thermo Fisher) and set the electroporation parameters: Voltage: 1200 V, Width: 10 ms, Pulse: 3 pulses.
4.6)用10μl Neon电转枪头(Thermo Fisher)吸取离心管中的细胞和RNP混合物,注意枪头中一定不能有气泡。然后将Neon电转枪头插入到加了3ml缓冲液E(Thermo Fisher)的Neon电转管中,点击Neon电转仪屏幕上的“Start键”。4.6) Use a 10μl Neon electroporation pipette tip (Thermo Fisher) to aspirate the cell and RNP mixture in the centrifuge tube. Be careful not to have bubbles in the tip. Then insert the Neon electroporation pipette tip into the Neon electroporation tube with 3ml buffer E (Thermo Fisher) added, and click the "Start" button on the Neon electroporation instrument screen.
4.7)将电转样品立即转移到相应孔的培养基中,电转结束后将孔板放入37℃/5%CO2培养箱中继续培养24h,再将细胞转移至不含小分子的培养基中培养。4.7) Immediately transfer the electroporated samples to the culture medium of the corresponding wells. After electroporation, place the well plate in a 37°C/5% CO2 incubator for further culturing for 24 hours, and then transfer the cells to a culture medium without small molecules for culturing.
5)敲入效率的检测方法5) Methods for detecting knock-in efficiency
5.1)电转后3天时取样,样品用PBS缓冲液清洗两遍,尽量去除细胞中的PBS缓冲液5.1) Take samples 3 days after electroporation and wash them twice with PBS buffer to remove as much PBS buffer as possible.
5.2)向细胞中加入50μl细胞裂解液(Lucigen),用枪头吹吸混匀,转移到PCR管中,PCR程序如下:
5.2) Add 50 μl of cell lysis buffer (Lucigen) to the cells, mix well by pipetting with a pipette, and transfer to a PCR tube. The PCR program is as follows:
5.3)取4μl上述反应产物为模板,在50μl体系中进行PCR扩增,基于KOD FX Polymerase(TOYOBO)的PCR体系如下:
5.3) Take 4 μl of the above reaction product as a template and perform PCR amplification in a 50 μl system. The PCR system based on KOD FX Polymerase (TOYOBO) is as follows:
TRAC-F和TRAC-R均在南京金斯瑞生物科技有限公司合成。TRAC-F and TRAC-R were synthesized at Nanjing GenScript Biotechnology Co., Ltd.
PCR程序:
PCR procedure:
5.4)上述PCR产物进行胶回收(天根)5.4) The above PCR products were recovered by gel (Tian Gen)
5.5)胶回收产物用Nanodrop进行浓度测定,取200ng胶回收产物进行HindIII(NEB)酶切,酶切体系如下:
5.5) The gel recovery product was measured for concentration using Nanodrop, and 200 ng of the gel recovery product was subjected to HindIII (NEB) digestion. The digestion system was as follows:
酶切程序如下:
The enzyme digestion procedure is as follows:
5.6)取1μl上述酶切产物,用Agilent 2100Bioanalyzer System检测酶切产物中每种长度片段的浓度,敲入效率的计算公式为:(Conc.酶切片段1+Conc.酶切片段2)/(Conc.酶切片段1+Conc.酶切片段2+Conc.全长)(如图19所示)。5.6) Take 1 μl of the above digestion product and use Agilent 2100 Bioanalyzer System to detect the concentration of each length fragment in the digestion product. The calculation formula for the knock-in efficiency is: (Conc. digestion fragment 1 + Conc. digestion fragment 2) / (Conc. digestion fragment 1 + Conc. digestion fragment 2 + Conc. full length) (as shown in Figure 19).
实验结论Experimental results
如图19所示,相比于IDT HDR增强剂,小分子1和小分子5联合使用,敲入效率提高31.9%。在HEK293T细胞的TRAC位点,优化的小分子组合,在提高敲入效率方面的作用优于商业化的IDT HDR增强剂。As shown in Figure 19, compared with the IDT HDR enhancer, the knock-in efficiency was increased by 31.9% when small molecules 1 and 5 were used in combination. At the TRAC site of HEK293T cells, the optimized small molecule combination was superior to the commercial IDT HDR enhancer in improving the knock-in efficiency.
CRISPR/Cas9系统是一种广泛使用的基因组编辑工具,能够实现特定位点的精准基因编辑。虽然目前筛选发现的通过调控细胞修复途径的选择来提高精准基因编辑效率的 小分子种类有很多,但是这些小分子具有一定的细胞类型特异性以及上下文依赖性,且各作者描述的提高程度也存在一定的差异。本发明将两种小分子化合物搭配使用,提供了一种适用范围广,能够显著提高CRISPR/Cas9介导的精准基因编辑效率的方法。The CRISPR/Cas9 system is a widely used genome editing tool that can achieve precise gene editing at specific sites. There are many types of small molecules, but these small molecules have certain cell type specificity and context dependence, and the degree of improvement described by each author also varies. The present invention uses two small molecule compounds together to provide a method with a wide range of applications that can significantly improve the efficiency of CRISPR/Cas9-mediated precision gene editing.
本发明将两种小分子化合物联合使用,提供一种能够更好提高CRISPR/Cas9介导的精准基因编辑效率的方法,所述方法包括:The present invention uses two small molecule compounds in combination to provide a method for better improving the efficiency of CRISPR/Cas9-mediated precise gene editing, the method comprising:
针对不同细胞的不同目的位点,验证小分子1、小分子2、小分子5这三种小分子不同浓度下的敲入效率,从而确定该细胞系该位点条件下每一种小分子的最适作用浓度;Targeting different target sites in different cells, the knock-in efficiency of small molecules 1, 2, and 5 at different concentrations was verified to determine the optimal concentration of each small molecule under the conditions of the cell line and site.
将最适作用浓度的小分子1或小分子2和小分子5联合使用,用于提高上述不同细胞不同位点的敲入效率。The small molecule 1 or small molecule 2 and small molecule 5 at the optimal concentration are used in combination to improve the knock-in efficiency at different sites in the above-mentioned different cells.
本发明将小分子1或小分子2和小分子5联合使用,在不同细胞系、不同位点以及不同供体长度的情况下,可以更好的提高敲入效率,且优于某些商业化的小分子增强剂。The present invention uses small molecule 1 or small molecule 2 and small molecule 5 in combination, which can better improve the knock-in efficiency in different cell lines, different sites and different donor lengths, and is superior to some commercial small molecule enhancers.
本文中提及的核苷酸序列如下:




The nucleotide sequences mentioned in this article are as follows:




Claims (23)

  1. 在细胞内增强CRISPR基因编辑系统介导的基因敲入效率的方法,包括在对所述细胞进行基因敲入操作过程中让所述细胞与DNA依赖性蛋白激酶抑制剂和细胞分裂周期蛋白7抑制剂接触。A method for enhancing the efficiency of gene knock-in mediated by a CRISPR gene editing system in a cell, comprising contacting the cell with a DNA-dependent protein kinase inhibitor and a cell division cycle protein 7 inhibitor during the gene knock-in operation on the cell.
  2. 如权利要求1所述的方法,其中所述CRISPR基因编辑系统包括Cas9蛋白。The method of claim 1, wherein the CRISPR gene editing system comprises a Cas9 protein.
  3. 如权利要求1或2所述的方法,其中所述CRISPR基因编辑系统还包括向导RNA和用于进行同源重组修复的供体模板。The method of claim 1 or 2, wherein the CRISPR gene editing system further comprises a guide RNA and a donor template for homologous recombination repair.
  4. 如权利要求1-3中任一项所述的方法,其中所述供体模板为单链或双链形式的DNA分子。The method according to any one of claims 1 to 3, wherein the donor template is a DNA molecule in single-stranded or double-stranded form.
  5. 如权利要求1-4中任一项所述的方法,其中所述DNA依赖性蛋白激酶抑制剂选自式(I)的化合物、式(II)的化合物及其组合,其中式(I)的化合物中X为氢或者氘:
    The method according to any one of claims 1 to 4, wherein the DNA-dependent protein kinase inhibitor is selected from a compound of formula (I), a compound of formula (II), and a combination thereof, wherein X in the compound of formula (I) is hydrogen or deuterium:
    其中优选地,式(I)的化合物为:
    Preferably, the compound of formula (I) is:
  6. 如权利要求1-5中任一项所述的方法,其中所述细胞分裂周期蛋白7抑制剂为式(V)的化合物或其盐酸盐:
    The method according to any one of claims 1 to 5, wherein the cell division cycle protein 7 inhibitor is a compound of formula (V) or its hydrochloride:
  7. 如权利要求5或6所述的方法,其中所述式(I)的化合物以不低于1μM的浓度与所述细胞接触;所述式(II)的化合物以不低于0.5μM的浓度与所述细胞接触;或者,所述式(V)的化合物以不低于10μM的浓度与所述细胞接触。The method of claim 5 or 6, wherein the compound of formula (I) is contacted with the cells at a concentration of not less than 1 μM; the compound of formula (II) is contacted with the cells at a concentration of not less than 0.5 μM; or the compound of formula (V) is contacted with the cells at a concentration of not less than 10 μM.
  8. 如权利要求5-7中任一项所述的方法,其中:The method according to any one of claims 5 to 7, wherein:
    1)所述式(I)的化合物以1-30μM的浓度与所述细胞接触,并且所述式(V)的化合物以10-50μM的浓度与所述细胞接触;或者1) the compound of formula (I) is contacted with the cells at a concentration of 1-30 μM, and the compound of formula (V) is contacted with the cells at a concentration of 10-50 μM; or
    2)所述式(II)的化合物以0.5-5μM的浓度与所述细胞接触,并且所述式(V)的化合物以10-50μM的浓度与所述细胞接触。2) The compound of formula (II) is contacted with the cells at a concentration of 0.5-5 μM, and the compound of formula (V) is contacted with the cells at a concentration of 10-50 μM.
  9. 如权利要求5-8中任一项所述的方法,其中:The method according to any one of claims 5 to 8, wherein:
    1)所述式(I)的化合物以5-20μM的浓度与所述细胞接触,并且所述式(V)的化合物以20-40μM的浓度与所述细胞接触;或者1) the compound of formula (I) is contacted with the cells at a concentration of 5-20 μM, and the compound of formula (V) is contacted with the cells at a concentration of 20-40 μM; or
    2)所述式(II)的化合物以2-4μM的浓度与所述细胞接触,并且所述式(V)的化合物以20-40μM的浓度与所述细胞接触。2) The compound of formula (II) is contacted with the cells at a concentration of 2-4 μM, and the compound of formula (V) is contacted with the cells at a concentration of 20-40 μM.
  10. 如权利要求5-9中任一项所述的方法,其中:The method according to any one of claims 5 to 9, wherein:
    1)所述式(I)的化合物以5μM的浓度与所述细胞接触,并且所述式(V)的化合物以30μM的浓度与所述细胞接触;或者 1) the compound of formula (I) is contacted with the cells at a concentration of 5 μM, and the compound of formula (V) is contacted with the cells at a concentration of 30 μM; or
    2)所述式(II)的化合物以2μM的浓度与所述细胞接触,并且所述式(V)的化合物以30μM的浓度与所述细胞接触。2) The compound of formula (II) is contacted with the cells at a concentration of 2 μM, and the compound of formula (V) is contacted with the cells at a concentration of 30 μM.
  11. 如权利要求1-10中任一项所述的方法,其中所述细胞为HEK293T、Jurkat、SJCRH30、H1975或A549细胞。The method of any one of claims 1 to 10, wherein the cell is a HEK293T, Jurkat, SJCRH30, H1975 or A549 cell.
  12. 如权利要求1-11中任一项所述的方法,其中所述细胞为T细胞。The method of any one of claims 1 to 11, wherein the cell is a T cell.
  13. 如权利要求5-12中任一项所述的方法,其中对所述细胞进行基因敲入操作过程中让所述细胞与所述式(I)的化合物、所述式(II)的化合物和所述式(V)的化合物接触。The method according to any one of claims 5 to 12, wherein the cell is contacted with the compound of formula (I), the compound of formula (II) and the compound of formula (V) during the gene knock-in operation on the cell.
  14. 如权利要求5-13中任一项所述的方法,其中所述基因敲入操作通过转染进行,并在所述转染之前使所述细胞与所述式(V)的化合物接触。The method according to any one of claims 5 to 13, wherein the gene knock-in operation is performed by transfection, and the cells are contacted with the compound of formula (V) before the transfection.
  15. 根据权利要求5-14中任一项所述的方法,其中所述基因敲入操作通过转染进行,所述式(I)的化合物和/或所述式(II)的化合物在转染步骤后与所述细胞接触,所述式(V)的化合物在转染步骤前和转染步骤后都与所述细胞接触。The method according to any one of claims 5 to 14, wherein the gene knock-in operation is performed by transfection, the compound of formula (I) and/or the compound of formula (II) is contacted with the cells after the transfection step, and the compound of formula (V) is contacted with the cells both before and after the transfection step.
  16. 在细胞内增强CRISPR基因编辑系统介导的基因敲入效率的试剂盒,包括DNA依赖性蛋白激酶抑制剂和细胞分裂周期蛋白7抑制剂。A kit for enhancing the efficiency of gene knock-in mediated by the CRISPR gene editing system in cells, including a DNA-dependent protein kinase inhibitor and a cell division cycle protein 7 inhibitor.
  17. 如权利要求16所述的试剂盒,其中所述试剂盒还包括Cas9蛋白或其编码核酸分子。The kit as claimed in claim 16, wherein the kit further comprises a Cas9 protein or a nucleic acid molecule encoding it.
  18. 如权利要求16或17所述的试剂盒,其中所述试剂盒还包括向导RNA和/或用于进行同源重组修复的供体模板。The kit of claim 16 or 17, wherein the kit further comprises a guide RNA and/or a donor template for homologous recombination repair.
  19. 如权利要求16-18中任一项所述的试剂盒,其中所述供体模板为单链或双链形式的DNA分子。The kit according to any one of claims 16 to 18, wherein the donor template is a DNA molecule in single-stranded or double-stranded form.
  20. 如权利要求16-19中任一项所述的试剂盒,其中所述DNA依赖性蛋白激酶抑制剂选自式(I)的化合物、式(II)的化合物及其组合,其中式(I)的化合物中X为氢或者氘:

    The kit according to any one of claims 16 to 19, wherein the DNA-dependent protein kinase inhibitor is selected from a compound of formula (I), a compound of formula (II), and a combination thereof, wherein X in the compound of formula (I) is hydrogen or deuterium:

    其中优选地,式(I)的化合物为:
    Preferably, the compound of formula (I) is:
  21. 如权利要求16-20中任一项所述的试剂盒,其中所述细胞分裂周期蛋白7抑制剂为式(V)的化合物或其盐酸盐:
    The kit according to any one of claims 16 to 20, wherein the cell division cycle protein 7 inhibitor is a compound of formula (V) or its hydrochloride:
  22. 如权利要求16-21中任一项所述的试剂盒,其中所述细胞为HEK293T、Jurkat、SJCRH30、H1975或A549细胞。The kit according to any one of claims 16 to 21, wherein the cell is a HEK293T, Jurkat, SJCRH30, H1975 or A549 cell.
  23. 如权利要求16-22中任一项所述的试剂盒,其中所述细胞为T细胞。 The kit of any one of claims 16 to 22, wherein the cell is a T cell.
PCT/CN2023/124585 2022-10-14 2023-10-13 Method for improving crispr/cas9-mediated genome knock-in editing efficiency WO2024078626A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202211263292 2022-10-14
CN202211263292.9 2022-10-14

Publications (1)

Publication Number Publication Date
WO2024078626A1 true WO2024078626A1 (en) 2024-04-18

Family

ID=90668891

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/124585 WO2024078626A1 (en) 2022-10-14 2023-10-13 Method for improving crispr/cas9-mediated genome knock-in editing efficiency

Country Status (1)

Country Link
WO (1) WO2024078626A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107429263A (en) * 2015-01-15 2017-12-01 斯坦福大学托管董事会 The method of controlling gene group editor
CN111139259A (en) * 2020-01-18 2020-05-12 潍坊医学院 Method for improving homologous recombination efficiency in gene editing
CN112359065A (en) * 2020-09-21 2021-02-12 中国医学科学院血液病医院(中国医学科学院血液学研究所) Small molecule composition for improving gene knock-in efficiency
CN113215193A (en) * 2021-03-18 2021-08-06 温州医科大学 Method for improving gene knockout and base editing system activity by using small molecular compound and application method thereof
WO2021166306A1 (en) * 2020-02-19 2021-08-26 Kyoto University Production method of genome-edited cells and kit therefor

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107429263A (en) * 2015-01-15 2017-12-01 斯坦福大学托管董事会 The method of controlling gene group editor
CN111139259A (en) * 2020-01-18 2020-05-12 潍坊医学院 Method for improving homologous recombination efficiency in gene editing
WO2021166306A1 (en) * 2020-02-19 2021-08-26 Kyoto University Production method of genome-edited cells and kit therefor
CN112359065A (en) * 2020-09-21 2021-02-12 中国医学科学院血液病医院(中国医学科学院血液学研究所) Small molecule composition for improving gene knock-in efficiency
CN113215193A (en) * 2021-03-18 2021-08-06 温州医科大学 Method for improving gene knockout and base editing system activity by using small molecular compound and application method thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
WIENERT BEEKE, NGUYEN DAVID N., GUENTHER ALEXIS, FENG SHARON J., LOCKE MELISSA N., WYMAN STACIA K., SHIN JIYUNG, KAZANE KATELYNN R: "Timed inhibition of CDC7 increases CRISPR-Cas9 mediated templated repair", NATURE COMMUNICATIONS, vol. 11, no. 1, 1 December 2020 (2020-12-01), pages 1 - 15, XP055839138, DOI: 10.1038/s41467-020-15845-1 *

Similar Documents

Publication Publication Date Title
Xu et al. Engineered miniature CRISPR-Cas system for mammalian genome regulation and editing
Murugan et al. The revolution continues: newly discovered systems expand the CRISPR-Cas toolkit
JP7114117B2 (en) Protein delivery in primary hematopoietic cells
US20220033858A1 (en) Crispr oligoncleotides and gene editing
AU2017277918B2 (en) High specificity genome editing using chemically modified guide RNAs
Ranganathan et al. Expansion of the CRISPR–Cas9 genome targeting space through the use of H1 promoter-expressed guide RNAs
Cong et al. Genome engineering using CRISPR-Cas9 system
JP6835726B2 (en) CRISPR hybrid DNA / RNA polynucleotide and usage
JP6739335B2 (en) Genome engineering
US20180282722A1 (en) Chimeric DNA:RNA Guide for High Accuracy Cas9 Genome Editing
JP2023552374A (en) Operated class 2 V-type CRISPR system
KR20190095412A (en) How to increase the efficiency of homologous directed repair (HDR) in the cellular genome
BR112016000571B1 (en) IN VITRO METHODS FOR MODULATING EXPRESSION AND FOR ALTERING ONE OR MORE TARGET NUCLEIC ACIDS IN A CELL SIMULTANEOUSLY WITH REGULATING THE EXPRESSION OF ONE OR MORE TARGET NUCLEIC ACIDS IN A CELL AS WELL AS A YEAST OR BACTERIAL CELL COMPRISING NUCLEIC ACIDS
US20190390229A1 (en) Gene editing reagents with reduced toxicity
US11834670B2 (en) Site-specific DNA modification using a donor DNA repair template having tandem repeat sequences
CN113215193B (en) Method for improving activity of gene knockout and base editing system by small molecule compound and application method thereof
Schatoff et al. Base editing the mammalian genome
US20240002834A1 (en) Adenine base editor lacking cytosine editing activity and use thereof
WO2024078626A1 (en) Method for improving crispr/cas9-mediated genome knock-in editing efficiency
Burnett et al. Examination of the cell cycle dependence of cytosine and adenine base editors
Shaikh et al. CRISPR/Cas9 genome editing tool: a promising tool for therapeutic applications on respiratory diseases
Hamaker Development of site-specific integration strategies and characterization of protein expression instability to improve CHO cell line engineering
Gopalappa et al. Efficient genome editing by FACS enrichment of paired D10A Cas9 nickases coupled with fluorescent proteins
KR20220077054A (en) Adenine base editors with enhanced thymine-cytosine sequence-specific cytosine editing activity and use thereof
Ojha et al. In vitro and in cellula site-directed RNA editing using the λNDD-BoxB system