WO2020037490A1 - Method of genome editing in mammalian stem cell - Google Patents

Method of genome editing in mammalian stem cell Download PDF

Info

Publication number
WO2020037490A1
WO2020037490A1 PCT/CN2018/101486 CN2018101486W WO2020037490A1 WO 2020037490 A1 WO2020037490 A1 WO 2020037490A1 CN 2018101486 W CN2018101486 W CN 2018101486W WO 2020037490 A1 WO2020037490 A1 WO 2020037490A1
Authority
WO
WIPO (PCT)
Prior art keywords
bcl
stem cell
cell
mammalian stem
sequence
Prior art date
Application number
PCT/CN2018/101486
Other languages
French (fr)
Inventor
Tao Cheng
Xiaolan Li
Jianping Zhang
Wei Wen
Changkai SUN
Original Assignee
Institute Of Hematology And Blood Diseases Hospital, Cams & Pumc
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 Institute Of Hematology And Blood Diseases Hospital, Cams & Pumc filed Critical Institute Of Hematology And Blood Diseases Hospital, Cams & Pumc
Priority to PCT/CN2018/101486 priority Critical patent/WO2020037490A1/en
Priority to CN201880030560.8A priority patent/CN110678553B/en
Publication of WO2020037490A1 publication Critical patent/WO2020037490A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4747Apoptosis related proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • C12N15/902Stable introduction of foreign DNA into chromosome using homologous recombination
    • C12N15/907Stable introduction of foreign DNA into chromosome using homologous recombination in mammalian 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases RNAses, DNAses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/20Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPRs]
    • 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
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/106Plasmid DNA for vertebrates
    • C12N2800/107Plasmid DNA for vertebrates for mammalian
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the invention relates to a method of genome editing in mammalian stem cell.
  • ESCs Human embryonic stem cells
  • ESCs Human embryonic stem cells
  • iPSC patient-specific induced pluripotent stem cell
  • DSB double-stranded DNA break
  • CRISPR-Cas9 RNA-guided endonuclease
  • ZFN zinc-finger nuclease
  • TALEN transcription activator-like effector nuclease
  • CRISPR-Cas9 RNA-guided endonuclease
  • the commonly used Cas9 is from streptococcus pyogenes (Sp) .
  • NHEJ non-homologous end joining
  • HDR homology-directed repair
  • HDR pathway could be used to integrate the sequence between HAs to create precise DNA deletion, substitution or insertion, leading to the correction of diseased genes or the targeted integration of genes of interest.
  • HDR-mediated knockin is often inefficient.
  • the invention provides a method for efficient precise gene editing.
  • a method of editing a target genomic DNA in a mammalian stem cell comprises introducing an apoptosis regulator and a genome editing composition into the mammalian stem cell to generate a modified mammalian stem cell that overexpress the apoptosis regulator, wherein the apoptosis regulator is BCL-XL, wherein the genome editing composition a genome editing endonuclease that cleaves within a desired target sequence of the genomic DNA of the mammalian stem cell and edits the target genomic DNA.
  • the BCL-XL comprises an amino acid sequence having at least 70%amino acid sequence identity to an amino acid sequence of SEQ ID NO: 1.
  • introducing the apoptosis regulator and the genome editing composition into the mammalian stem cell may comprise: introducing the apoptosis regulator into the mammalian stem cell to generate the modified mammalian stem cell that overexpress the apoptosis regulator; and introducing the genome editing composition into the modified mammalian stem cell.
  • the genome editing composition may comprise a RNA-guided endonuclease and a guide RNA.
  • the RNA-guided endonuclease may be Cas9.
  • the guide RNA may comprise a clustered regularly interspaced short palindromic repeats RNA and a tracrRNA.
  • the genome editing composition may comprise a donor plasmid comprising a donor sequence, wherein the donor sequence is inserted into the genome at an insertion site through homology-directed repair.
  • the mammalian stem cell may comprise an embryonic stem cell or a pluripotent stem cell.
  • the mammalian stem cell may be an induced pluripotent stem cell.
  • the method further may comprise introducing a BCL inhibitor into the cell.
  • the BCL inhibitor may be ABT-263.
  • the BCL-XL is stably expressed.
  • the BCL-XL is transiently overexpressed.
  • the BCL-XL is overexpressed for a period of time from about 1 hour to about 72 hour.
  • the BCL-XL is overexpressed by at least 5-fold over background.
  • FIG. 1 depicts a schematic design of stable BCL-XL overexpression in human iPSCs.
  • FIG. 2 depicts a schematic of HDR editing at PRDM14.
  • FIG. 3 depicts flow cytometry analysis of iPSC-Lenti-BCL-XL lines and iPSC-Lenti-control lines after co-transfection of CRISPR plasmids.
  • FIG. 4 depicts a schematic design of transient BCL-XL overexpression in human iPSCs.
  • FIG. 5 depicts flow cytometry analysis of transient BCL-XL overexpression group and control group.
  • FIG. 6 depicts a schematic of HDR editing at CTNNB1.
  • FIG. 7 depicts a schematic of HDR editing at OCT4.
  • FIG. 8 depicts a flow cytometry analysis indicating KI efficiency at CTNNB1 and OCT4 in iPSC.
  • FIG. 9 depicts a schematic of NHEJ-mediated knockout at CD326.
  • FIG. 10 depicts a schematic of NHEJ-mediated knockout at CD9.
  • FIG. 11 depicts a flow cytometry analysis indicating KO efficiency at CD326 and CD9 in iPSC.
  • FIG. 12 depicts dynamic changes in relative cell numbers after electroporation with genome editing plasmids together with or without BCL-XL.
  • FIG. 13 depicts flow cytometry analysis of iPSCs with BCL-XL, BCL2 or MCL1.
  • FIG. 14 depicts expression levels of Cas9 and sgRNA.
  • FIG. 16 depicts effects of BBC3 knockout on iPSC cell survival and editing efficiency.
  • FIG. 17 depicts effects of various dose and treatment period of ABT-263 on cell survival of iPSC after electroporation.
  • FIG. 18 depicts effects of various dose and treatment period of ABT-263 on HDR efficiency of iPSC after electroporation.
  • FIG. 19 depicts effects of ABT-263 on iPSC cell survival and editing efficiency.
  • FIG. 20 depicts a schematic of HDR-mediated dual KI at PRDM14 and CTNNB1 in iPSCs.
  • FIG. 21 depicts a flow cytometry analysis indicating KI efficiency at CTNNB1 in iPSC.
  • FIG. 22 depicts a schematic of Dual editing at PRDM14 by HDR and CD326 by NHEJ.
  • FIG. 23 depicts a flow cytometry analysis indicating KO efficiency at CD326 in iPSC.
  • FIG. 24 depicts a schematic for gene knockout by biallelic HDR insertion of section cassettes.
  • FIG. 25 depicts a flow cytometry analysis indicating KO efficiency at CD326 in iPSC.
  • FIG. 26 depicts effects of transient BCL-XL, BCL2 or MCL1 overexpression on HDR efficiency in mouse ES cells.
  • FIG. 27 depicts effects of transient BCL-XL, BCL2 or MCL1 overexpression on HDR efficiency in K562 cells.
  • FIG. 28 depicts effects of transient BCL-XL, BCL2 or MCL1 overexpression on HDR efficiency in 293T cells.
  • FIG. 29 depicts effects of transient BCL-XL, BCL2 or MCL1 overexpression on HDR efficiency in Jurkat cells.
  • the present invention provides a novel genome editing method with significantly higher efficiency compared to traditional genome editing methods using RNA-guided endonuclease such as CRISPR/Cas9.
  • the method of the present application utilizes an apoptosis regulator BCL-XL.
  • BCL-XL apoptosis regulator
  • the improved genome editing system provides useful tools for applications ranging from manipulating human iPSC genomes to creating gene-modified animal models.
  • nucleic acid refers to a deoxyribonucleotide or ribonucleotide polymer, in linear or circular conformation, and in either single-or double-stranded form.
  • polynucleotide refers to a deoxyribonucleotide or ribonucleotide polymer, in linear or circular conformation, and in either single-or double-stranded form.
  • these terms are not to be construed as limiting with respect to the length of a polymer.
  • the terms can encompass known analogues of natural nucleotides, as well as nucleotides that are modified in the base, sugar and/or phosphate moieties (e.g., phosphorothioate backbones) .
  • an analogue of a particular nucleotide has the same base-pairing specificity; i.e., an analogue of A will base-pair with T.
  • polypeptide, ” “peptide” and “protein” are used interchangeably to refer to a polymer of amino acid residues.
  • the term also applies to amino acid polymers in which one or more amino acids are chemical analogues or modified derivatives of a corresponding naturally-occurring amino acids.
  • sequence refers to a nucleotide sequence of any length, which can be DNA or RNA; can be linear, circular or branched and can be either single-stranded or double stranded.
  • homologous nucleic acid as used herein includes a nucleic acid sequence that is either identical or substantially similar to a known reference sequence. In one embodiment, the term “homologous nucleic acid” is used to characterize a sequence that is at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or even 100%identical to a known reference sequence.
  • HDR homology-directed repair
  • This process requires nucleotide sequence homology, uses a "donor” molecule to template repair of a "target” molecule (i.e., the one that experienced the double-strand break) , and leads to the transfer of genetic information from the donor to the target.
  • Homology-directed repair may result in an alteration of the sequence of the target molecule (e.g., insertion, deletion, mutation) , if the donor polynucleotide differs from the target molecule and part or all of the sequence of the donor polynucleotide is incorporated into the target DNA.
  • non-homologous end joining refers to the repair of double-strand breaks in DNA by direct ligation of the break ends to one another without the need for a homologous template (in contrast to homology-directed repair, which requires a homologous sequence to guide repair) . NHEJ often results in the loss (deletion) of nucleotide sequence near the site of the double-strand break.
  • stem cell is used herein to refer to a cell ⁇ e.g., plant stem cell, vertebrate stem cell) that has the ability both to self-renew and to generate a differentiated cell type.
  • stem cell e.g., plant stem cell, vertebrate stem cell
  • differentiated is a relative term.
  • a “differentiated cell” is a cell that has progressed further down the developmental pathway than the cell it is being compared with.
  • Pluripotent stem cell or "PSC” is used herein to mean a stem cell capable of producing all cell types of the organism. Therefore, a PSC can give rise to cells of all germ layers of the organism (e.g., the endoderm, mesoderm, and ectoderm of a vertebrate) . Pluripotent cells are capable of forming teratomas and of contributing to ectoderm, mesoderm, or endoderm tissues in a living organism. Pluripotent stem cells of plants are capable of giving rise to all cell types of the plant (e.g., cells of the root, stem, leaves, etc. ) .
  • PSC refers to pluripotent stem cells regardless of their derivation
  • the term PSC encompasses the terms ESC and iPSC, as well as the term embryonic germ stem cells (EGSC) , which are another example of a PSC.
  • PSCs may be in the form of an established cell line, they may be obtained directly from primary embryonic tissue, or they may be derived from a somatic cell. PSCs can be target cells of the methods described herein.
  • iPSC induced pluripotent stem cell
  • PSC induced pluripotent stem cell
  • iPSCs can be derived from multiple different cell types, including terminally differentiated cells. iPSCs have an ES cell-like morphology, growing as flat colonies with large nucleo-cytoplasmic ratios, defined borders and prominent nuclei.
  • donor sequence refers to a nucleic acid to be inserted into the chromosome of a host cell.
  • a donor sequence can be of any length, for example between 2 and 10,000 nucleotides in length (or any integer value therebetween or thereabove) .
  • references to “about” a value or parameter herein includes (and describes) variations that are directed to that value or parameter per se. For example, description referring to “about X” includes description of “X” .
  • compositions and methods of the present invention may comprise, consist of, or consist essentially of the essential elements and limitations of the invention described herein, as well as any additional or optional ingredients, components, or limitations described herein or otherwise useful in a nutritional or pharmaceutical application.
  • the present invention provides a method for genome editing in mammalian stem cells.
  • the method involves modifying the mammalian stem cells by overexpressing an apoptosis regulator, and introducing a genome editing composition into the mammalian stem cells.
  • Genome editing comprises NHEJ and HDR.
  • a genome-editing endonuclease generates a single-or double-strand break in a target genomic DNA, and the single-or double-strand break is repaired. Repair that occurs via NHEJ is sometimes referred to an "indel" (insertion or deletion) ; DNA repair via HDR is sometimes referred to as "gene correction" or "gene modification.
  • editing a target genomic DNA involves generating a substitution of one or more nucleotides in the target genomic DNA, generating an edited target genomic DNA. In some cases, editing a target genomic DNA involves deletion of one or more nucleotides from the target genomic DNA, generating an edited target genomic DNA. In some cases, editing a target genomic DNA involves insertion of one or more nucleotides from the target genomic DNA, generating an edited target genomic DNA.
  • the method comprises introducing an apoptosis regulator and a genome editing composition into the mammalian stem cell to generate a modified mammalian stem cell that overexpress the apoptosis regulator, wherein the apoptosis regulator is BCL-XL, wherein the genome editing composition a genome editing endonuclease that cleaves within a desired target sequence of the genomic DNA of the mammalian stem cell and edits the target genomic DNA.
  • BCL-XL encoded by the BCL2-like 1 (BCL1L1) gene, maintains the outer mitochondrial membrane integrity and thereby prevent the release of mitochondrial contents such as cytochrome c, an apoptosis activator.
  • BCL-XL comprises an amino acid sequence having at least 70%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence of SEQ ID NO: 1.
  • the mammalian stem cells described herein may be embryonic stem cell or a pluripotent stem cell. In some embodiments, the mammalian stem cells may be induced pluripotent stem cell. In some embodiments, the mammalian stem cells may be human induced pluripotent stem cell.
  • the BCL-XL is introduced into the mammalian stem cell in the form of a protein.
  • the BCL-XL is introduced into the mammalian stem cell in the form of a nucleic acid, such as a messenger RNA (mRNA) , or a cDNA.
  • Nucleic acids can be delivered as part of a larger construct, such as a plasmid or viral vector, or directly, e.g., by electroporation, lipid vesicles, viral transporters, and microinjection.
  • the BCL-XL may be introduced into the cell by a variety of means known in the art, including transfection, calcium phosphate-DNA co-precipitation, DEAE-dextran-mediated transfection, polybrene-mediated transfection, electroporation, microinjection, transduction, cell fusion, liposome fusion, lipofection, protoplast fusion, retroviral infection, use of a gene gun, use of a DNA vector transporter, and biolistics (e.g., particle bombardment) .
  • a nucleotide sequence encoding the BCL-XL can be operably linked to a transcriptional control element (s) , e.g., a promoter, where the promoter is active in a mammalian stem cell.
  • a transcriptional control element e.g., a promoter
  • the promoter is a constitutive promoter.
  • the BCL-XL is stably expressed.
  • the promoter is an inducible promoter.
  • a nucleotide sequence encoding the BCL-XL is introduced into the mammalian stem cell and then inserted to the genome of the mammalian stem cell.
  • the genome editing composition and the BCL-XL are introduced into the cell simultaneously.
  • the BCL-XL may be introduced into the cell first, and the genome editing composition is subsequently introduced.
  • introducing the apoptosis regulator (e.g., BCL-XL) and the genome editing composition into the mammalian stem cell comprising the following steps. First, the apoptosis regulator (e.g., BCL-XL) is introduced into the mammalian stem cell to generate the modified mammalian stem cell that overexpress the apoptosis regulator (e.g., BCL-XL) first. Then, the genome editing composition is introduced into the modified mammalian stem cell.
  • the BCL-XL is constitutively overexpressed in the cell. In some embodiments, the BCL-XL is transiently overexpressed. For example, in some cases, the BCL-XL is overexpressed for a period of time of from about 1 hour to about 72 hours. For example, in some cases, the BCL-XL is overexpressed for a period of time of from about 1 hour to about 4 hours, from about 4 hours to about 8 hours, from about 8 hours to about 12 hours, from about 12 hours to about 16 hours, from about 16 hours to about 20 hours, from about 20 hours to about 24 hours, from about 24 hours to about 30 hours, from about 30 hours to about 36 hours, from about 36 hours to about 42 hours, or from about 42 hours to about 48 hours. In some cases, the BCL-XL is overexpressed for a period of time of from about 12 hour to about 48 hours. In some cases, the BCL-XL is overexpressed for a period of time of from about 12 hours to about 24 hours.
  • the BCL-XL is introduced in to the cell such that the level of the BCL-XL in the cell is at least 2-fold, or more than 2-fold, higher than the background level of the BCL-XL in a control (unmodified) cell.
  • the BCL-XL is introduced in to the cell such that the level of the BCL-XL in the cell is from 2-fold to 10-fold, or from 5-fold to 10-fold, higher than the background level of the BCL-XL in a control (unmodified) cell.
  • Overexpressing the BCL-XL in a mammalian stem cell generates a modified mammalian stem cell.
  • the step of introducing a genome editing composition into the mammalian stem cell occurs during a period of time in which the BCL-XL is overexpressed.
  • overexpression of the BCL-XL in the mammalian stem cell increases the survival of the mammalian stem cell when the mammalian stem cell is contacted with a genome editing composition by at least 5-fold, at least 10-fold, at least 20-fold, or more than 20-fold, compared with the survival of a control stem cell that does not overexpress the BCL-XL, and is contacted with the genome editing composition.
  • overexpression of the BCL-XL in the mammalian stem cell increases the survival of the mammalian stem cell when the mammalian stem cell is contacted with a genome editing composition by 10-fold to 20-fold, compared with the survival of a control stem cell that does not overexpress the BCL-XL, and is contacted with the genome editing composition.
  • overexpression of the BCL-XL in the mammalian stem cell increase HDR-mediated KI (knockin) efficiency when the mammalian stem cell is contacted with a genome editing composition by at least 10-fold, at least 20-fold, at least 50-fold, at least 100-fold, or more than 100-fold, compared with the survival of a control stem cell that does not overexpress the BCL-XL, and is contacted with the genome editing composition.
  • overexpression of the BCL-XL in the mammalian stem cell increase HDR-mediated KI (knockin) efficiency when the mammalian stem cell is contacted with a genome editing composition by 20-fold to 100-fold, compared with the survival of a control stem cell that does not overexpress the BCL-XL, and is contacted with the genome editing composition.
  • overexpression of the BCL-XL in the mammalian stem cell increase NHEJ-mediated KO (knockout) efficiency when the mammalian stem cell is contacted with a genome editing composition by at least 1-fold, at least 2-fold, at least 5-fold, or more than 5-fold, compared with the survival of a control stem cell that does not overexpress the BCL-XL, and is contacted with the genome editing composition.
  • overexpression of the BCL-XL in the mammalian stem cell increase NHEJ-mediated KO (knockout) efficiency when the mammalian stem cell is contacted with a genome editing composition by 1-fold to 5-fold, compared with the survival of a control stem cell that does not overexpress the BCL-XL, and is contacted with the genome editing composition.
  • NHEJ-mediated KO knockout
  • the efficiency of genome editing of a mammalian stem cell is increased when the mammalian stem cell overexpresses the BCL-XL.
  • the genome editing composition comprises a RNA-guided endonuclease and a guide RNA.
  • the RNA-guided endonuclease is introduced into the eukaryotic cell in the form of a protein, or in the form of a nucleic acid encoding the RNA-guided endonuclease, such as a messenger RNA (mRNA) , or a cDNA.
  • Nucleic acids can be delivered as part of a larger construct, such as a plasmid or viral vector, or directly, e.g., by electroporation, lipid vesicles, viral transporters, and microinjection.
  • the RNA-guided endonuclease can be introduced into the cell by a variety of means known in the art, including transfection, calcium phosphate-DNA co-precipitation, DEAE-dextran-mediated transfection, polybrene-mediated transfection, electroporation, microinjection, transduction, cell fusion, liposome fusion, lipofection, protoplast fusion, retroviral infection, use of a gene gun, use of a DNA vector transporter, and biolistics (e.g., particle bombardment) .
  • the nucleic acid encoding the RNA-guided endonuclease is introduced into the cell by transfection (including for example transfection through electroporation) . In some embodiments, the nucleic acid encoding the RNA-guided endonuclease is introduced into the cell by injection.
  • the guide RNA can be introduced, for example, as RNA or as a plasmid or other nucleic acid vector encoding the guide RNA.
  • the RNA-guided endonuclease binds to the gRNA and the target DNA to which the gRNA binds and cleaves the chromosome at the designed specific site.
  • the guide RNA can be introduced into the cell by a variety of means known in the art, including transfection, calcium phosphate-DNA co-precipitation, DEAE-dextran-mediated transfection, polybrene-mediated transfection, electroporation, microinjection, transduction, cell fusion, liposome fusion, lipofection, protoplast fusion, retroviral infection, use of a gene gun, use of a DNA vector transporter, and biolistics (e.g., particle bombardment) .
  • the RNA-guided endonuclease is a sequence-specific nuclease.
  • sequence-specific nuclease refers to a protein that recognizes and binds to a polynucleotide at a specific nucleic acid sequence and catalyzes a double-strand break in the polynucleotide.
  • the RNA-guided endonuclease cleaves the chromosome only once, i.e., a single double-strand break is introduced at the designed specific site during the methods described herein.
  • RNA-guided endonuclease system that can be used with the methods and compositions described herein includes the Cas/CRISPR system.
  • the Cas/CRISPR (Clustered Regularly interspaced Short Palindromic Repeats) system exploits RNA-guided DNA-binding and sequence-specific cleavage of target DNA.
  • a guide RNA (gRNA) contains about 20-25 (such as 20) nucleotides that are complementary to a target genomic DNA sequence upstream of a genomic PAM (protospacer adjacent motifs) site and a constant RNA scaffold region.
  • the target sequence is associated with a PAM, which is a short sequence recognized by the CRISPR complex.
  • PAMs are typically 2-5 bp sequences adjacent to the protospacer (that is, the target sequence) .
  • Examples of PAM sequences are known in the art, and the skilled person will be able to identify further PAM sequences for use with a given CRISPR enzyme.
  • target sites for Cas9 from S. pyogenes, with PAM sequences NGG may be identified by searching for 5’-Nx-NGG-3’both on an input sequence and on the reverse-complement of the input.
  • the genomic PAM site used herein is NGG, NNG, NAG, NGGNG, or NNAGAAW.
  • the Streptococcus pyogenes Cas9 (SpCas9) is used and the corresponding PAM is NGG.
  • different Cas9 enzymes from different bacterial strains use different PAM sequences.
  • the Cas (CRISPR-associated) protein binds to the gRNA and the target DNA to which the gRNA binds and introduces a double-strand break in a defined location upstream of the PAM site.
  • the CRISPR/Cas, Cas/CRISPR, or the CRISPR-Cas system does not require the generation of customized proteins to target specific sequences but rather a single Cas enzyme can be programmed by a short RNA molecule to recognize a specific DNA target, i.e., the Cas enzyme can be recruited to a specific DNA target using the short RNA molecule.
  • the RNA-guided endonuclease is a type II Cas protein. In some embodiments, the RNA-guided endonuclease is Cas9, a homolog thereof, or a modified version thereof. In some embodiments, a combination of two or more Cas proteins can be used. In some embodiments, the CRISPR enzyme is Cas9, and may be Cas9 from S. pyogenes or S. pneumoniae. In some embodiments, Cas9 is used in the methods described herein. Cas9 harbors two independent nuclease domains homologous to HNH and RuvC endonucleases, and can cut double strands of DNA to generate blunt-ended double strand breaks (DSBs) under the guidance of gRNA.
  • DSBs blunt-ended double strand breaks
  • a guide RNA is an RNA comprising a 5’region comprising at least one repeat from a CRISPR locus and a 3’region that is complementary to the predetermined insertion site on the chromosome.
  • the 5’region comprises a sequence that is complementary to the predetermined cleavage site on the chromosome
  • the 3’region comprises at least one repeat from a CRISPR locus.
  • the 3’region of the guide RNA further comprises the one or more structural sequences of crRNA and/or tracrRNA.
  • the guide RNA comprises a crRNA and a tracrRNA, and the two pieces of RNA form a complex through hybridization.
  • the genome editing composition further comprises a donor plasmid comprising a donor sequence.
  • the donor plasmid can be introduced into the cell by a variety of means known in the art, including transfection, calcium phosphate-DNA co-precipitation, DEAE-dextran-mediated transfection, polybrene-mediated transfection, electroporation, microinjection, transduction, cell fusion, liposome fusion, lipofection, protoplast fusion, retroviral infection, use of a gene gun, use of a DNA vector transporter, and biolistics (e.g., particle bombardment) .
  • the donor sequence is flanked with a 5’homology arm and a 3’homology arm, wherein the 5’homology arm is homologous to a 5’target sequence upstream of the insertion site on the genome and the 3’homology arm is homologous to a 3’target sequence downstream of the insertion site on the genome, wherein the donor sequence is inserted in to the genome at the insertion site through homology-directed repair.
  • the insertion of the donor sequence can be evaluated using any methods known in the art. For example, a 5’primer corresponding to a sequence upstream of the 5’homology arm and a corresponding 3’primer corresponding to a region in the donor sequence can be designed to assess the 5’-junction of the insertion. Similarly, a 3’primer corresponding to a sequence downstream of the 3’homology arm and a corresponding 5’primer corresponding to a region in the donor sequence can be designed to assess the 3’-junction of the insertion.
  • the insertion site can be at any desired site, so long as RNA-guided endonuclease can be designed to effect cleavage at such site.
  • the insertion site is at a target gene locus. In some embodiments, the insertion site is not a gene locus.
  • the donor nucleic acid is a sequence not present in the host cell. In some embodiments, the donor sequence is an endogenous sequence present at a site other than the predetermined target site. In some embodiments, the donor sequence is a coding sequence. In some embodiments, the donor sequence is a non-coding sequence. In some embodiments, the donor sequence is a mutant locus of a gene.
  • the size of the donor sequence can range from about 1 bp to about 100 kb. In certain embodiments, the size of the donor sequence is between about 1 bp and about 10 bp, between about 10 bp and about 50 bp, between about 50 bp and about 100 bp, between about 100 bp and about 500 bp, between about 500 bp and about 1 kb, between about 1 kb and about 10 kb, between about 10 kb and about 50 kb, between about 50 kb and about 100 kb, or more than about 100 kb.
  • the donor sequence is an exogenous gene to be inserted into the chromosome.
  • the donor sequence is modified sequence that replaces the endogenous sequence at the target site.
  • the donor sequence may be a gene harboring a desired mutation, and can be used to replace the endogenous gene present on the chromosome.
  • the donor sequence is a regulatory element.
  • the donor sequence is a tag or a coding sequence encoding a reporter protein and/or RNA.
  • the donor sequence is inserted in frame into the coding sequence of a target gene which will allow expression of a fusion protein comprising an exogenous sequence fused to the N-or C-terminus of the target protein.
  • the donor plasmid described herein is cleaved within the cell to produce a linear nucleic acid.
  • the linear nucleic acid described herein comprises a 5’homology arm, a donor sequence, and a 3’homology arm.
  • the donor sequence is flanked with a 5’homology arm and a 3’homology arm.
  • the homology arms are at least about 50 bp in length, for example at least about any of 50 bp, 100 bp, 200 bp, 300 bp, 600 bp, 900 bp, 1 kb, 1.5 kb, 2 kb, 4kb, 6kb, 10kb, 15kb and 20 kp in length. In some embodiments, the homology arms are at least about 300 bp in length.
  • the homology arms may range from about 50 bp to about 2000 bp, from about 100 bp to about 2000 bp, from about 150 bp to about 2000 bp, from about 300 bp to about 2000 bp, from about 300 bp to about 1500 bp, from about 300 bp to about 1000 bp in length.
  • the length of the 5’homology arm and the length of the 3’homology arm are the same. In some embodiments, the length of the 5’homology arm is different from that of the 3’homology arm.
  • the 5’homology arm is homologous to a 5’target sequence upstream of the insertion site on the genome and the 3’homology arm is homologous to a 3’target sequence downstream of the insertion site (e.g. DSB) on the genome, thereby allowing homology-directed repair to occur.
  • the 5’and/or 3’homology arms may be homologous to corresponding target sequences that is less than 200 bp away from the insertion site (e.g. DNA cleavage site) .
  • the 5’and/or 3’homology arms may be homologous to a target sequence that is 0 bp away from the DNA cleavage site.
  • the 5’target sequence and the 3’target sequence may be separated by less than 200 bp.
  • the donor plasmid is cleaved within the cell (for example by a RNA-guided endonuclease recognizing a cleavage site on the plasmid) to produce a linear nucleic acid described herein.
  • the donor plasmid may comprise flanking sequences upstream of the 5′homology arm and downstream of the 3′homology arm. Such flanking sequences in some embodiments do not exist in the genomic sequences of the host cell thus allowing cleavage to only occur on the donor plasmid.
  • the guide RNA recognizes the 5’flanking sequence and the 3’flanking sequence.
  • RNA-guided endonuclease can then be designed accordingly to effect cleavage at the 5’flanking sequence and the 3’flanking sequence under the guidance of guide RNA that allows the release of the linear nucleic acid without affecting the host sequences.
  • the flanking sequences can be, for example, about 20 to about 23 bp.
  • the method further comprises introducing a BCL inhibitor into the cell.
  • the BCL inhibitor is introduced into the eukaryotic cell in the form of a protein, or in the form of a nucleic acid encoding the BCL inhibitor, such as a messenger RNA (mRNA) , or a cDNA.
  • Nucleic acids can be delivered as part of a larger construct, such as a plasmid or viral vector, or directly, e.g., by electroporation, lipid vesicles, viral transporters, and microinjection.
  • the BCL inhibitor may be introduced into the cell by a variety of means known in the art, including transfection, calcium phosphate-DNA co-precipitation, DEAE-dextran-mediated transfection, polybrene-mediated transfection, electroporation, microinjection, transduction, cell fusion, liposome fusion, lipofection, protoplast fusion, retroviral infection, use of a gene gun, use of a DNA vector transporter, and biolistics (e.g., particle bombardment) .
  • the BCL inhibitor is directly added into medium and then is introduced into the cell by contacting the cell with the BCL inhibitor.
  • at least two BCL inhibitors are introduced into the cell.
  • the apoptosis regulator e.g., BCL-XL
  • the genome editing composition and the BCL inhibitor are introduced into the cell simultaneously.
  • at least one of the three components is introduced into the cell at a different time from the other components.
  • the BCL-XL may be introduced into the cell first, and the genome editing composition, and the BCL inhibitor are subsequently introduced.
  • the BCL-XL is introduced into the cell first, and the BCL inhibitor, and the genome editing composition are subsequently introduced.
  • all three components are introduced at a different time point relative to each other.
  • the three components can be administered in a sequence, one after another at a specific order.
  • the BCL-XL, and the genome editing composition are introduced into the cell first, and the BCL inhibitor is subsequently introduced.
  • the BCL inhibitor is introduced in to the cell such that the level of the BCL inhibitor in the cell is at least 20%, at least 50%, at least 75%, at least 1-fold, at least 2-fold, or more than 2-fold, higher than the background level of BCL inhibitor in a control cell.
  • the BCL inhibitor is introduced in to the cell such that the level of the BCL inhibitor in the cell is from 25%to 50%, from 50%to 75%, from 75%to 1-fold, or from 1-fold to 2-fold, higher than the background level of the BCL inhibitor in a control cell.
  • the BCL inhibitor is directly added into medium and then is introduced into the cell by contacting the cell with the BCL inhibitor.
  • a concentration of the BCL inhibitor in the medium ranges from about 0.5 ⁇ M to 1 ⁇ M.
  • the BCL inhibitor comprises ABT-263.
  • FIG. 1 depicts a schematic design of stable BCL-XL overexpression in human iPSCs.
  • the iPCSs are used to examine the effects of stable overexpression of BCL-XL on editing efficiency.
  • a iPSC-Lenti-BCL-XL cell line is established (FIG. 1) .
  • Lentiviral vector (Lenti-EF1-BCL-XL-E2A-Puro-Wpre) containing a BCL-XL sequence between Puro and Wpre element was constructed in the following steps.
  • the complementary DNA (cDNA) for the puromycin resistant gene (Puro) and BCL-XL were amplified by PCR and purified using KAPA HiFi polymerase (KAPA Biosystems) and a GeneJET Gel Extraction Kit (Thermo Fisher Scientific) , respectively.
  • KAPA HiFi polymerase KAPA Biosystems
  • GeneJET Gel Extraction Kit Thermo Fisher Scientific
  • iPSCs were transduced with lentiviral vectors (Lenti-EF1-BCL-XL-E2A-Puro-Wpre) at a low MOI of 0.1-0.2, and stably transduced cells were selected by culturing cells in mTeSR1 medium supplemented with 1 ⁇ g/mL puromycin for 1 week. After one week of antibiotic selection, iPSC-BCL-XL cell line expressing BCL-XL were established. As a control, iPSCs-Lenti-control cell lines were also established by transduction with the puro resistant gene only.
  • FIG. 2 depicts a schematic of HDR editing at PRDM14.
  • the following plasmids were constructed: pEF1-Cas9, pU6-sgPRDM14, pD-PRDM14-E2A-mNeonGreen-sg, and pU6-sgDocut.
  • All Cas9 plasmid (pEF1-Cas9) and sgRNA plasmid (pU6-sgPRDM14 and pU6-sgDocut) were constructed with a NEBuilder HiFi DNA Assembly Kit (New England Biolabs) .
  • PCR products were produced using KAPA HiFi polymerase (KAPA Biosystems) and purified using a GeneJET Gel Extraction Kit (Thermo Fisher Scientific) .
  • the linear PCR products were then assembled into plasmids in a DNA assembly reaction (20 ⁇ l) , on ice, according to the manufacturer’s instructions.
  • the reaction contained NEBuilder HiFi DNA Assembly Master Mix (10 ⁇ l) , equal ratios of PCR products (0.2-0.5 pmols) , and water.
  • the ligation reaction was briefly vortexed and centrifuged prior to incubation at 50°C for 5-30 min.
  • NEB 5-alpha Competent E. coli cells were then transformed with the assembled DNA products and plated on LB agar plates with ampicillin. Multiple colonies were chosen for Sanger sequencing (MCLAB) to identify the correct clones.
  • the sgDocut sequence was GGGTGCAGATGAACTTCA (SEQ ID NO: 2) .
  • the sgPRDM14 sequence was GAAGACTACTAGCCCTGCC (SEQ ID NO: 3) .
  • sgPRDM14 was designed to cut PRDM14, and sgDocut for cutting double cut donor plasmid (pD-sg, ie pD-PRDM14-E2A-mNeonGreen-sg) .
  • the double cut donor plasmid (pD-PRDM14-E2A-mNeonGreen-sg) was generated by using the NEBuilder HiFi DNA Assembly kit (New England Biolabs) as detailed above.
  • all the fragments included in a pDonor-sg (left homology arm, desired fragments for knockin, right homology arm) were amplified by PCR using KAPA HiFi polymerase (KAPA Biosystems) and purified using a GeneJET Gel Extraction Kit (Thermo Fisher Scientific) .
  • the HA sequences of ⁇ 600 bp in length were amplified from human gDNA and a sgDocut recognition sequence was added at the upstream of the left HA and downstream of the right HA. All the vectors were verified by Sanger sequencing.
  • CRISPR plasmids including pEF1-Cas9, pU6-sgPRDM14, pD-PRDM14-E2A-mNeonGreen-sg and pU6-sgDocut, were co-electroporated into iPSC-Lenti-BCL-XL lines and iPSC-Lenti-control lines, leading to the precise integration of E2A-mNeonGreen at the PRDM14 stop codon through the HDR pathway.
  • iPSCs For electroporation of human iPSCs, cells were transfected by electroporation using the Human Stem Cell Kit 2 and the program B-016, following the manufacturer’s instructions.
  • FIG. 3 depicts flow cytometry analysis of iPSC-Lenti-BCL-XL lines and iPSC-Lenti-control lines after co-transfection of CRISPR plasmids. Cell survival was determined by relative ratio of survived cell count at day 1 to cell count at electroporation.
  • FIG. 4 depicts a schematic design of transient BCL-XL overexpression in human iPSCs.
  • a plasmid pEF1-BCL-XL
  • the pEF1-BCL-XL plasmid was generated by using the NEBuilder HiFi DNA Assembly kit (New England Biolabs) as detailed above.
  • CRISPR plasmids (pEF1-Cas9, pU6-sgPRDM14, pD-PRDM14-E2A-mNeonGreen-sg and pU6-sgDocut) together with the pEF1-BCL-XL plasmid were co-transfected into iPSCs by electroporation (FIG. 4) .
  • the vector provides only transient BCL-XL overexpression, because of vector depletion after cell division and gradual silencing of plasmid in mammalian cells.
  • control group was also established by transduction with the CRISPR plasmids only (without BCL-XL) .
  • FIG. 5 depicts flow cytometry analysis of transient BCL-XL overexpression group and control group.
  • Cell survival at day 1 and HDR knockin (KI) efficiency at day 3 were detected, as detailed above.
  • KI HDR knockin
  • FIG. 5 depicts flow cytometry analysis of transient BCL-XL overexpression group and control group.
  • Cell survival at day 1 and HDR knockin (KI) efficiency at day 3 were detected, as detailed above.
  • KI HDR knockin
  • Example 3 Effects of BCL-XL on iPSC survival and HDR efficiency at the CTNNB1 or OCT4 locus in iPSCs
  • FIG. 6 depicts a schematic of HDR editing at CTNNB1.
  • FIG. 7 depicts a schematic of HDR editing at OCT4.
  • the following plasmids were constructed: pU6-sgCTNNB1, pD-CTNNB1-E2A-mNeonGreen-sg, pU6-sgOCT4 and pD-OCT4-E2A-Puro-E2A-Crimson-sg.
  • CTNNB1 is a pivotal gene in the canonical WNT pathway that is constitutively expressed in iPSCs and other cells.
  • pU6-sgCTNNB1 and pD-CTNNB1-E2A-mNeonGreen-sg were used to target at 39 bp before the stop codon and insert an E2A-mNeonGreen cassette into CTNNB1 locusA.
  • the sgCTNNB1 sequence was GCTGATTGCTGTCACCTGG (SEQ ID NO: 4) .
  • An E2A-puro-E2A-Crimson cassette was in-frame inserted before the OCT4 stop codon from a promoterless double cut HDR donor pD-OCT4-E2A-Puro-E2A-Crimson-sg, with the left HA (600bp) spanning from Intron 2 to Exon 4 and right HA (1000bp) starting from the stop codon, resulting in the replacement of Intron 4 and Exon 5 on genome with Exon5-E2A-puro-E2A-Crimson on pDonor.
  • An sgOCT4 sequence targeting OCT4 Intron 4 was GTGAGTGCCATGTCTCTCTG (SEQ ID NO: 5) .
  • CRISPR plasmids (pEF1-Cas9, pU6-sgCTNNB1, pD-CTNNB1-E2A-mNeonGreen-sg and pU6-sgDocut) and CRISPR plasmids (pEF1-Cas9, pU6-sgOCT4, pD-OCT4-E2A-Puro-E2A-Crimson-sg and pU6-sgDocut) together with the pEF1-BCL-XL plasmid were co-transfected into iPSCs by electroporation, respectively. After HDR knockin of the promoterless pDonor vector, expression of the fluorescent reporter mNeonGreen or Crimson will be driven by the endogenous CTNNB1 or OCT4 transcription machinery.
  • FIG. 8 depicts a flow cytometry analysis indicating KI efficiency at CTNNB1 and OCT4 in iPSC.
  • HDR knockin (KI) efficiency at day 3 was detected, as detailed above.
  • BCL-XL co-transfection led to a dramatic improvement in KI efficiency of 20-fold at both CTNNB1 (1.6%vs. 32%) and OCT4 (0.4%vs. 7.6%) (FIG. 8) .
  • BCL-XL increases survival of stressed iPSCs after electroporation.
  • FIG. 9 depicts a schematic of NHEJ-mediated knockout at CD326.
  • FIG. 10 depicts a schematic of NHEJ-mediated knockout at CD9.
  • the following plasmids were constructed: pU6-sgCD326 and pU6-sgCD9.
  • the sgCD9 was designed to target Exon 1 of CD326.
  • the sgCD326 sequence was GGTTCTCACTCGCTCAGAGC (SEQ ID NO: 6) .
  • the sgCD9 was designed to target Exon 2 of CD9.
  • the sgCD9 sequence was GCTTCTACACAGGTGAGGGA (SEQ ID NO: 7) .
  • CRISPR plasmids (pEF1-Cas9 and pU6-sgCD326) and CRISPR plasmids (pEF1-Cas9 and pU6-sgCD9) together with the pEF1-BCL-XL plasmid were co-transfected into iPSCs by electroporation, respectively.
  • Knockout of these 2 genes because they are expressed at high levels in human iPSCs and can be detected by antibody (anti-human CD9 FUTC (eBioscience) and anti-human CD326 PE (eBioscience) ) staining of surface markers followed with flow cytometry.
  • CD326 knockout efficiency was determined at 1 week after electroporation by staining with Anti-CD326-PE and gating PE-negative cells.
  • CD9 knockout efficiency in 6 iPSC lines was determined at 1 week after electroporation by staining with Anti-CD9-FITC and gating FITC-negative cells.
  • FIG. 11 depicts a flow cytometry analysis indicating KO efficiency at CD326 and CD9 in iPSC.
  • Significant improvement in KO efficiency by BCL-XL is observed, ⁇ 5.5-fold at CD326 (10%vs. 55%) and ⁇ 2.5-fold at CD9 (9%vs. 22%) .
  • Transient BCL-XL overexpression considerably increases both HDR knockin efficiency and NHEJ-mediated knockout efficiency.
  • FIG. 12 depicts dynamic changes in relative cell numbers after electroporation with genome editing plasmids together with or without BCL-XL.
  • pEF1-BCL2 and pEF1-MCL1 plasmids are constructed to compare with the pEF1-BCL-XL plasmid.
  • the pEF1-BCL2 and pEF1-MCL1 plasmids were generated by using the NEBuilder HiFi DNA Assembly kit (New England Biolabs) as detailed above.
  • pEF1-BCL-XL, pEF1-BCL2, pEF1-MCL1 plasmids were tested in both HDR knockin and NHEJ knockout systems by using similar method to example 2 and example 4 above.
  • control group was also established by transduction with the CRISPR plasmids only.
  • Cell survival was determined by cell count at 1 day after electroporation. KI or KO efficiency was determined at 3 or 7 days by FACS.
  • FIG. 13 depicts flow cytometry analysis of iPSCs with BCL-XL, BCL2 or MCL1.
  • BCL2 also improved survival rate ( ⁇ 5-fold) , but at lower levels compared with BCL-XL ( ⁇ 8-fold) , whereas MCL1 did not significantly increase iPSC survival after plasmid transfection.
  • BCL2 moderately increased HDR-mediated KI efficiency (0.2%vs. 7.5%, ⁇ 35-fold improvement) and NHEJ-mediated KO efficiency (9.2%vs.
  • FIG. 14 depicts expression levels of Cas9 and sgRNA.
  • a 5-to 10-fold increase in Cas9 and 3-to 6-fold increase in sgRNA levels were observed.
  • BCL-XL binds to proapoptotic counterparts BAX and BAK1, preventing the formation of lethal pores in the mitochondrial outer membrane and thereby interrupting apoptosis.
  • Human iPSCs predominantly express BAX instead of BAK1 ( ⁇ 300 vs. ⁇ 50 TPM or transcripts per million) (not shown) .
  • four BAX KO cell lines were established by targeting BAX Exon 1 or Exon 2 with Cas9-sgRNA, in two different iPSC lines. The wild type iPSCs were used as a control.
  • sgRNA targeting BAX (sgBAX-E1, sgBAX-E2) were designed. And a pD-EF1-Puro-PolyA-sg was designed and constructed. Two human iPSC lines were electroporated with plasmids of Cas9, sgRNA (targeting BAX) , pD-EF1-Puro-PolyA-sg and BCL-XL. 1 ⁇ g/ml puromycin was added into culture medium for 1 week to select stable BAX knockout cell lines.
  • sgBAX-E1 sequence was GCGGCGGTGATGGACGGGTC (SEQ ID NO: 8) .
  • sgBAX-E2 sequence was GACAGGGGCCCTTTTGCTTC (SEQ ID NO: 9) .
  • FIG. 15 depicts effects of BAX knockout on iPSC cell survival and editing efficiency.
  • BAX KO in these four lines greatly improved cell survival and HDR-mediated KI at both PDRM14 and CTNNB1 loci (FIG. 15) .
  • addition of BCL-XL significantly increased HDR efficiency, suggesting that BAX KO alone cannot replace BCL-XL.
  • BBC3 also known as PUMA, interacts with BCL2 family members, thus freeing BAX or BAK1 and inducing apoptosis.
  • four BBC3 KO cell lines were established by targeting BBC3 Exon 1 or 2 with Cas9-sgRNA, in two different iPSC lines.
  • sgRNA targeting BBC (sgBBC3-E1, sgBBC3-E2) were designed. And a pD-EF1-Puro-PolyA-sg was designed and constructed. Two human iPSC lines were electroporated with plasmids of Cas9, sgRNA (targeting BBC3) , pD-EF1-Puro-PolyA-sg and BCL-XL. 1 ⁇ g/ml puromycin was added into culture medium for 1 week to select stable BAX knockout cell lines.
  • sgBBC3-E1 sequence was GACTCACCACAAATCTGGCA (SEQ ID NO: 10) .
  • sgBBC3-E2 sequence was GTAGAGGGCCTGGCCCGCGA (SEQ ID NO: 11) .
  • FIG. 16 depicts effects of BBC3 knockout on iPSC cell survival and editing efficiency. Similar to BAX KO, BBC3 KO greatly improved cell survival and editing, but addition of BCL-XL further increased HDR efficiency at two loci (FIG. 16) , suggesting that BBC3 depletion cannot replace BCL-XL either
  • iPSCs with high copy number of CRISPR plasmids may lead to improved editing efficiency, hence enriching these cells would increase the editing efficiency in bulk population.
  • ABT-263 Navitoclax
  • iPSCs were equally split into several wells after electroporation with CRISPR plasmids (pEF1-Cas9, pU6-sgPRDM14, pD-PRDM14-E2A-mNeonGreen-sg and pU6-sgDocut) and pEF1-BCL-XL.
  • BCL inhibitor ABT-263 (Navitoclax; Selleck Chemicals) were firstly diluted in 50 ⁇ l culture medium to make a master mix, 50 ⁇ l diluted small molecules were then added evenly into each well with desired working concentration and treatment time. The medium was changed with fresh medium thereafter. A parallel well added with DMSO only (0.1%) was carried out as a control. Three days after electroporation, cells were harvested for FACS analysis to determine cell survival and editing efficiency in each condition.
  • FIG. 17 depicts effects of various dose and treatment period of ABT-263 on cell survival of iPSC after electroporation.
  • FIG. 18 depicts effects of various dose and treatment period of ABT-263 on HDR efficiency of iPSC after electroporation.
  • FIG. 19 depicts effects of ABT-263 on iPSC cell survival and editing efficiency. It can be known from FIGs. 17, 18, 19 that, with 1 ⁇ M of ABT-263, there was a 50-70%reduction in cells survival, and a 70%and 40%improvement in KI and KO editing efficiency, respectively. These results indicate that the use of BCL-XL in iPSC genome editing allows for cell selection and enrichment of successfully edited cells by administration of BCL inhibitors.
  • Example 10 Rapid high-level knockin or knockout by dual or biallelic editing
  • FIG. 20 depicts a schematic of HDR-mediated dual KI at PRDM14 and CTNNB1 in iPSCs.
  • a double cut donor pD-E2A-Puro-E2A-Crimson-sg
  • pD-E2A-Puro-E2A-Crimson-sg is designed to target the PDRM14 locus.
  • CRISPR plasmids (pD-E2A-Puro-E2A-Crimson-sg, pD-CTNNB1-E2A-mNeonGreen-sg, pU6-sgCTNNB1, pU6-sgPRDM14, pEF1-Cas9, pU6-sgDocut) together with BCL-XL plasmid were co-electroporated into iPSCs.
  • Knockin of the E2A-Puro-E2A-Crimson cassette through HDR at PRDM14 allows for puromycin selection to enrich iPSCs with HDR editing at CTNNB1. Specifically, 1 ⁇ g/ml of Puromycin was added 2 days after electroporation for selection. KI efficiency (mNeonGreenpositive) at CTNNB1 was determined by FACS.
  • FIG. 21 depicts a flow cytometry analysis indicating KI efficiency at CTNNB1 in iPSC. It can be known from FIG. 21 that, the HDR KI efficiency at CTNNB1 increased from 17%to 95%after puromycin selection.
  • FIG. 22 depicts a schematic of Dual editing at PRDM14 by HDR and CD326 by NHEJ.
  • CRISPR plasmids pD-E2A-Puro-E2A-Crimson-sg, pU6-sgCD326, pU6-sgPRDM14, pEF1-Cas9, pU6-sgDocut
  • BCL-XL plasmid were co-electroporated into iPSCs.
  • Knockin of the E2A-Puro-E2A-Crimson cassette through HDR at PRDM14 allows for puromycin selection to enrich iPSCs with NHEJ editing at CD326.
  • KO efficiency at CD326 (CD326-PE negative) was determined by FACS.
  • FIG. 23 depicts a flow cytometry analysis indicating KO efficiency at CD326 in iPSC. It can be known from FIG. 23 that, the cells with CD326 KO was enriched from 21%to 98%. These data demonstrate that a dual editing strategy can be readily used to increase HDR or KO editing efficiency to over 95%in bulk iPSCs.
  • FIG. 24 depicts a schematic for gene knockout by biallelic HDR insertion of section cassettes.
  • two double cut HDR donor pD--EF1-puro-sg and pD-EF1-zeocin-sg
  • pD--EF1-puro-sg and pD-EF1-zeocin-sg were designed with 600bp homology arms to insert puro or zeocin resistant genes at CD326, leading to biallelic disruption of the open reading frame.
  • FIG. 25 depicts a flow cytometry analysis indicating KO efficiency at CD326 in iPSC. It can be known from FIG. 25 that, a KO efficiency of 38%is observed.
  • a promoterless pDonor-mNeonGreen was designed to target the intron before the stop codon-located exon of universally expressed genes EEF1A1 and GAPDH.
  • two sgRNAs sgEef1a1-1, sgEef1a1-2 were designed to target the Eef1a1 intron.
  • sgEef1a1-1 sequence was GAGTGTAGCAGACTCAGATC (SEQ ID NO: 12) .
  • sgEef1a1-2 sequence was GTAGCAAAGATACTGATAAA (SEQ ID NO: 13) .
  • sgEEF1A1-1 and sgGAPDH were designed to target the EEF1A1 and GAPDH intron.
  • sgEEF1A1-1 sequence was GTAGTCATCCTTACCCAA (SEQ ID NO: 14) .
  • sgGAPDH sequence was GACAACTCTTTTCATCTTCT (SEQ ID NO: 15) .
  • Mouse ES cells were purchased from ATCC and maintained in Dulbecco's modified Eagle's medium (DMEM) , supplemented with 5%fetal bovine serum (Gibco) , 5%Serum replacement (Gibco) , 2 mM glutamine (Gibco) , 10 ng/ml mouse leukemia inhibitory factor (Lif) , 0.1 mM 2-mercaptoethanol (Gibco) , 3 ⁇ M GSK inhibitor (CHIR99021, Selleck) , 1 ⁇ M MEK inhibitor and 1%Penicillin/streptomycin (Invitrogen) , and seeded on 0.1%Gelatin (Sigma) coated tissue-treated 6-well plates. Cells were fed with fresh medium each day and split by 6 to 8-fold every 2-3 days. Electroporation process was performed similar to that used for human iPSCs, except that the A-013 program was used.
  • DMEM Dulbecco's modified Eagle's medium
  • Gibco 5%
  • K562 (ATCC, CCL-243) cells were grown in RPMI-1640 (VWR Life Science) with 10%FBS (Gibco) and 1%penicillin/streptomycin (Invitrogen) .
  • RPMI-1640 VWR Life Science
  • 10%FBS Gibco
  • penicillin/streptomycin Invitrogen
  • AmaxaTM Cell Line NucleofectorTM Kit V Loxza
  • Program T-016 were used, following manufacturer’s instructions.
  • Jurkat (ATCC, Clone E6-1) cells were grown in RPMI-1640 (VWR Life Science) with 10%FBS (Gibco) and 1%penicillin/streptomycin (Invitrogen) .
  • RPMI-1640 VWR Life Science
  • 10%FBS Gibco
  • penicillin/streptomycin Invitrogen
  • Amaxa TM Cell Line Nucleofector TM Kit V Loxza
  • Program X-001 were used according to manufacturer’s instructions.
  • HEK 293T cells were cultured in DMEM (Dulbecco’s modified Eagle medium, Sigma) supplemented with 10%fetal bovine serum (FBS; Gibco) and 1%penicillin/streptomycin (Invitrogen) .
  • FBS fetal bovine serum
  • penicillin/streptomycin Invitrogen
  • Mouse ESCs, K562, 293T cells were electroporated with editing plasmids with or without BCL plasmid. 293T cells were transfected by lipofection. HDR knockin efficiency, reflected by portion of mNeonGreen-positive cells, was determined 3 days after transfection by FACS.
  • FIG. 26 depicts effects of transient BCL-XL, BCL2 or MCL1 overexpression on HDR efficiency in mouse ES cells.
  • BCL-XL significantly increased HDR editing in 2 of 2 mouse ESC experiments, whereas BCL2 and MCL1 were less effective.
  • FIG. 27 depicts effects of transient BCL-XL, BCL2 or MCL1 overexpression on HDR efficiency in K562 cells.
  • BCL-XL and MCL1 had no obvious benefits, while BCL2 significantly decreased HDR efficiency at both EEF1A1 and GAPDH.
  • FIG. 28 depicts effects of transient BCL-XL, BCL2 or MCL1 overexpression on HDR efficiency in 293T cells.
  • BCL-XL and BCL2 significantly decreased HDR efficiency at both EEF1A1 and GAPDH, while MCL1 had no obvious effect.
  • FIG. 29 depicts effects of transient BCL-XL, BCL2 or MCL1 overexpression on HDR efficiency in Jurkat cells.
  • BCL-XL and MCL1 showed a trend for enhancing editing efficiency, but the differences were insignificant.

Abstract

A method of editing a target genomic DNA in a mammalian stem cell is provided. The method comprises introducing an apoptosis regulator and a genome editing composition into the mammalian stem cell to generate a modified mammalian stem cell that overexpress the apoptosis regulator, wherein the apoptosis regulator is BCL-XL, wherein the genome editing composition a genome editing endonuclease that cleaves within a desired target sequence of the genomic DNA of the mammalian stem cell and edits the target genomic DNA.

Description

METHOD OF GENOME EDITING IN MAMMALIAN STEM CELL BACKGROUND Technical Field
The invention relates to a method of genome editing in mammalian stem cell.
Description of Related Art
Human embryonic stem cells (ESCs) provide a sufficient cell source for regenerative medicine since ESCs have an unlimited self-renewal capacity, however, their derivation from the blastocyst possess ethical issues. The discovery of patient-specific induced pluripotent stem cell (iPSC) solved both the immunogenic problem associated with transplantation of allogeneic cells and the ethical concerns. Recently, considerable progress has been made to generate iPSCs, from readily available cell sources like peripheral blood, using non-integrating vectors that express reprogramming factors. However, to realize the potentials of iPSCs in regenerative medicine and disease modeling, the disease-causing genes often need to be corrected or modified prior to conducting therapy. While these approaches are generic, they often lead to the integration of the entire donor plasmid and may induce mutagenic junctions caused by erroneous NHEJ, limiting the application potentials, and therefore the development of a novel method for efficient precise gene knockin is an important current object.
Gene targeting in mouse ESCs was achieved decades ago, albeit at extremely low efficiency. Further studies led to a realization that the early success has unwittingly exploited cell intrinsic repair mechanism after DNA break. However, a naturally occurring double-stranded DNA break (DSB) surrounding a targeting locus is extremely rare, thus often limiting the targeting efficiency to levels of one in a million even with the use of homology arms extending 10 kilobase pairs long. To enhance gene targeting, tremendous effort over the past two decades has focused on creating DSBs at certain locus by targetable endonucleases. While the development of engineered endonucleases, like zinc-finger nuclease (ZFN) or transcription activator-like effector nuclease (TALEN) , have generated excitement, their limitations in design or cloning rendered them impractical for routine laboratory use. The latest generation of RNA-guided endonuclease, or CRISPR-Cas9, has been widely used due to its simplicity in vector design and robustness in performance. CRISPR-Cas9 is an adaptive immune system evolved in bacteria and archaea to identify and destroy invading agents such as bacteriophages or plasmids. The commonly used Cas9 is from streptococcus pyogenes (Sp) .
After creating a double-stranded DNA break (DSB) , cellular mechanisms repair the damage primarily through two pathways: non-homologous end joining (NHEJ) and homology-directed repair (HDR) . In the absence of a template, the NHEJ pathway is utilized, introducing variable insertions or deletions (indels) at the DSB site, which may disrupt the open reading frame of the gene and generate a knockout (KO) phenotype. This editing approach is relatively efficient and had been widely used in animal modeling, disease modeling and functional genomic research. In the presence of a donor template flanked with homology arms (HA) , the HDR pathway could be used to integrate the sequence between HAs to create precise DNA deletion, substitution or insertion, leading to the correction of diseased genes or the targeted integration of genes of interest. Unfortunately, HDR-mediated knockin is often inefficient.
The inefficiency in editing human PSCs is largely due to poor survival after manipulation. In contrast to mouse PSCs, dissassication of human PSCs into single cell suspension often induces massive cell death. The use of ROCK inhibitor  considerably increases cloning efficiency by preventing anoikis, a dissociation-induced apoptosis. However, this has solved only one problem. To precisely edit PSCs, the CRISPR components, Cas9 and sgRNA, together with a DNA donor template need to be delivered into cells. The most efficient way for vector delivery is electroporation or its improved version nucleofection, which however induces cell death.
As massive cell death, during and following nucleofection of plasmid vectors, remains a major barrier in iPSCs genome editing, and therefore the development of a novel method for genome editing in pluripotent stem cell is an important current object.
SUMMARY
The invention provides a method for efficient precise gene editing.
In one embodiment, disclosed herein is a method of editing a target genomic DNA in a mammalian stem cell is provided. The method comprises introducing an apoptosis regulator and a genome editing composition into the mammalian stem cell to generate a modified mammalian stem cell that overexpress the apoptosis regulator, wherein the apoptosis regulator is BCL-XL, wherein the genome editing composition a genome editing endonuclease that cleaves within a desired target sequence of the genomic DNA of the mammalian stem cell and edits the target genomic DNA.
In some embodiments, the BCL-XL comprises an amino acid sequence having at least 70%amino acid sequence identity to an amino acid sequence of SEQ ID NO: 1.
In some embodiments, introducing the apoptosis regulator and the genome editing composition into the mammalian stem cell may comprise: introducing the apoptosis regulator into the mammalian stem cell to generate the modified mammalian stem cell that overexpress the apoptosis regulator; and introducing the genome editing composition into the modified mammalian stem cell.
In some embodiments, the genome editing composition may comprise a RNA-guided endonuclease and a guide RNA.
In some embodiments, the RNA-guided endonuclease may be Cas9.
In some embodiments, the guide RNA may comprise a clustered regularly interspaced short palindromic repeats RNA and a tracrRNA.
In some embodiments, the genome editing composition may comprise a donor plasmid comprising a donor sequence, wherein the donor sequence is inserted into the genome at an insertion site through homology-directed repair.
In some embodiments, the mammalian stem cell may comprise an embryonic stem cell or a pluripotent stem cell.
In some embodiments, the mammalian stem cell may be an induced pluripotent stem cell.
In some embodiments, the method further may comprise introducing a BCL inhibitor into the cell.
In some embodiments, the BCL inhibitor may be ABT-263.
In some embodiments, the BCL-XL is stably expressed.
In some embodiments, the BCL-XL is transiently overexpressed.
In some embodiments, the BCL-XL is overexpressed for a period of time from about 1 hour to about 72 hour.
In some embodiments, the BCL-XL is overexpressed by at least 5-fold over background.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 depicts a schematic design of stable BCL-XL overexpression in human iPSCs.
FIG. 2 depicts a schematic of HDR editing at PRDM14.
FIG. 3 depicts flow cytometry analysis of iPSC-Lenti-BCL-XL lines and iPSC-Lenti-control lines after co-transfection of CRISPR plasmids.
FIG. 4 depicts a schematic design of transient BCL-XL overexpression in human iPSCs.
FIG. 5 depicts flow cytometry analysis of transient BCL-XL overexpression group and control group.
FIG. 6 depicts a schematic of HDR editing at CTNNB1.
FIG. 7 depicts a schematic of HDR editing at OCT4.
FIG. 8 depicts a flow cytometry analysis indicating KI efficiency at CTNNB1 and OCT4 in iPSC.
FIG. 9 depicts a schematic of NHEJ-mediated knockout at CD326.
FIG. 10 depicts a schematic of NHEJ-mediated knockout at CD9.
FIG. 11 depicts a flow cytometry analysis indicating KO efficiency at CD326 and CD9 in iPSC.
FIG. 12 depicts dynamic changes in relative cell numbers after electroporation with genome editing plasmids together with or without BCL-XL.
FIG. 13 depicts flow cytometry analysis of iPSCs with BCL-XL, BCL2 or MCL1.
FIG. 14 depicts expression levels of Cas9 and sgRNA.
FIG. 15 depicts effects of BAX knockout on iPSC cell survival and editing efficiency.
FIG. 16 depicts effects of BBC3 knockout on iPSC cell survival and editing efficiency.
FIG. 17 depicts effects of various dose and treatment period of ABT-263 on cell survival of iPSC after electroporation.
FIG. 18 depicts effects of various dose and treatment period of ABT-263 on HDR efficiency of iPSC after electroporation.
FIG. 19 depicts effects of ABT-263 on iPSC cell survival and editing efficiency.
FIG. 20 depicts a schematic of HDR-mediated dual KI at PRDM14 and CTNNB1 in iPSCs.
FIG. 21 depicts a flow cytometry analysis indicating KI efficiency at CTNNB1 in iPSC.
FIG. 22 depicts a schematic of Dual editing at PRDM14 by HDR and CD326 by NHEJ.
FIG. 23 depicts a flow cytometry analysis indicating KO efficiency at CD326 in iPSC.
FIG. 24 depicts a schematic for gene knockout by biallelic HDR insertion of section cassettes.
FIG. 25 depicts a flow cytometry analysis indicating KO efficiency at CD326 in iPSC.
FIG. 26 depicts effects of transient BCL-XL, BCL2 or MCL1 overexpression on HDR efficiency in mouse ES cells.
FIG. 27 depicts effects of transient BCL-XL, BCL2 or MCL1 overexpression on HDR efficiency in K562 cells.
FIG. 28 depicts effects of transient BCL-XL, BCL2 or MCL1 overexpression on HDR efficiency in 293T cells.
FIG. 29 depicts effects of transient BCL-XL, BCL2 or MCL1 overexpression on HDR efficiency in Jurkat cells.
DESCRIPTION OF THE EMBODIMENTS
The present invention provides a novel genome editing method with significantly higher efficiency compared to traditional genome editing methods using RNA-guided endonuclease such as CRISPR/Cas9. The method of the present application utilizes an apoptosis regulator BCL-XL. By overexpressing BCL-XL during iPSCs transfection, the survival of iPSCs could be improved after electroporation, and the HDR KI efficiency and the KO efficiency also could be improved. The improved genome editing system provides useful tools for applications ranging from manipulating human iPSC genomes to creating gene-modified animal models.
The terms “nucleic acid, ” “polynucleotide, ” and “oligonucleotide” are used interchangeably and refer to a deoxyribonucleotide or ribonucleotide polymer, in linear or circular conformation, and in either single-or double-stranded form. For the purposes of the present disclosure, these terms are not to be construed as limiting with respect to the length of a polymer. The terms can encompass known analogues of natural nucleotides, as well as nucleotides that are modified in the base, sugar and/or phosphate moieties (e.g., phosphorothioate backbones) . In general, an analogue of a particular nucleotide has the same base-pairing specificity; i.e., an analogue of A will base-pair with T.
The terms “polypeptide, ” “peptide” and “protein” are used interchangeably to refer to a polymer of amino acid residues. The term also applies to amino acid polymers in which one or more amino acids are chemical analogues or modified derivatives of a corresponding naturally-occurring amino acids.
The term “sequence” refers to a nucleotide sequence of any length, which can be DNA or RNA; can be linear, circular or branched and can be either single-stranded or double stranded.
The term “homologous nucleic acid” as used herein includes a nucleic acid sequence that is either identical or substantially similar to a known reference sequence. In one embodiment, the term “homologous nucleic acid” is used to characterize a sequence that is at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or even 100%identical to a known reference sequence.
The term "homology-directed repair (HDR) " refers to the specialized form DNA repair that takes place, for example, during repair of double-strand breaks in cells. This process requires nucleotide sequence homology, uses a "donor" molecule to template repair of a "target" molecule (i.e., the one that experienced the double-strand break) , and leads to the transfer of genetic information from the donor to the target. Homology-directed repair may result in an alteration of the sequence of the target molecule (e.g., insertion, deletion, mutation) , if the donor polynucleotide differs from the target molecule and part or all of the sequence of the donor polynucleotide is incorporated into the target DNA.
The term "non-homologous end joining (NHEJ) " refers to the repair of double-strand breaks in DNA by direct ligation of the break ends to one another without the need for a homologous template (in contrast to homology-directed repair, which requires a homologous sequence to guide repair) . NHEJ often results in the loss (deletion) of nucleotide sequence near the site of the double-strand break.
The term "stem cell" is used herein to refer to a cell {e.g., plant stem cell, vertebrate stem cell) that has the ability both to  self-renew and to generate a differentiated cell type. In the context of cell ontogeny, the adjective "differentiated" , or "differentiating" is a relative term. A "differentiated cell" is a cell that has progressed further down the developmental pathway than the cell it is being compared with.
The term "pluripotent stem cell" or "PSC" is used herein to mean a stem cell capable of producing all cell types of the organism. Therefore, a PSC can give rise to cells of all germ layers of the organism (e.g., the endoderm, mesoderm, and ectoderm of a vertebrate) . Pluripotent cells are capable of forming teratomas and of contributing to ectoderm, mesoderm, or endoderm tissues in a living organism. Pluripotent stem cells of plants are capable of giving rise to all cell types of the plant (e.g., cells of the root, stem, leaves, etc. ) . Because the term PSC refers to pluripotent stem cells regardless of their derivation, the term PSC encompasses the terms ESC and iPSC, as well as the term embryonic germ stem cells (EGSC) , which are another example of a PSC. PSCs may be in the form of an established cell line, they may be obtained directly from primary embryonic tissue, or they may be derived from a somatic cell. PSCs can be target cells of the methods described herein.
The term "induced pluripotent stem cell" or "iPSC" refers to a PSC that is derived from a cell that is not a PSC {i.e., from a cell this is differentiated relative to a PSC) . iPSCs can be derived from multiple different cell types, including terminally differentiated cells. iPSCs have an ES cell-like morphology, growing as flat colonies with large nucleo-cytoplasmic ratios, defined borders and prominent nuclei.
The term “donor sequence” as used herein refers to a nucleic acid to be inserted into the chromosome of a host cell. A donor sequence can be of any length, for example between 2 and 10,000 nucleotides in length (or any integer value therebetween or thereabove) .
Reference to “about” a value or parameter herein includes (and describes) variations that are directed to that value or parameter per se. For example, description referring to “about X” includes description of “X” .
As used herein and in the appended claims, the singular forms “a” , “or” , and “the” include plural referents unless the context clearly dictates otherwise.
The compositions and methods of the present invention may comprise, consist of, or consist essentially of the essential elements and limitations of the invention described herein, as well as any additional or optional ingredients, components, or limitations described herein or otherwise useful in a nutritional or pharmaceutical application.
The present invention provides a method for genome editing in mammalian stem cells. The method involves modifying the mammalian stem cells by overexpressing an apoptosis regulator, and introducing a genome editing composition into the mammalian stem cells. Genome editing comprises NHEJ and HDR. A genome-editing endonuclease generates a single-or double-strand break in a target genomic DNA, and the single-or double-strand break is repaired. Repair that occurs via NHEJ is sometimes referred to an "indel" (insertion or deletion) ; DNA repair via HDR is sometimes referred to as "gene correction" or "gene modification. " In some cases, editing a target genomic DNA involves generating a substitution of one or more nucleotides in the target genomic DNA, generating an edited target genomic DNA. In some cases, editing a target genomic DNA involves deletion of one or more nucleotides from the target genomic DNA, generating an edited target genomic DNA. In some cases, editing a target genomic DNA involves insertion of one or more nucleotides from the target genomic DNA, generating an edited target genomic DNA.
In some embodiments, the method comprises introducing an apoptosis regulator and a genome editing composition into the  mammalian stem cell to generate a modified mammalian stem cell that overexpress the apoptosis regulator, wherein the apoptosis regulator is BCL-XL, wherein the genome editing composition a genome editing endonuclease that cleaves within a desired target sequence of the genomic DNA of the mammalian stem cell and edits the target genomic DNA. BCL-XL, encoded by the BCL2-like 1 (BCL1L1) gene, maintains the outer mitochondrial membrane integrity and thereby prevent the release of mitochondrial contents such as cytochrome c, an apoptosis activator.
In some embodiments, BCL-XL comprises an amino acid sequence having at least 70%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%, amino acid sequence identity to the amino acid sequence of SEQ ID NO: 1.
In some embodiments, the mammalian stem cells described herein may be embryonic stem cell or a pluripotent stem cell. In some embodiments, the mammalian stem cells may be induced pluripotent stem cell. In some embodiments, the mammalian stem cells may be human induced pluripotent stem cell.
Overexpression of the BCL-XL in a mammalian stem cell can be achieved by any known method. In some cases, the BCL-XL is introduced into the mammalian stem cell in the form of a protein. In some cases, the BCL-XL is introduced into the mammalian stem cell in the form of a nucleic acid, such as a messenger RNA (mRNA) , or a cDNA. Nucleic acids can be delivered as part of a larger construct, such as a plasmid or viral vector, or directly, e.g., by electroporation, lipid vesicles, viral transporters, and microinjection. In some embodiments, the BCL-XL may be introduced into the cell by a variety of means known in the art, including transfection, calcium phosphate-DNA co-precipitation, DEAE-dextran-mediated transfection, polybrene-mediated transfection, electroporation, microinjection, transduction, cell fusion, liposome fusion, lipofection, protoplast fusion, retroviral infection, use of a gene gun, use of a DNA vector transporter, and biolistics (e.g., particle bombardment) . In some embodiments, a nucleotide sequence encoding the BCL-XL can be operably linked to a transcriptional control element (s) , e.g., a promoter, where the promoter is active in a mammalian stem cell. In some cases, the promoter is a constitutive promoter. In such case, the BCL-XL is stably expressed. In some cases, the promoter is an inducible promoter. In some embodiments, a nucleotide sequence encoding the BCL-XL is introduced into the mammalian stem cell and then inserted to the genome of the mammalian stem cell.
In some embodiments, the genome editing composition and the BCL-XL are introduced into the cell simultaneously. In some embodiments, the BCL-XL may be introduced into the cell first, and the genome editing composition is subsequently introduced. In some embodiments, introducing the apoptosis regulator (e.g., BCL-XL) and the genome editing composition into the mammalian stem cell comprising the following steps. First, the apoptosis regulator (e.g., BCL-XL) is introduced into the mammalian stem cell to generate the modified mammalian stem cell that overexpress the apoptosis regulator (e.g., BCL-XL) first. Then, the genome editing composition is introduced into the modified mammalian stem cell.
In some embodiments, the BCL-XL is constitutively overexpressed in the cell. In some embodiments, the BCL-XL is transiently overexpressed. For example, in some cases, the BCL-XL is overexpressed for a period of time of from about 1 hour to about 72 hours. For example, in some cases, the BCL-XL is overexpressed for a period of time of from about 1 hour to about 4 hours, from about 4 hours to about 8 hours, from about 8 hours to about 12 hours, from about 12 hours to about 16 hours, from about 16 hours to about 20 hours, from about 20 hours to about 24 hours, from about 24 hours to about 30 hours, from about 30 hours to about 36 hours, from about 36 hours to about 42 hours, or from about 42 hours to about 48 hours. In some cases, the BCL-XL is overexpressed for a period of time of from about 12 hour to about 48 hours. In some cases, the BCL-XL is overexpressed for a period of time of from about 12 hours to about 24 hours.
In some embodiments, the BCL-XL is introduced in to the cell such that the level of the BCL-XL in the cell is at least 2-fold, or more than 2-fold, higher than the background level of the BCL-XL in a control (unmodified) cell. For example, in some cases, the BCL-XL is introduced in to the cell such that the level of the BCL-XL in the cell is from 2-fold to 10-fold, or from 5-fold to 10-fold, higher than the background level of the BCL-XL in a control (unmodified) cell.
Overexpressing the BCL-XL in a mammalian stem cell generates a modified mammalian stem cell. Thus, the step of introducing a genome editing composition into the mammalian stem cell occurs during a period of time in which the BCL-XL is overexpressed.
In some embodiments, overexpression of the BCL-XL in the mammalian stem cell increases the survival of the mammalian stem cell when the mammalian stem cell is contacted with a genome editing composition by at least 5-fold, at least 10-fold, at least 20-fold, or more than 20-fold, compared with the survival of a control stem cell that does not overexpress the BCL-XL, and is contacted with the genome editing composition. For example, in some cases, overexpression of the BCL-XL in the mammalian stem cell increases the survival of the mammalian stem cell when the mammalian stem cell is contacted with a genome editing composition by 10-fold to 20-fold, compared with the survival of a control stem cell that does not overexpress the BCL-XL, and is contacted with the genome editing composition.
In some embodiments, overexpression of the BCL-XL in the mammalian stem cell increase HDR-mediated KI (knockin) efficiency when the mammalian stem cell is contacted with a genome editing composition by at least 10-fold, at least 20-fold, at least 50-fold, at least 100-fold, or more than 100-fold, compared with the survival of a control stem cell that does not overexpress the BCL-XL, and is contacted with the genome editing composition. For example, in some cases, overexpression of the BCL-XL in the mammalian stem cell increase HDR-mediated KI (knockin) efficiency when the mammalian stem cell is contacted with a genome editing composition by 20-fold to 100-fold, compared with the survival of a control stem cell that does not overexpress the BCL-XL, and is contacted with the genome editing composition.
In some embodiments, overexpression of the BCL-XL in the mammalian stem cell increase NHEJ-mediated KO (knockout) efficiency when the mammalian stem cell is contacted with a genome editing composition by at least 1-fold, at least 2-fold, at least 5-fold, or more than 5-fold, compared with the survival of a control stem cell that does not overexpress the BCL-XL, and is contacted with the genome editing composition. For example, in some cases, overexpression of the BCL-XL in the mammalian stem cell increase NHEJ-mediated KO (knockout) efficiency when the mammalian stem cell is contacted with a genome editing composition by 1-fold to 5-fold, compared with the survival of a control stem cell that does not overexpress the BCL-XL, and is contacted with the genome editing composition. Thus, the efficiency of genome editing of a mammalian stem cell is increased when the mammalian stem cell overexpresses the BCL-XL.
In some embodiments, the genome editing composition comprises a RNA-guided endonuclease and a guide RNA.
In some embodiments, the RNA-guided endonuclease is introduced into the eukaryotic cell in the form of a protein, or in the form of a nucleic acid encoding the RNA-guided endonuclease, such as a messenger RNA (mRNA) , or a cDNA. Nucleic acids can be delivered as part of a larger construct, such as a plasmid or viral vector, or directly, e.g., by electroporation, lipid vesicles, viral transporters, and microinjection. For example, the RNA-guided endonuclease can be introduced into the cell by a variety of means known in the art, including transfection, calcium phosphate-DNA co-precipitation, DEAE-dextran-mediated transfection, polybrene-mediated transfection, electroporation, microinjection, transduction, cell fusion, liposome fusion, lipofection, protoplast fusion, retroviral infection, use of a gene gun, use of a DNA vector transporter,  and biolistics (e.g., particle bombardment) .
In some embodiments, the nucleic acid encoding the RNA-guided endonuclease is introduced into the cell by transfection (including for example transfection through electroporation) . In some embodiments, the nucleic acid encoding the RNA-guided endonuclease is introduced into the cell by injection.
In some embodiments, the guide RNA (gRNA) can be introduced, for example, as RNA or as a plasmid or other nucleic acid vector encoding the guide RNA. The RNA-guided endonuclease binds to the gRNA and the target DNA to which the gRNA binds and cleaves the chromosome at the designed specific site. For example, the guide RNA (gRNA) can be introduced into the cell by a variety of means known in the art, including transfection, calcium phosphate-DNA co-precipitation, DEAE-dextran-mediated transfection, polybrene-mediated transfection, electroporation, microinjection, transduction, cell fusion, liposome fusion, lipofection, protoplast fusion, retroviral infection, use of a gene gun, use of a DNA vector transporter, and biolistics (e.g., particle bombardment) .
In some embodiments, the RNA-guided endonuclease is a sequence-specific nuclease. The term “sequence-specific nuclease, ” as used herein, refers to a protein that recognizes and binds to a polynucleotide at a specific nucleic acid sequence and catalyzes a double-strand break in the polynucleotide. In certain embodiments, the RNA-guided endonuclease cleaves the chromosome only once, i.e., a single double-strand break is introduced at the designed specific site during the methods described herein.
An example of a RNA-guided endonuclease system that can be used with the methods and compositions described herein includes the Cas/CRISPR system. The Cas/CRISPR (Clustered Regularly interspaced Short Palindromic Repeats) system exploits RNA-guided DNA-binding and sequence-specific cleavage of target DNA. A guide RNA (gRNA) contains about 20-25 (such as 20) nucleotides that are complementary to a target genomic DNA sequence upstream of a genomic PAM (protospacer adjacent motifs) site and a constant RNA scaffold region. In certain embodiments, the target sequence is associated with a PAM, which is a short sequence recognized by the CRISPR complex. The precise sequence and length requirements for the PAM differ depending on the CRISPR enzyme used, but PAMs are typically 2-5 bp sequences adjacent to the protospacer (that is, the target sequence) . Examples of PAM sequences are known in the art, and the skilled person will be able to identify further PAM sequences for use with a given CRISPR enzyme. For example, target sites for Cas9 from S. pyogenes, with PAM sequences NGG, may be identified by searching for 5’-Nx-NGG-3’both on an input sequence and on the reverse-complement of the input. In certain embodiments, the genomic PAM site used herein is NGG, NNG, NAG, NGGNG, or NNAGAAW. In particular embodiments, the Streptococcus pyogenes Cas9 (SpCas9) is used and the corresponding PAM is NGG. In some aspects, different Cas9 enzymes from different bacterial strains use different PAM sequences. The Cas (CRISPR-associated) protein binds to the gRNA and the target DNA to which the gRNA binds and introduces a double-strand break in a defined location upstream of the PAM site. In one aspect, the CRISPR/Cas, Cas/CRISPR, or the CRISPR-Cas system (these terms are used interchangeably throughout this application) does not require the generation of customized proteins to target specific sequences but rather a single Cas enzyme can be programmed by a short RNA molecule to recognize a specific DNA target, i.e., the Cas enzyme can be recruited to a specific DNA target using the short RNA molecule.
In some embodiments, the RNA-guided endonuclease is a type II Cas protein. In some embodiments, the RNA-guided endonuclease is Cas9, a homolog thereof, or a modified version thereof. In some embodiments, a combination of two or more  Cas proteins can be used. In some embodiments, the CRISPR enzyme is Cas9, and may be Cas9 from S. pyogenes or S. pneumoniae. In some embodiments, Cas9 is used in the methods described herein. Cas9 harbors two independent nuclease domains homologous to HNH and RuvC endonucleases, and can cut double strands of DNA to generate blunt-ended double strand breaks (DSBs) under the guidance of gRNA.
In some embodiments, a guide RNA is an RNA comprising a 5’region comprising at least one repeat from a CRISPR locus and a 3’region that is complementary to the predetermined insertion site on the chromosome. In certain embodiments, the 5’region comprises a sequence that is complementary to the predetermined cleavage site on the chromosome, and the 3’region comprises at least one repeat from a CRISPR locus. In some aspects, the 3’region of the guide RNA further comprises the one or more structural sequences of crRNA and/or tracrRNA. In some embodiments, the guide RNA comprises a crRNA and a tracrRNA, and the two pieces of RNA form a complex through hybridization.
In some embodiments, the genome editing composition further comprises a donor plasmid comprising a donor sequence. The donor plasmid can be introduced into the cell by a variety of means known in the art, including transfection, calcium phosphate-DNA co-precipitation, DEAE-dextran-mediated transfection, polybrene-mediated transfection, electroporation, microinjection, transduction, cell fusion, liposome fusion, lipofection, protoplast fusion, retroviral infection, use of a gene gun, use of a DNA vector transporter, and biolistics (e.g., particle bombardment) .
In some embodiments, the donor sequence is flanked with a 5’homology arm and a 3’homology arm, wherein the 5’homology arm is homologous to a 5’target sequence upstream of the insertion site on the genome and the 3’homology arm is homologous to a 3’target sequence downstream of the insertion site on the genome, wherein the donor sequence is inserted in to the genome at the insertion site through homology-directed repair.
In some embodiments, the insertion of the donor sequence can be evaluated using any methods known in the art. For example, a 5’primer corresponding to a sequence upstream of the 5’homology arm and a corresponding 3’primer corresponding to a region in the donor sequence can be designed to assess the 5’-junction of the insertion. Similarly, a 3’primer corresponding to a sequence downstream of the 3’homology arm and a corresponding 5’primer corresponding to a region in the donor sequence can be designed to assess the 3’-junction of the insertion.
In some embodiments, the insertion site can be at any desired site, so long as RNA-guided endonuclease can be designed to effect cleavage at such site. In some embodiments, the insertion site is at a target gene locus. In some embodiments, the insertion site is not a gene locus.
In some embodiments, the donor nucleic acid is a sequence not present in the host cell. In some embodiments, the donor sequence is an endogenous sequence present at a site other than the predetermined target site. In some embodiments, the donor sequence is a coding sequence. In some embodiments, the donor sequence is a non-coding sequence. In some embodiments, the donor sequence is a mutant locus of a gene.
In some embodiments, the size of the donor sequence can range from about 1 bp to about 100 kb. In certain embodiments, the size of the donor sequence is between about 1 bp and about 10 bp, between about 10 bp and about 50 bp, between about 50 bp and about 100 bp, between about 100 bp and about 500 bp, between about 500 bp and about 1 kb, between about 1 kb and about 10 kb, between about 10 kb and about 50 kb, between about 50 kb and about 100 kb, or more than about 100 kb.
In some embodiments, the donor sequence is an exogenous gene to be inserted into the chromosome. In some embodiments,  the donor sequence is modified sequence that replaces the endogenous sequence at the target site. For example, the donor sequence may be a gene harboring a desired mutation, and can be used to replace the endogenous gene present on the chromosome. In some embodiments, the donor sequence is a regulatory element. In some embodiments, the donor sequence is a tag or a coding sequence encoding a reporter protein and/or RNA. In some embodiments, the donor sequence is inserted in frame into the coding sequence of a target gene which will allow expression of a fusion protein comprising an exogenous sequence fused to the N-or C-terminus of the target protein.
In some embodiments, the donor plasmid described herein is cleaved within the cell to produce a linear nucleic acid. The linear nucleic acid described herein comprises a 5’homology arm, a donor sequence, and a 3’homology arm. In other words, the donor sequence is flanked with a 5’homology arm and a 3’homology arm.
In some embodiments, the homology arms are at least about 50 bp in length, for example at least about any of 50 bp, 100 bp, 200 bp, 300 bp, 600 bp, 900 bp, 1 kb, 1.5 kb, 2 kb, 4kb, 6kb, 10kb, 15kb and 20 kp in length. In some embodiments, the homology arms are at least about 300 bp in length. In certain embodiments, the homology arms may range from about 50 bp to about 2000 bp, from about 100 bp to about 2000 bp, from about 150 bp to about 2000 bp, from about 300 bp to about 2000 bp, from about 300 bp to about 1500 bp, from about 300 bp to about 1000 bp in length. In some embodiments, the length of the 5’homology arm and the length of the 3’homology arm are the same. In some embodiments, the length of the 5’homology arm is different from that of the 3’homology arm.
In some embodiments, the 5’homology arm is homologous to a 5’target sequence upstream of the insertion site on the genome and the 3’homology arm is homologous to a 3’target sequence downstream of the insertion site (e.g. DSB) on the genome, thereby allowing homology-directed repair to occur. In some embodiments, the 5’and/or 3’homology arms may be homologous to corresponding target sequences that is less than 200 bp away from the insertion site (e.g. DNA cleavage site) . In some embodiments, the 5’and/or 3’homology arms may be homologous to a target sequence that is 0 bp away from the DNA cleavage site. In some embodiments, the 5’target sequence and the 3’target sequence may be separated by less than 200 bp.
In some embodiments, the donor plasmid is cleaved within the cell (for example by a RNA-guided endonuclease recognizing a cleavage site on the plasmid) to produce a linear nucleic acid described herein. For example, the donor plasmid may comprise flanking sequences upstream of the 5′homology arm and downstream of the 3′homology arm. Such flanking sequences in some embodiments do not exist in the genomic sequences of the host cell thus allowing cleavage to only occur on the donor plasmid. The guide RNA recognizes the 5’flanking sequence and the 3’flanking sequence. RNA-guided endonuclease can then be designed accordingly to effect cleavage at the 5’flanking sequence and the 3’flanking sequence under the guidance of guide RNA that allows the release of the linear nucleic acid without affecting the host sequences. The flanking sequences can be, for example, about 20 to about 23 bp.
In some embodiments, the method further comprises introducing a BCL inhibitor into the cell. In some embodiments, the BCL inhibitor is introduced into the eukaryotic cell in the form of a protein, or in the form of a nucleic acid encoding the BCL inhibitor, such as a messenger RNA (mRNA) , or a cDNA. Nucleic acids can be delivered as part of a larger construct, such as a plasmid or viral vector, or directly, e.g., by electroporation, lipid vesicles, viral transporters, and microinjection. In some embodiments, the BCL inhibitor may be introduced into the cell by a variety of means known in the art, including transfection, calcium phosphate-DNA co-precipitation, DEAE-dextran-mediated transfection, polybrene-mediated transfection,  electroporation, microinjection, transduction, cell fusion, liposome fusion, lipofection, protoplast fusion, retroviral infection, use of a gene gun, use of a DNA vector transporter, and biolistics (e.g., particle bombardment) . In some embodiments, the BCL inhibitor is directly added into medium and then is introduced into the cell by contacting the cell with the BCL inhibitor. In some embodiments, at least two BCL inhibitors are introduced into the cell.
In some embodiments, the apoptosis regulator (e.g., BCL-XL) , the genome editing composition, and the BCL inhibitor are introduced into the cell simultaneously. In some embodiments, at least one of the three components is introduced into the cell at a different time from the other components. For example, the BCL-XL may be introduced into the cell first, and the genome editing composition, and the BCL inhibitor are subsequently introduced. In some embodiments, the BCL-XL is introduced into the cell first, and the BCL inhibitor, and the genome editing composition are subsequently introduced. In some embodiments, all three components are introduced at a different time point relative to each other. For example, the three components can be administered in a sequence, one after another at a specific order. In some embodiments, the BCL-XL, and the genome editing composition are introduced into the cell first, and the BCL inhibitor is subsequently introduced.
In some embodiments, the BCL inhibitor is introduced in to the cell such that the level of the BCL inhibitor in the cell is at least 20%, at least 50%, at least 75%, at least 1-fold, at least 2-fold, or more than 2-fold, higher than the background level of BCL inhibitor in a control cell. For example, in some cases, the BCL inhibitor is introduced in to the cell such that the level of the BCL inhibitor in the cell is from 25%to 50%, from 50%to 75%, from 75%to 1-fold, or from 1-fold to 2-fold, higher than the background level of the BCL inhibitor in a control cell.
In some embodiments, the BCL inhibitor is directly added into medium and then is introduced into the cell by contacting the cell with the BCL inhibitor. A concentration of the BCL inhibitor in the medium ranges from about 0.5 μM to 1 μM.
In some embodiments, the BCL inhibitor comprises ABT-263.
In the following, the above embodiments are described in more detail with reference to examples. However, the examples are not to be construed as limiting the scope of the invention in any sense.
Example 1: Stable BCL-XL overexpression
FIG. 1 depicts a schematic design of stable BCL-XL overexpression in human iPSCs. In this experiment, the iPCSs are used to examine the effects of stable overexpression of BCL-XL on editing efficiency. To this purpose, a iPSC-Lenti-BCL-XL cell line is established (FIG. 1) .
Lentiviral vector (Lenti-EF1-BCL-XL-E2A-Puro-Wpre) containing a BCL-XL sequence between Puro and Wpre element was constructed in the following steps. The complementary DNA (cDNA) for the puromycin resistant gene (Puro) and BCL-XL were amplified by PCR and purified using KAPA HiFi polymerase (KAPA Biosystems) and a GeneJET Gel Extraction Kit (Thermo Fisher Scientific) , respectively. The fragments of BCL-XL, E2A linker and Puro, were inserted into a lentiviral vector with the EF1 promoter using the NEBuilder HiFi DNA Assembly Kit (New England Biolabs) . All constructs were verified by Sanger sequencing (MCLAB) . Correct clones were grown in CircleGrow Medium (MP Biomedicals) and DNA plasmids were purified using Endo-Free Plasmid Maxi Kits (Qiagen) . A standard calcium phosphate precipitation protocol was used for lentivirus production. The lentiviral vectors were concentrated a 100-fold by centrifugation at 6000 g for 24 hours at 4℃ to reach biological titers of 2-10 x 10 7/ml.
iPSCs were transduced with lentiviral vectors (Lenti-EF1-BCL-XL-E2A-Puro-Wpre) at a low MOI of 0.1-0.2, and stably  transduced cells were selected by culturing cells in mTeSR1 medium supplemented with 1 μg/mL puromycin for 1 week. After one week of antibiotic selection, iPSC-BCL-XL cell line expressing BCL-XL were established. As a control, iPSCs-Lenti-control cell lines were also established by transduction with the puro resistant gene only.
The effects of stable BCL-XL overexpression are tested by creating a green fluorescent protein reporter line of PDRM14. FIG. 2 depicts a schematic of HDR editing at PRDM14. The following plasmids were constructed: pEF1-Cas9, pU6-sgPRDM14, pD-PRDM14-E2A-mNeonGreen-sg, and pU6-sgDocut.
All Cas9 plasmid (pEF1-Cas9) and sgRNA plasmid (pU6-sgPRDM14 and pU6-sgDocut) were constructed with a NEBuilder HiFi DNA Assembly Kit (New England Biolabs) . First, PCR products were produced using KAPA HiFi polymerase (KAPA Biosystems) and purified using a GeneJET Gel Extraction Kit (Thermo Fisher Scientific) . The linear PCR products were then assembled into plasmids in a DNA assembly reaction (20 μl) , on ice, according to the manufacturer’s instructions. The reaction contained NEBuilder HiFi DNA Assembly Master Mix (10 μl) , equal ratios of PCR products (0.2-0.5 pmols) , and water. The ligation reaction was briefly vortexed and centrifuged prior to incubation at 50℃ for 5-30 min. NEB 5-alpha Competent E. coli cells were then transformed with the assembled DNA products and plated on LB agar plates with ampicillin. Multiple colonies were chosen for Sanger sequencing (MCLAB) to identify the correct clones. The sgDocut sequence was GGGTGCAGATGAACTTCA (SEQ ID NO: 2) . The sgPRDM14 sequence was GAAGACTACTAGCCCTGCC (SEQ ID NO: 3) . sgPRDM14 was designed to cut PRDM14, and sgDocut for cutting double cut donor plasmid (pD-sg, ie pD-PRDM14-E2A-mNeonGreen-sg) .
The double cut donor plasmid (pD-PRDM14-E2A-mNeonGreen-sg) was generated by using the NEBuilder HiFi DNA Assembly kit (New England Biolabs) as detailed above. In short, all the fragments included in a pDonor-sg (left homology arm, desired fragments for knockin, right homology arm) were amplified by PCR using KAPA HiFi polymerase (KAPA Biosystems) and purified using a GeneJET Gel Extraction Kit (Thermo Fisher Scientific) . The HA sequences of ~600 bp in length were amplified from human gDNA and a sgDocut recognition sequence was added at the upstream of the left HA and downstream of the right HA. All the vectors were verified by Sanger sequencing.
CRISPR plasmids, including pEF1-Cas9, pU6-sgPRDM14, pD-PRDM14-E2A-mNeonGreen-sg and pU6-sgDocut, were co-electroporated into iPSC-Lenti-BCL-XL lines and iPSC-Lenti-control lines, leading to the precise integration of E2A-mNeonGreen at the PRDM14 stop codon through the HDR pathway. For electroporation of human iPSCs, cells were transfected by electroporation using the Human Stem Cell
Figure PCTCN2018101486-appb-000001
Kit 2 and the program B-016, following the manufacturer’s instructions. Since PRDM14 is actively expressed in iPSCs, its endogenous transcription machinery drives the expression of mNeonGreen, which can be quantified by FACS analysis at day 3 following electroporation (Fig. 2) . Cell survival was determined by the survived cell count at day 1 relative to cell number used for electroporation. FIG. 3 depicts flow cytometry analysis of iPSC-Lenti-BCL-XL lines and iPSC-Lenti-control lines after co-transfection of CRISPR plasmids. Cell survival was determined by relative ratio of survived cell count at day 1 to cell count at electroporation. 3%iPSC-Lenti-control cells survived at day 1, whereas 90%iPSC-Lenti-BCL-XL cells survived, suggesting that stable BCL-XL overexpression leads a 30-fold improvement in cell survival after electroporation with plasmids (Fig. 3) . KI (knockin) efficiency, which is reflected by percentage of mNeonGreen-positive cells, was determined by FACS at 3 days after electroporation. HDR efficiency at PRDM14 was increased from 0.17%in the control group to ~15%in the Lenti-BCL-XL iPSC lines, a 90-fold improvement (Fig. 3) . These results suggest that increased iPSC survival by BCL-XL leads to enhanced HDR editing in survived cells.
Example 2: Transient BCL-XL overexpression
FIG. 4 depicts a schematic design of transient BCL-XL overexpression in human iPSCs. In this experiment, a plasmid (pEF1-BCL-XL) encoding BCL-XL under the control of the EF1 promoter is constructed. The pEF1-BCL-XL plasmid was generated by using the NEBuilder HiFi DNA Assembly kit (New England Biolabs) as detailed above.
CRISPR plasmids (pEF1-Cas9, pU6-sgPRDM14, pD-PRDM14-E2A-mNeonGreen-sg and pU6-sgDocut) together with the pEF1-BCL-XL plasmid were co-transfected into iPSCs by electroporation (FIG. 4) . The vector provides only transient BCL-XL overexpression, because of vector depletion after cell division and gradual silencing of plasmid in mammalian cells. As a control, control group was also established by transduction with the CRISPR plasmids only (without BCL-XL) .
FIG. 5 depicts flow cytometry analysis of transient BCL-XL overexpression group and control group. Cell survival at day 1 and HDR knockin (KI) efficiency at day 3 were detected, as detailed above. Compared with a 3%cell survival in the control group without BCL-XL, 27%of cells survived in the transient BCL-XL overexpression group, a 9-fold increase (FIG. 5) . The increased survival led to a ~150-fold improvement in KI efficiency (0.17%vs. 25%) at the PRDM14 locus (FIG. 5) . Of note, transient BCL-XL exhibited higher editing efficiency compared to stable BCL-XL overexpression (25%vs. 15%) (FIG. 5, FIG. 3) . This may be explained by greater selection pressure after transient BCL-XL transfection, because cells transfected with greater copy numbers of BCL-XL and editing plasmids are more likely to survive the stress and have higher editing efficiency.
Example 3: Effects of BCL-XL on iPSC survival and HDR efficiency at the CTNNB1 or OCT4 locus in iPSCs
To generalize the effects of BCL-XL on iPSC survival and HDR-mediated KI, similar experiments at two more loci, CTNNB1 and OCT4 are conducted. FIG. 6 depicts a schematic of HDR editing at CTNNB1. FIG. 7 depicts a schematic of HDR editing at OCT4. The following plasmids were constructed: pU6-sgCTNNB1, pD-CTNNB1-E2A-mNeonGreen-sg, pU6-sgOCT4 and pD-OCT4-E2A-Puro-E2A-Crimson-sg.
CTNNB1 is a pivotal gene in the canonical WNT pathway that is constitutively expressed in iPSCs and other cells. pU6-sgCTNNB1 and pD-CTNNB1-E2A-mNeonGreen-sg were used to target at 39 bp before the stop codon and insert an E2A-mNeonGreen cassette into CTNNB1 locusA. The sgCTNNB1 sequence was GCTGATTGCTGTCACCTGG (SEQ ID NO: 4) .
An E2A-puro-E2A-Crimson cassette was in-frame inserted before the OCT4 stop codon from a promoterless double cut HDR donor pD-OCT4-E2A-Puro-E2A-Crimson-sg, with the left HA (600bp) spanning from Intron 2 to Exon 4 and right HA (1000bp) starting from the stop codon, resulting in the replacement of Intron 4 and Exon 5 on genome with Exon5-E2A-puro-E2A-Crimson on pDonor. An sgOCT4 sequence targeting OCT4 Intron 4 was GTGAGTGCCATGTCTCTCTG (SEQ ID NO: 5) .
In this experiment, all of the sgRNA plasmid and donor plasmids used in this experiment were generated by using the NEBuilder HiFi DNA Assembly kit (New England Biolabs) as detailed above. To further increase reproducibility, 6 different iPSC lines are used in this experiment.
CRISPR plasmids (pEF1-Cas9, pU6-sgCTNNB1, pD-CTNNB1-E2A-mNeonGreen-sg and pU6-sgDocut) and CRISPR plasmids (pEF1-Cas9, pU6-sgOCT4, pD-OCT4-E2A-Puro-E2A-Crimson-sg and pU6-sgDocut) together with the pEF1-BCL-XL plasmid were co-transfected into iPSCs by electroporation, respectively. After HDR knockin of the  promoterless pDonor vector, expression of the fluorescent reporter mNeonGreen or Crimson will be driven by the endogenous CTNNB1 or OCT4 transcription machinery.
FIG. 8 depicts a flow cytometry analysis indicating KI efficiency at CTNNB1 and OCT4 in iPSC. HDR knockin (KI) efficiency at day 3 was detected, as detailed above. Again, BCL-XL co-transfection led to a dramatic improvement in KI efficiency of 20-fold at both CTNNB1 (1.6%vs. 32%) and OCT4 (0.4%vs. 7.6%) (FIG. 8) . Based on the above, BCL-XL increases survival of stressed iPSCs after electroporation.
Example 4: Effects of BCL-XL on NHEJ-mediated knockout at CD326 and CD9 locus in iPSCs
FIG. 9 depicts a schematic of NHEJ-mediated knockout at CD326. FIG. 10 depicts a schematic of NHEJ-mediated knockout at CD9. The following plasmids were constructed: pU6-sgCD326 and pU6-sgCD9. The sgCD9 was designed to target Exon 1 of CD326. The sgCD326 sequence was GGTTCTCACTCGCTCAGAGC (SEQ ID NO: 6) . The sgCD9 was designed to target Exon 2 of CD9. The sgCD9 sequence was GCTTCTACACAGGTGAGGGA (SEQ ID NO: 7) .
CRISPR plasmids (pEF1-Cas9 and pU6-sgCD326) and CRISPR plasmids (pEF1-Cas9 and pU6-sgCD9) together with the pEF1-BCL-XL plasmid were co-transfected into iPSCs by electroporation, respectively.
Knockout of these 2 genes because they are expressed at high levels in human iPSCs and can be detected by antibody (anti-human CD9 FUTC (eBioscience) and anti-human CD326 PE (eBioscience) ) staining of surface markers followed with flow cytometry. CD326 knockout efficiency was determined at 1 week after electroporation by staining with Anti-CD326-PE and gating PE-negative cells. CD9 knockout efficiency in 6 iPSC lines was determined at 1 week after electroporation by staining with Anti-CD9-FITC and gating FITC-negative cells.
FIG. 11 depicts a flow cytometry analysis indicating KO efficiency at CD326 and CD9 in iPSC. Significant improvement in KO efficiency by BCL-XL is observed, ~5.5-fold at CD326 (10%vs. 55%) and ~2.5-fold at CD9 (9%vs. 22%) . Transient BCL-XL overexpression considerably increases both HDR knockin efficiency and NHEJ-mediated knockout efficiency.
Example 5: Dynamics analysis of cell death after electroporation with or without BCL-XL
In this experiment, live cells at 2-48 hours post-transfection are counted to investigate the dynamics of cell death after electroporation of iPSCs, with plasmids, in the absence or presence of BCL-XL.
FIG. 12 depicts dynamic changes in relative cell numbers after electroporation with genome editing plasmids together with or without BCL-XL. At ~0.5 hour after electroporation, ~50%of iPSCs in both groups survived the shock of electroporation. From 0.5 hour to 2 hour, cells from both groups became more stably attached, no additional cell death was observed (FIG. 12) . However, starting from hours 2 to 8, the vast majority of cells in control started to die. In contrast, the ever-increasing expression of BCL-XL started to protect cells from death, leading to a 60%increased survival at hour 4. Six hours later, with the accumulation of BCL-XL, cell death was not significantly increased henceforth. At 24 hours after electroporation with BCL-XL, cells started to divide, leading to an increase of relative survival rates from ~10-fold to ~20-fold compared to no BCL-XL control (FIG. 12) . These results confirm that rapid cell death occurs from 2 to 8 hours after electroporation, whereas overexpressed BCL-XL prevents cell death starting 4 hours after electroporation.
Example 6: Effects of BCL2 family on cell survival and editing
In this experiment, pEF1-BCL2 and pEF1-MCL1 plasmids are constructed to compare with the pEF1-BCL-XL plasmid. The pEF1-BCL2 and pEF1-MCL1 plasmids were generated by using the NEBuilder HiFi DNA Assembly kit (New England  Biolabs) as detailed above.
The pEF1-BCL-XL, pEF1-BCL2, pEF1-MCL1 plasmids were tested in both HDR knockin and NHEJ knockout systems by using similar method to example 2 and example 4 above. As a control, control group was also established by transduction with the CRISPR plasmids only. Cell survival was determined by cell count at 1 day after electroporation. KI or KO efficiency was determined at 3 or 7 days by FACS.
FIG. 13 depicts flow cytometry analysis of iPSCs with BCL-XL, BCL2 or MCL1. In both systems (HDR knockin and NHEJ knockout) , there were no differences in cell survival for each vector, and thus were combined for analysis. BCL2 also improved survival rate (~5-fold) , but at lower levels compared with BCL-XL (~8-fold) , whereas MCL1 did not significantly increase iPSC survival after plasmid transfection. Accordingly, BCL2 moderately increased HDR-mediated KI efficiency (0.2%vs. 7.5%, ~35-fold improvement) and NHEJ-mediated KO efficiency (9.2%vs. 37%, ~4-fold improvement) , whereas MCL1 showed no significant improvement in both KI and KO efficiency (FIG. 13) . These data demonstrate that the striking effects of BCL-XL cannot be replaced by either BCL2 or MCL1. In other words, BCL2 or MCL1 is inferior to BCL-XL in iPSC survival and editing.
Furthermore, Cas9 mRNA and sgRNA expression in iPSCs at 8 hours after transfection were measured by RT-PCR.
FIG. 14 depicts expression levels of Cas9 and sgRNA. A 5-to 10-fold increase in Cas9 and 3-to 6-fold increase in sgRNA levels were observed. These data suggest that BCL-XL or BCL2 preferentially protects cells transfected with more copies of plasmids from death, leading to higher levels of CRISPR components in survived iPSCs, which in turn improves editing efficiency.
Example 7: Effects of BCL-XL on BAX knockout cell line
BCL-XL binds to proapoptotic counterparts BAX and BAK1, preventing the formation of lethal pores in the mitochondrial outer membrane and thereby interrupting apoptosis. Human iPSCs predominantly express BAX instead of BAK1 (~300 vs. ~50 TPM or transcripts per million) (not shown) . In this experiment, four BAX KO cell lines were established by targeting BAX Exon 1 or Exon 2 with Cas9-sgRNA, in two different iPSC lines. The wild type iPSCs were used as a control.
sgRNA targeting BAX (sgBAX-E1, sgBAX-E2) were designed. And a pD-EF1-Puro-PolyA-sg was designed and constructed. Two human iPSC lines were electroporated with plasmids of Cas9, sgRNA (targeting BAX) , pD-EF1-Puro-PolyA-sg and BCL-XL. 1 μg/ml puromycin was added into culture medium for 1 week to select stable BAX knockout cell lines. sgBAX-E1 sequence was GCGGCGGTGATGGACGGGTC (SEQ ID NO: 8) . sgBAX-E2 sequence was GACAGGGGCCCTTTTGCTTC (SEQ ID NO: 9) .
FIG. 15 depicts effects of BAX knockout on iPSC cell survival and editing efficiency. BAX KO in these four lines greatly improved cell survival and HDR-mediated KI at both PDRM14 and CTNNB1 loci (FIG. 15) . However, addition of BCL-XL significantly increased HDR efficiency, suggesting that BAX KO alone cannot replace BCL-XL.
Example 8: Effects of BCL-XL on BBC3 knockout cell line
BBC3, also known as PUMA, interacts with BCL2 family members, thus freeing BAX or BAK1 and inducing apoptosis. In this experiment, four BBC3 KO cell lines were established by targeting  BBC3 Exon  1 or 2 with Cas9-sgRNA, in two different iPSC lines.
sgRNA targeting BBC (sgBBC3-E1, sgBBC3-E2) were designed. And a pD-EF1-Puro-PolyA-sg was designed and  constructed. Two human iPSC lines were electroporated with plasmids of Cas9, sgRNA (targeting BBC3) , pD-EF1-Puro-PolyA-sg and BCL-XL. 1 μg/ml puromycin was added into culture medium for 1 week to select stable BAX knockout cell lines. sgBBC3-E1 sequence was GACTCACCACAAATCTGGCA (SEQ ID NO: 10) . sgBBC3-E2 sequence was GTAGAGGGCCTGGCCCGCGA (SEQ ID NO: 11) .
FIG. 16 depicts effects of BBC3 knockout on iPSC cell survival and editing efficiency. Similar to BAX KO, BBC3 KO greatly improved cell survival and editing, but addition of BCL-XL further increased HDR efficiency at two loci (FIG. 16) , suggesting that BBC3 depletion cannot replace BCL-XL either
Example 9: Effects of BCL inhibitor ABT-263 on iPSC editing
iPSCs with high copy number of CRISPR plasmids may lead to improved editing efficiency, hence enriching these cells would increase the editing efficiency in bulk population. To preferentially deplete cells transfected with low copies of plasmids, cells were treated with ABT-263 (Navitoclax) , a potent inhibitor of BCL-XL, BCL-2 and BCL-W.
To test the effect of ABT-263, iPSCs were equally split into several wells after electroporation with CRISPR plasmids (pEF1-Cas9, pU6-sgPRDM14, pD-PRDM14-E2A-mNeonGreen-sg and pU6-sgDocut) and pEF1-BCL-XL. BCL inhibitor ABT-263 (Navitoclax; Selleck Chemicals) were firstly diluted in 50 μl culture medium to make a master mix, 50 μl diluted small molecules were then added evenly into each well with desired working concentration and treatment time. The medium was changed with fresh medium thereafter. A parallel well added with DMSO only (0.1%) was carried out as a control. Three days after electroporation, cells were harvested for FACS analysis to determine cell survival and editing efficiency in each condition.
FIG. 17 depicts effects of various dose and treatment period of ABT-263 on cell survival of iPSC after electroporation. FIG. 18 depicts effects of various dose and treatment period of ABT-263 on HDR efficiency of iPSC after electroporation.
It can be known from FIG. 17 and FIG. 18 that, adding ABT-263 (0.2μM, 0.5μM) right after electroporation, when no exogenous BCL-XL was expressed, led to sharp reduction in cell survival and no significant improvement in editing efficiency. In contrast, when ABT-263 was administered starting 8 hours, when robust BCL-XL was expressed, till 24 hours, a dose-dependent gradual decrease in cell survival and increase in editing efficiency were observed.
Furthermore, the effect of ABT-263 on KO efficiency was tested. KO efficiency was determined by a method similar to example 4. FIG. 19 depicts effects of ABT-263 on iPSC cell survival and editing efficiency. It can be known from FIGs. 17, 18, 19 that, with 1μM of ABT-263, there was a 50-70%reduction in cells survival, and a 70%and 40%improvement in KI and KO editing efficiency, respectively. These results indicate that the use of BCL-XL in iPSC genome editing allows for cell selection and enrichment of successfully edited cells by administration of BCL inhibitors.
Example 10: Rapid high-level knockin or knockout by dual or biallelic editing
FIG. 20 depicts a schematic of HDR-mediated dual KI at PRDM14 and CTNNB1 in iPSCs. In this experiment, a double cut donor (pD-E2A-Puro-E2A-Crimson-sg) , with the puro resistant gene, is designed to target the PDRM14 locus. CRISPR plasmids (pD-E2A-Puro-E2A-Crimson-sg, pD-CTNNB1-E2A-mNeonGreen-sg, pU6-sgCTNNB1, pU6-sgPRDM14, pEF1-Cas9, pU6-sgDocut) together with BCL-XL plasmid were co-electroporated into iPSCs. Knockin of the E2A-Puro-E2A-Crimson cassette through HDR at PRDM14 allows for puromycin selection to enrich iPSCs with HDR editing at CTNNB1. Specifically, 1 μg/ml of Puromycin was added 2 days after electroporation for selection. KI efficiency  (mNeonGreenpositive) at CTNNB1 was determined by FACS.
FIG. 21 depicts a flow cytometry analysis indicating KI efficiency at CTNNB1 in iPSC. It can be known from FIG. 21 that, the HDR KI efficiency at CTNNB1 increased from 17%to 95%after puromycin selection.
FIG. 22 depicts a schematic of Dual editing at PRDM14 by HDR and CD326 by NHEJ. CRISPR plasmids (pD-E2A-Puro-E2A-Crimson-sg, pU6-sgCD326, pU6-sgPRDM14, pEF1-Cas9, pU6-sgDocut) together with BCL-XL plasmid were co-electroporated into iPSCs. Knockin of the E2A-Puro-E2A-Crimson cassette through HDR at PRDM14 allows for puromycin selection to enrich iPSCs with NHEJ editing at CD326. KO efficiency at CD326 (CD326-PE negative) was determined by FACS.
FIG. 23 depicts a flow cytometry analysis indicating KO efficiency at CD326 in iPSC. It can be known from FIG. 23 that, the cells with CD326 KO was enriched from 21%to 98%. These data demonstrate that a dual editing strategy can be readily used to increase HDR or KO editing efficiency to over 95%in bulk iPSCs.
FIG. 24 depicts a schematic for gene knockout by biallelic HDR insertion of section cassettes. In this experiment, two double cut HDR donor (pD--EF1-puro-sg and pD-EF1-zeocin-sg) were designed with 600bp homology arms to insert puro or zeocin resistant genes at CD326, leading to biallelic disruption of the open reading frame.
All CRISPR plasmids (pD--EF1-puro-sg, pD-EF1-zeocin-sg, pU6-sgCD326, pEF1-Cas9, pU6-sgDocut) together with BCL-XL plasmid were co-electroporated into iPSCs. One week after electroporation, antibody staining and FACS analysis are performed. FIG. 25 depicts a flow cytometry analysis indicating KO efficiency at CD326 in iPSC. It can be known from FIG. 25 that, a KO efficiency of 38%is observed. Selection with either puromycin or zeocin increased KO efficiency to 95-96%, while double selection with both puro and zeocin led to 100%KO (FIG. 25) . These data demonstrate that knockout in virtually 100%cells can be achieved by biallelic knockin of selective cassettes.
Example 11: Differential effects of BCL2 family on editing of multiple cell lines
In this experiment, effects of BCL-XL, BCL2 and MCL1 in mouse ESCs, 293T (human embryonic kidney cells) , K562 (human erythroleukemia cells) , and Jurkat (T cell leukemia) cells are tested. In all of these lines, electroporation with BCL did not increase cell survival (data not shown) .
To facilitate detection of HDR editing, a promoterless pDonor-mNeonGreen was designed to target the intron before the stop codon-located exon of universally expressed genes EEF1A1 and GAPDH. In mouse ESCs, two sgRNAs (sgEef1a1-1, sgEef1a1-2) were designed to target the Eef1a1 intron. sgEef1a1-1 sequence was GAGTGTAGCAGACTCAGATC (SEQ ID NO: 12) . sgEef1a1-2 sequence was GTAGCAAAGATACTGATAAA (SEQ ID NO: 13) . Furthermore, in 293T, K562, and Jurkat cells, sgEEF1A1-1 and sgGAPDH were designed to target the EEF1A1 and GAPDH intron. sgEEF1A1-1 sequence was GTAGTCATCCTTACCCAA (SEQ ID NO: 14) . sgGAPDH sequence was GACAACTCTTTTCATCTTCT (SEQ ID NO: 15) .
Mouse ES cells were purchased from ATCC and maintained in Dulbecco's modified Eagle's medium (DMEM) , supplemented with 5%fetal bovine serum (Gibco) , 5%Serum replacement (Gibco) , 2 mM glutamine (Gibco) , 10 ng/ml mouse leukemia inhibitory factor (Lif) , 0.1 mM 2-mercaptoethanol (Gibco) , 3 μM GSK inhibitor (CHIR99021, Selleck) , 1 μM MEK inhibitor 
Figure PCTCN2018101486-appb-000002
and 1%Penicillin/streptomycin (Invitrogen) , and seeded on 0.1%Gelatin (Sigma) coated tissue-treated 6-well plates. Cells were fed with fresh medium each day and split by 6 to 8-fold every 2-3 days. Electroporation process was performed  similar to that used for human iPSCs, except that the A-013 program was used.
K562 (ATCC, CCL-243) cells were grown in RPMI-1640 (VWR Life Science) with 10%FBS (Gibco) and 1%penicillin/streptomycin (Invitrogen) . For electroporation of K562 cells, AmaxaTM Cell Line NucleofectorTM Kit V (Lonza) and Program T-016 were used, following manufacturer’s instructions.
Jurkat (ATCC, Clone E6-1) cells were grown in RPMI-1640 (VWR Life Science) with 10%FBS (Gibco) and 1%penicillin/streptomycin (Invitrogen) . For electroporation of Jurkat cells, Amaxa TM Cell Line Nucleofector TM Kit V (Lonza) and Program X-001 were used according to manufacturer’s instructions.
HEK 293T cells were cultured in DMEM (Dulbecco’s modified Eagle medium, Sigma) supplemented with 10%fetal bovine serum (FBS; Gibco) and 1%penicillin/streptomycin (Invitrogen) . For transfection of 293T cells, Lipofectamine 3000 (Life Technologies) was used according to manufacturer’s instructions.
Mouse ESCs, K562, 293T cells were electroporated with editing plasmids with or without BCL plasmid. 293T cells were transfected by lipofection. HDR knockin efficiency, reflected by portion of mNeonGreen-positive cells, was determined 3 days after transfection by FACS.
FIG. 26 depicts effects of transient BCL-XL, BCL2 or MCL1 overexpression on HDR efficiency in mouse ES cells. BCL-XL significantly increased HDR editing in 2 of 2 mouse ESC experiments, whereas BCL2 and MCL1 were less effective.
FIG. 27 depicts effects of transient BCL-XL, BCL2 or MCL1 overexpression on HDR efficiency in K562 cells. In K562 cells, BCL-XL and MCL1 had no obvious benefits, while BCL2 significantly decreased HDR efficiency at both EEF1A1 and GAPDH.
FIG. 28 depicts effects of transient BCL-XL, BCL2 or MCL1 overexpression on HDR efficiency in 293T cells. In 293T cells, BCL-XL and BCL2 significantly decreased HDR efficiency at both EEF1A1 and GAPDH, while MCL1 had no obvious effect.
FIG. 29 depicts effects of transient BCL-XL, BCL2 or MCL1 overexpression on HDR efficiency in Jurkat cells. In Jurkat cells, BCL-XL and MCL1 showed a trend for enhancing editing efficiency, but the differences were insignificant.
Taken together, these data indicate that BCL-XL increases HDR efficiency in mouse ESCs and BCL2 members have differential effects on gene editing in different cell lines, by mechanisms other than affecting cell survival.
It will be apparent to those skilled in the art that various modifications and variations can be made to the disclosed embodiments without departing from the scope or spirit of the disclosure. In view of the foregoing, it is intended that the disclosure covers modifications and variations provided that they fall within the scope of the following claims and their equivalents.

Claims (16)

  1. A method of editing a target genomic DNA in a mammalian stem cell, the method comprising introducing an apoptosis regulator and a genome editing composition into the mammalian stem cell to generate a modified mammalian stem cell that overexpress the apoptosis regulator,
    wherein the apoptosis regulator is BCL-XL,
    wherein the genome editing composition a genome editing endonuclease that cleaves within a desired target sequence of the genomic DNA of the mammalian stem cell and edits the target genomic DNA.
  2. The method of claim 1, wherein the BCL-XL comprises an amino acid sequence having at least 70%amino acid sequence identity to an amino acid sequence of SEQ ID NO: 1.
  3. The method of claim 1, wherein introducing the apoptosis regulator and the genome editing composition into the mammalian stem cell comprising:
    introducing the apoptosis regulator into the mammalian stem cell to generate the modified mammalian stem cell that overexpress the apoptosis regulator; and
    introducing the genome editing composition into the modified mammalian stem cell.
  4. The method of claim 1, wherein the genome editing composition comprises a RNA-guided endonuclease and a guide RNA.
  5. The method of claim 4, wherein the RNA-guided endonuclease is Cas9.
  6. The method of claim 4, wherein the guide RNA comprises a clustered regularly interspaced short palindromic repeats RNA and a tracrRNA.
  7. The method of claim 4, wherein the genome editing composition further comprises a donor plasmid comprising a donor sequence, wherein the donor sequence is inserted into the genome at an insertion site through homology-directed repair.
  8. The method of claim 1, wherein the mammalian stem cell comprises an embryonic stem cell or a pluripotent stem cell.
  9. The method of claim 1, wherein the mammalian stem cell is an induced pluripotent stem cell.
  10. The method of claim 1, wherein the mammalian stem cell is an induced pluripotent stem cell.
  11. The method of claim 1, further comprising introducing a BCL inhibitor into the cell.
  12. The method of claim 11, wherein the BCL inhibitor is ABT-263.
  13. The method of claim 1, wherein the BCL-XL is stably expressed.
  14. The method of claim 1, wherein the BCL-XL is transiently overexpressed.
  15. The method of claim 14, wherein the BCL-XL is overexpressed for a period of time from about 1 hours to about 72 hours.
  16. The method of claim 1, wherein the BCL-XL is overexpressed by at least 5-fold over background.
PCT/CN2018/101486 2018-08-21 2018-08-21 Method of genome editing in mammalian stem cell WO2020037490A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
PCT/CN2018/101486 WO2020037490A1 (en) 2018-08-21 2018-08-21 Method of genome editing in mammalian stem cell
CN201880030560.8A CN110678553B (en) 2018-08-21 2018-08-21 Method for genome editing in mammalian stem cells

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2018/101486 WO2020037490A1 (en) 2018-08-21 2018-08-21 Method of genome editing in mammalian stem cell

Publications (1)

Publication Number Publication Date
WO2020037490A1 true WO2020037490A1 (en) 2020-02-27

Family

ID=69066943

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2018/101486 WO2020037490A1 (en) 2018-08-21 2018-08-21 Method of genome editing in mammalian stem cell

Country Status (2)

Country Link
CN (1) CN110678553B (en)
WO (1) WO2020037490A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023158755A1 (en) * 2022-02-16 2023-08-24 Maxcyte, Inc. A method of cell electroporation

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112899237A (en) * 2021-01-28 2021-06-04 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) CDKN1A gene reporter cell line and construction method and application thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150132269A1 (en) * 2013-11-13 2015-05-14 Children's Medical Center Corporation Nuclease-mediated regulation of gene expression
US20150232833A1 (en) * 2012-12-17 2015-08-20 President And Fellows Of Harvard College RNA-Guided Human Genome Engineering
WO2016014794A1 (en) * 2014-07-25 2016-01-28 Sangamo Biosciences, Inc. Methods and compositions for modulating nuclease-mediated genome engineering in hematopoietic stem cells
WO2017143071A1 (en) * 2016-02-18 2017-08-24 The Regents Of The University Of California Methods and compositions for gene editing in stem cells

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11459586B2 (en) * 2016-04-19 2022-10-04 President And Fellows Of Harvard College Methods for increasing efficiency of nuclease-mediated gene editing in stem cells

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150232833A1 (en) * 2012-12-17 2015-08-20 President And Fellows Of Harvard College RNA-Guided Human Genome Engineering
US20150132269A1 (en) * 2013-11-13 2015-05-14 Children's Medical Center Corporation Nuclease-mediated regulation of gene expression
WO2016014794A1 (en) * 2014-07-25 2016-01-28 Sangamo Biosciences, Inc. Methods and compositions for modulating nuclease-mediated genome engineering in hematopoietic stem cells
WO2017143071A1 (en) * 2016-02-18 2017-08-24 The Regents Of The University Of California Methods and compositions for gene editing in stem cells

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
ROSE, J.C. ET AL.: "Rheostatic Control of Cas9-Mediated DNA Double Strand Break (DSB) Generation and Genome Editing", ACS CHEM. BIOL, vol. 13, no. 2, 12 September 2017 (2017-09-12), pages 438 - 442, XP055688345 *
WEN, W. ET AL.: "High-Level Precise Knockin of iPSCs by Simultaneous Reprogramming an( Genome Editing of Human Peripheral Blood Mononuclear Cells", STEM CELL REPORTS, vol. 10, no. 6, 5 June 2018 (2018-06-05), pages 1821 - 1834, XP055688344 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023158755A1 (en) * 2022-02-16 2023-08-24 Maxcyte, Inc. A method of cell electroporation

Also Published As

Publication number Publication date
CN110678553B (en) 2023-08-11
CN110678553A (en) 2020-01-10

Similar Documents

Publication Publication Date Title
Li et al. Highly efficient genome editing via CRISPR–Cas9 in human pluripotent stem cells is achieved by transient BCL-XL overexpression
US11884917B2 (en) Conditional CRISPR sgRNA expression
Steyer et al. Scarless genome editing of human pluripotent stem cells via transient puromycin selection
Xie et al. Seamless gene correction of β-thalassemia mutations in patient-specific iPSCs using CRISPR/Cas9 and piggyBac
US20190330603A1 (en) Crispr-cas system, materials and methods
RU2764757C2 (en) Genomic engineering
CA3040030A1 (en) Self-limiting cas9 circuitry for enhanced safety (slices) plasmid and lentiviral system thereof
Arbab et al. Cloning-free CRISPR
EP3559230B1 (en) Methods for increasing the efficiency of homology directed repair (hdr) in the cellular genome
CN109415729B (en) Gene editing reagents with reduced toxicity
WO2016061374A1 (en) Methods and compositions for generating or maintaining pluripotent cells
WO2016057951A2 (en) Crispr oligonucleotides and gene editing
US11459586B2 (en) Methods for increasing efficiency of nuclease-mediated gene editing in stem cells
US20200318140A1 (en) Methods and compositions for increasing rna activity in a cell
US20190225989A1 (en) Gene knockin method and kit for gene knockin
Giacalone et al. CRISPR‐Cas9‐based genome editing of human induced pluripotent stem cells
TW202023605A (en) Methods of achieving high specificity of genome editing
Verma et al. CRISPR/Cas-mediated knockin in human pluripotent stem cells
WO2020037490A1 (en) Method of genome editing in mammalian stem cell
Jiang et al. Robust genome and RNA editing via CRISPR nucleases in PiggyBac systems
US20230046668A1 (en) Targeted integration in mammalian sequences enhancing gene expression
TWI704224B (en) Composition and method for editing a nucleic acid sequence
Sun et al. Organoid Easytag: an efficient workflow for gene targeting in human organoids
US20230070047A1 (en) Method to enhance gene editing

Legal Events

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

Ref document number: 18930530

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18930530

Country of ref document: EP

Kind code of ref document: A1