CN113355362B - Use of chemically modified CRISPR/Cpf1 complex - Google Patents

Use of chemically modified CRISPR/Cpf1 complex Download PDF

Info

Publication number
CN113355362B
CN113355362B CN202110668136.XA CN202110668136A CN113355362B CN 113355362 B CN113355362 B CN 113355362B CN 202110668136 A CN202110668136 A CN 202110668136A CN 113355362 B CN113355362 B CN 113355362B
Authority
CN
China
Prior art keywords
lys
leu
glu
cells
ile
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202110668136.XA
Other languages
Chinese (zh)
Other versions
CN113355362A (en
Inventor
刘涛
凌鑫宇
常丽颖
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Peking University
Original Assignee
Peking University
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 Peking University filed Critical Peking University
Priority to CN202110668136.XA priority Critical patent/CN113355362B/en
Publication of CN113355362A publication Critical patent/CN113355362A/en
Application granted granted Critical
Publication of CN113355362B publication Critical patent/CN113355362B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0008Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • 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
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0645Macrophages, e.g. Kuepfer cells in the liver; Monocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0646Natural killers cells [NK], NKT 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0656Adult fibroblasts
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0684Cells of the urinary tract or kidneys
    • C12N5/0686Kidney 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0693Tumour cells; Cancer cells
    • C12N5/0694Cells of blood, e.g. leukemia cells, myeloma cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies
    • 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
    • C12N2510/00Genetically modified 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
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Cell Biology (AREA)
  • Microbiology (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Hematology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biophysics (AREA)
  • Epidemiology (AREA)
  • Physics & Mathematics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Virology (AREA)
  • Oncology (AREA)
  • Plant Pathology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Urology & Nephrology (AREA)
  • Toxicology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Rheumatology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The invention relates to the technical field of gene editing, in particular to application of a chemically modified CRISPR/Cpf1 complex. The invention discloses an application of a CRISPR/Cpf1 complex in gene editing of cells, wherein a chemically modified Cpf1 protein in the complex is coupled with crRNA and is marked as cCpf 1. The gene editing using the complex can improve the gene editing efficiency. When the antigen-binding fragment is applied to the preparation of immune cells, the preparation efficiency of CAR-T can be improved, and more importantly, the preparation efficiency of multi-site gene editing and universal CAR-T is improved.

Description

Use of chemically modified CRISPR/Cpf1 complex
Technical Field
The invention relates to the technical field of gene editing, in particular to application of a chemically modified CRISPR/Cpf1 complex.
Background
Tumors are a serious disease that afflicts human health. Due to the high complexity, diversity and variability of the biological characteristics of tumors, understanding the mechanisms underlying tumorigenesis and finding ways to treat tumors are a great challenge to scientists. In recent years, with the continuous and deep knowledge of the relationship between tumor and host, especially the anti-tumor immune response and tumor immune escape mechanism of the organism, the application of the intervention means based on immune cells, molecules and genes to tumor therapy becomes a great focus of attention of scientists and obtains exciting clinical test results. Immunotherapy of tumors has become a fourth class of anti-tumor therapies that have been demonstrated to have significant clinical therapeutic effects and advantages following surgery, radiation therapy, and chemotherapy. Research approaches (CAR-T) in which genetic engineering techniques have been adapted to enhance the specificity of T cells to enhance their killing of tumor cells have made significant progress. The therapy achieves the purpose of rapidly killing tumor cells by separating immune T cells in peripheral blood of a patient, culturing in vitro, then carrying out genetic engineering modification, namely carrying out specific genetic modification and in vitro amplification according to the tumor type suffered by the patient, and finally infusing back into the body of the patient.
Early clinical trials of CAR-T cell therapy in patients with advanced refractory leukemia and lymphoma have shown very encouraging results. One clinical trial, published by the university of pennsylvania researcher in the world journal of top medicine, new england journal of medicine, 10 months this year, showed that in 30 subjects with relapsed or refractory ALL who received CTL019 infusion (CAR-T therapy targeting CD19 antigen), 27 patients achieved complete remission 1 month after treatment, including even 15 patients who had received a bone marrow stem cell transplant. The non-recurrent survival rate at 6 months was 67% (20 people), and the overall survival rate was 78% (23 people). In addition 1 patient continued to have complete remission at 2 years follow-up. CAR-T therapy has also achieved a series of exciting advances in the treatment of solid tumors in recent years, showing good promise for immunotherapy.
Although CAR-T therapy currently has achieved an exciting set of clinical outcomes, it still carries many risks, such as over-killing, cytokine storm, inhibition of the solid tumor microenvironment, limited target selectivity, and the like. In addition, the current CAR-T preparation usually uses a virus system to integrate the exogenous CAR gene, and the integration has randomness, so that the integration position of the exogenous gene cannot be controlled, and the integration has the possible carcinogenic risk, and after the blue bird biotechnology company in the recent journal of science uses lentiviral vector to perform gene modification on blood stem cells, the modified blood stem cells are delivered to human bodies for the research of treating sickle cell diseases, and two people develop leukemia after receiving the treatment for five years and are considered to be the carcinogenic risk caused by gene random introduction. The fact that random integration caused by retrovirus has carcinogenicity is also proved in past clinical experiments, so that the development of site-directed controllable high-efficiency targeted gene integration T cells is one of the important directions for the development of future CAR-T therapy and is also a core problem mainly concerned in the field.
In recent years, gene editing technology has been developed rapidly, and is a new molecular biotechnology that artificially changes a specific gene site in a certain nucleotide sequence, inserts, deletes, replaces or modifies a specific target gene in a genome, and changes its expression trait. With the development of the technology, three generations of gene editing technologies are widely applied, namely zinc finger protein nucleases, TALEN and CRISPR technologies, wherein the CRISPR technology is developed vigorously in the last decade as a third generation gene editing technology since people find that CRISPR-Cas9 has RNA-mediated DNA endonuclease activity in 2012. Compared with the first two generations of gene editing technology, the CRISPR/Cas9 technology is easier to design and operate, has higher targeting efficiency and has unique development advantages. CRISPR-Cas systems can be divided into two classes based on the properties of the effector nucleases, one class of effector nucleases being multi-protein complexes and the second class of effector nucleases being single proteins. The two systems are divided into 6 types and 19 subtypes, and the different systems have different target substrates and cleavage mechanisms, so that the possibility is provided for the diversified application of the CRISPR-Cas system. One type of system is common in bacteria and archaea, accounting for about 90% of all established CRISPR-Cas loci, with the remaining about 10% of the two system being almost entirely present in bacteria. The second class of CRISPR-Cas systems comprises three types ii, v, and vi, with type ii nuclease Cas9 being the first effector nuclease for genome editing in mammalian cells, type v nuclease including DNA-targeting nucleases Cpf1 and Cas12b (previously referred to as Cpf1 and C2C1, respectively), and type vi nuclease including RNA-mediated RNA nucleases Cas13a and Cas13 b.
Among these, CRISPR/Cas9 is most widely used in mammalian cells, and is widely used in mammalian gene editing and immune cell preparation due to its high editing efficiency and wide genome recognition, CAR-T prepared by knocking out T cell TRAC gene and performing in situ fixed point integration into CAR gene using CRISPR/Cas9 has been in clinical stage at present, but CRISPR/Cas9 system has high off-target effect due to its own existence, and lacks evaluation of clinical long-term safety, and is still unknown for the existence of carcinogenic risk for a long time.
CRISPR/Cpf1 is the second CRISPR family enzyme found after CRISPR/Cas9 that can be applied to mammalian cell editing, because it has different preferences for genome recognition and its off-target efficiency is significantly lower than Cas9, Cpf1 becomes a powerful complement to Cas9 and a CRISPR family gene editing tool with more biomedical prospects. In general, CRISPR is used for gene editing of T cells in vitro, DNA or mRNA is introduced into the cells for editing, and then CAR-T cells are prepared by introducing exogenous donor DNA or delivering a donor through AAV, DNA delivery editing has a potential integration risk and persistent expression of the DNA may improve off-target efficiency, mRNA delivery editing methods also have an adverse effect that an expression peak is long and off-target is easily improved, compared with direct delivery of a ribonucleoprotein complex (RNP), protein and RNA are directly introduced into cells, can rapidly respond to cleavage and rapidly degrade the protein after editing is realized, the currently known most safe delivery method for gene editing is provided, and the off-target editing efficiency is significantly lower than other delivery methods, so that the method is widely applied to CAR-T cell preparation. The CRISPR-Cpf1 is reported based on an mRNA delivery system, the site-directed TRAC gene CAR-T can be prepared through systematic optimization, the multiple gene knockout CAR-T can be prepared, and the preparation by an RNP mode is a better choice but no related method is reported in documents at present. The reason for this is that the overall gene editing efficiency of Cpf1 is not ideal, the preparation efficiency of CAR-T cells using RNP complexes of Cpf1-crRNA is very low, which greatly limits the wide application of CAR-T cells in immune cells, and there is a strong need in the art to provide a method for efficiently preparing CAR-T cells by RNP delivery using improved Cpf 1.
Disclosure of Invention
In view of the above, the technical problem to be solved by the present invention is to provide the use of the chemically modified CRISPR/Cpf1 complex in immune cell gene editing.
The invention provides the use of a chemically modified CRISPR/Cpf1 complex in gene editing of a cell.
In the present invention, the cell is a stem cell, an embryonic cell, a somatic cell and/or an immune cell.
The cell of the invention is a human cell or an animal cell. The stem cells comprise hematopoietic stem cells, embryonic stem cells and mesenchymal stem cells. The animal cell is derived from model animals such as mouse, rat, guinea pig, hamster, rabbit, monkey, etc.
In the present example, the gene editing effect of mouse cells was verified by using mouse-derived embryonic cells NIH-3T3 as an example.
The verification of the gene editing effect of the human body cells comprises the verification of the human body cells and immune cells. In some embodiments, the somatic cells are 293T cells or K562 cells. The immune cells are Jurkat cells, T cells, NK cells or Macrophage cells and the like.
In the invention, the chemically modified CRISPR/Cpf1 complex consists of a Cpf1 family protein and crRNA; wherein the Cpf1 family protein is selected from AsCpf1, SpCas9, FnCpf1, CmCpf1, MbCpf1, PcCpf1 or LbCpf 1. At least one of the amino acid residues at positions 806, 834, 835, 860 and/or 1086 of said AsCpf1 protein is chemically modified.
The amino acid residue at least one of positions 3, 757, 759, 768, 785 and/or 786 of the LbCpf1 protein is chemically modified.
The chemically modified amino acid is pAcF, AcF, NAEK, p-AzF, o-AzbK, AzK, ACPK, PrpF, PrpK, CpOK, TetF, Tet1, Tet2 or Tet 3.
In the present invention, the crRNA targets an endogenous gene.
In some embodiments, the targeted endogenous gene comprises at least one of TRAC, β 2M, PD1, CTLA4, or mHPRT 1.
The invention also provides a gene editing method of a cell, which comprises the following steps: the chemically modified CRISPR/Cpf1 complex is transformed into a cell and cultured to obtain a gene-edited cell.
In the present invention, the method of transformation is electrical transformation or chemical transformation.
The gene editing of the invention is also combined with plasmid vector, lentivirus or adenovirus mediated gene transformation methods.
The invention also provides a cell prepared by the gene editing method.
The invention also provides application of the chemically modified CRISPR/Cpf1 complex in preparing a medicament for treating diseases.
The invention also provides the application of the cell prepared by the gene editing method in preparing a medicament for treating diseases.
The diseases include: immune system diseases or tumors. The treatment comprises: autologous cell therapy or allogeneic cell therapy.
The tumor is at least one of bladder cancer, blood cancer, bone cancer, brain cancer, breast cancer, central nervous system cancer, cervical cancer, colon cancer, endometrial cancer, esophageal cancer, gall bladder cancer, gastrointestinal cancer, genitourinary tract cancer, head cancer, kidney cancer, laryngeal cancer, liver cancer, lung cancer, muscle tissue cancer, neck cancer, oral or nasal mucosa cancer, ovarian cancer, pancreatic cancer, prostate cancer, skin cancer, spleen cancer, small intestine cancer, large intestine cancer, stomach cancer, testicular cancer and/or thyroid cancer.
The invention relates to a medicament for treating diseases, which comprises a chemically modified CRISPR/Cpf1 complex and/or a cell prepared by the gene editing method.
The present invention also provides a method for treating a disease, which comprises administering the cell obtained by the gene editing method of the present invention. Or editing the autoimmune cells or allogeneic immune cells with the chemically modified CRISPR/Cpf1 complex and then administering to the patient.
The invention discloses an application of a CRISPR/Cpf1 complex in gene editing of cells, wherein a chemically modified Cpf1 protein in the complex is coupled with crRNA and is marked as cCpf 1. The gene editing using the complex can improve the gene editing efficiency. When the antigen-binding fragment is applied to the preparation of immune cells, the preparation efficiency of CAR-T can be improved, and more importantly, the preparation efficiency of multi-site gene editing and universal CAR-T is improved.
Drawings
FIG. 1 shows a synthetic route to unnatural amino acid AeF;
FIG. 2 shows protein purity and activity validation of M806 modified with unnatural amino acid AeF;
FIG. 3 shows in vitro conjugation and validation of activity of unnatural amino acid AeF modified M806;
FIG. 4 shows the gene editing efficiency evaluation of the cCpf1 system;
FIG. 5 illustrates the safety evaluation of the cpcf 1 system;
FIG. 6 shows that the cpcpf 1 system improves the efficiency of precision knock-in fragments to achieve precise gene repair;
FIG. 7 shows the use of cpcf 1 in combination with AAV for the generation of CAR-Jurkat cells;
FIG. 8 shows genome level evaluation of the site-directed introduction of the CAR gene by cCpf1 into the TRAC gene;
FIG. 9 shows the application of cCpf1 to the preparation of site-directed integration of CAR-T in T cells;
FIG. 10 shows an immunological evaluation of cCpf1 for the preparation of CAR-T cells;
FIG. 11 shows the application of cCpf1 to multi-site gene knockout and knock-in for the preparation of generic CAR-T;
FIG. 12 is a schematic diagram showing that cpcf 1 improves efficiency of CAR-T production in gene editing, site-directed precise repair.
Detailed Description
The invention provides the application of the chemically modified CRISPR/Cpf1 complex, and the technical personnel can appropriately modify the technological parameters for realizing the CRISPR/Cpf1 complex by referring to the content. It is expressly intended that all such similar substitutes and modifications which would be obvious to one skilled in the art are deemed to be included in the invention. While the methods and applications of this invention have been described in terms of preferred embodiments, it will be apparent to those of ordinary skill in the art that variations and modifications in the methods and applications described herein, as well as other suitable variations and combinations, may be made to implement and use the techniques of this invention without departing from the spirit and scope of the invention.
In the invention, the chemically modified CRISPR/Cpf1 complex is prepared by performing site-specific mutation on a CRISPR family protein by using an unnatural amino acid and then coupling the CRISPR family protein with nucleic acid, and is also called as cCpf1 in the invention. Among them, CRISPR family proteins include, but are not limited to, Cas9 protein, Cpf1 protein, CasX protein, Cas phi protein, Cas12g protein, Cas13X protein, or their related inactive homologous proteins that do not have cleavage activity but retain binding activity (e.g., dCas9 protein, dCpf1 protein, or ddCpf1 protein, etc.). In the invention, the CRISPR family protein is SpCas9, and the mutation exists in one or more of the K3 position, D39 position, H41 position, H116 position, D576 position, E945 position, K1151 position and G1367 position of SEQ ID NO. 2. Alternatively, the CRISPR family protein is ascipf 1, also designated as AsCas12a, which is mutated at the following positions: one or more of positions M806, L834, S835, K860, K1086 of SEQ ID NO. 1; the CRISPR family protein is LbCpf1, also designated LbCas12a, which is mutated at the following sites: position 3, 757, 759, 768, 785 or 786 of SEQ ID NO. 3.
The unnatural amino acids have orthogonal chemical reactivity. The unnatural amino acid comprises an azide, alkynyl, aldehyde, keto, cyclopropene, or tetrazine group, and in some embodiments, is selected from the group consisting of:
Figure BDA0003117757000000041
Figure BDA0003117757000000051
in some embodiments, the unnatural amino acid is selected from the group consisting of: pAcF, AeF, PrpF, NAEK, or TetF. In the present invention, the CRISPR family protein is modified with unnatural amino acid AeF also denoted Cpf1 (M806). Namely the Cpf1 protein with the AeF modified and mutated amino acid 806.
In the present invention, the nucleic acid in the cpcpf 1 complex includes oligo DNA, donor DNA, crRNA and modified nucleic acids related thereto. In the present invention, the nucleic acid in the cpcf 1 complex is crRNA, which leads CRISPR family protein to the vicinity of the target region by base pairing with the target DNA and changes its conformation to form ribonucleoprotein complex (RNP) having DNA cleavage activity, and the activated Cas9 or Cpf1 can cleave the target DNA to form double-stranded nick (DSB). In the present invention, the crRNA targets a target gene. In the present invention, the crRNA may target any endogenous gene that needs to be knocked out and/or knocked out, in the case of constructing a gene-editing cell line or animal model. In some embodiments, the crRNA may target genes associated with immune or other functions when constructing cells for cell therapy. In some embodiments, the targeted endogenous gene comprises at least one of a T cell endogenous TCR α constant region (TRAC), T cell β 2 microglobulin (β 2M), T cell programmed death receptor 1(PD1), T cell cytotoxic T lymphocyte-associated protein 4(CTLA4), or mHPRT 1.
In the gene editing method provided by the invention, the improvement of the editing efficiency mainly depends on the CRISPR/Cpf1 complex. Therefore, the specific sequence of the crRNA is not limited in the present invention, and any crRNA sequence that meets the design rules in the art can achieve the desired effect.
The invention provides the use of a chemically modified CRISPR/Cpf1 complex in gene editing of a cell. The animal cell is derived from model animals such as mouse, rat, guinea pig, hamster, rabbit, monkey, etc. In the present example, the gene editing effect of mouse cells was verified by using mouse-derived embryonic cells NIH-3T3 as an example. The verification of the gene editing effect of the human body cells comprises the verification of the human body cells and immune cells. In some embodiments, the somatic cells are 293T cells or K562 cells. The immune cells are Jurkat cells, T cells, NK cells or Macrophage cells and the like.
The gene editing according to the present invention includes gene knock-out and/or knock-in. In the case of gene knock-out, the system of electrotransformation, which comprises only the complex of the invention, in which the crRNA is designed for the target gene. In gene knock-in, a fragment of the homology arm of the knock-in gene is provided in addition to the complex.
In embodiments of the invention, methods of gene editing are provided. In this method, a chemically modified CRISPR/Cpf1 complex is first prepared. The preparation method of the compound comprises the following steps: (1) expressing Cpf1 protein containing unnatural amino acid by using gene codon expansion technology; (2) the Cpf1 protein containing the modified crRNA at the 5' end and the unnatural amino acid was coupled by orthogonal reaction. The complex is then used to edit the cells.
In the gene editing method of the present invention, an adeno-associated virus vector-mediated gene editing means, a lentiviral vector-mediated gene editing means, and an adenoviral vector-mediated gene editing means are combined, or a plasmid vector or single-stranded/double-stranded DNA is used to introduce a target gene. Alternatively, RNA interference techniques may be combined to reduce or silence the expression of the gene of interest. For example, in embodiments of the invention, an adenoviral vector is used to integrate the CAR gene in a cell, and the immunosuppressive factor is knocked out in the cell. Immune cells for cell therapy were constructed. The method can be applied to T cells, NK cells, Macrophage cells and the like. In the examples of the present invention, T cells were used as the subjects.
The method provided by the invention can improve the gene editing efficiency, so that the obtained cells can be more effectively used for cell therapy. The cell therapy of the present invention includes the treatment of diseases of the immune system or tumors. The means of treatment include autologous cell therapy or allogeneic cell therapy. In the invention, K562 cells are edited so as to be used as a medicine for treating blood tumor diseases; editing hematopoietic stem cells as a drug for treating hematological neoplasms; the immune cells are edited to be used as a medicine for treating immune system diseases or tumors. For example, Jurkat cells are edited and used to treat tumors.
The gene editing method provided by the invention can also be used for constructing a cell line and/or an animal model. For example, a specific gene-edited mouse cell line was obtained by editing 3T3 cells.
The invention relates to a medicament for treating diseases, which comprises a chemically modified CRISPR/Cpf1 complex and/or a cell prepared by the gene editing method.
The medicine for treating diseases comprises cells prepared by the gene editing method and pharmaceutically acceptable auxiliary materials. For example, cell buffers, osmo-regulators. More specifically, the medicine may include cells and physiological saline. Albumin, hyaluronic acid, and the like may also be included.
The medicament for treating diseases comprises a chemically modified CRISPR/Cpf1 complex. The medicament also comprises reagents required in the electrotransformation process. Cell culture media and the like may also be included.
The present invention also provides a method for treating a disease, which comprises administering the cell obtained by the gene editing method of the present invention. Or editing the autoimmune cells or allogeneic immune cells with the chemically modified CRISPR/Cpf1 complex and then administering to the patient.
The present invention is directed to improving the gene editing efficiency of the Cpf1 system while broadening its efficiency of production in immune cell production such as CAR-T. The inventor considers that the affinity of Cpf1 and crRNA thereof is low, and dissociation of a complex can be caused in a cell under a low-concentration RNP form, and then the function of gene editing is lost, so the inventor realizes covalent coupling of two molecules by a strategy of non-natural amino acid site-directed coupling technology and bio-orthogonal reaction of Cpf1 and crRNA for gene editing based on a strategy of protein chemical modification, and improves the application of Cpf1 in the fields of gene editing and CAR-T by increasing the affinity to infinity. In detail, the inventor develops a mode of coupling CRISPR/Cpf1 family protein fixed points and nucleic acids through chemical covalent bonds, delivers the protein and the nucleic acids into T cells as a whole to exert the gene editing function of the T cells, and introduces a donor containing a homology arm and an insertion fragment through AAV virus to perform fixed point gene integration at a specific position of a T cell gene to efficiently obtain the fixed point integrated CAR-T cells. The inventor edits T cells by chemical coupling of Cpf1, realizes efficient preparation of CAR-T cells by using the currently safest RNP delivery mode, and solves the problem of low efficiency of site-specific integration CAR-T cell preparation in the prior art.
The test materials adopted by the invention are all common commercial products and can be purchased in the market. The reagents used in the examples include the following:
2 × Phanta Max Master Mix (Novozam), KOD one TM PCR Master Mix (TOYOBO), DpnI (New England Biolabs, NEB), spectinomycin (Inalco Pharmaceuticals), ampicillin (Inalco Pharmaceuticals), IPTG (Inalco Pharmaceuticals), DMEM medium (Macgene), RPMI-1640 medium (Macgene), X-VIVOTM15 medium (Lonza), 1 XTrysin-EDTA 0.25% (Macgene), 1XPBS (Macgene), Nickel medium (GE healthcare), c-Myc mouse monoclonal (Invitrogen), Ann-mouse IgG (H + L), F (ab')2Fragment (Alexa B)
Figure BDA0003117757000000061
647Conjugate) (Cell Signaling Technology), APC anti-human CD3(Biolegend), FITC anti-human β 2-microlobulin (Biolegend), PE anti-human CD279(Biolegend), PE/Cyanine7 anti-human CD152(Biolegend), RNA transcription kit (NEB), Cell genome extraction kit (Nozan), Total Bovine Serum (Gibco), DynabeadsTM UntouchedTMHuman T Cells Kit (Invitrogen), Ficoll-Paque PLUS (GE Healthcare), XYbeads Human CD3/CD 28T Cell Activator (SAILY BIO), T7 Endonuclase I (Norzam), ELISA Kit (Invitrogen), LDH detection Kit (Promega)
Examples the apparatus used comprises the following:
PCR apparatus (Bio-Rad), electrophoresis apparatus (Tanon), solar gel imaging system (Tanon), ultrasonication apparatus (SONICS), autoclave (STIK), water purification apparatus (Millipore), nanodrop (Thermo), table centrifuge (Thermo), sub-ultracentrifuge (Beckman), AKTA protein purification system (GE healthcare), flow cytometer (Beckmann Cytoflex), cell incubator (Thermo), microplate reader (BioTek), magnetic stand (Invitrogen), cell electro-transport apparatus (Celetrix).
The invention is further illustrated by the following examples:
example 1: construction of Gene vector comprising site-directed mutagenesis AsCpf1
(1) Acquisition of helper plasmids
pUltra-MjPolyRS (available from Addgene, Inc.) (hereinafter referred to as helper plasmid) which expresses tRNA and tRNA synthetase for specifically recognizing and inserting unnatural amino acid AeF.
(2) Obtaining of plasmid of AsCpf1
The protein sequence in the pET22b-AsCpf1 plasmid is obtained from Addgene plasmid #102565 through PCR, then 5 'NdeI and 3' XhoI enzyme digestion are introduced into the tail end of the PCR primer, and the complete plasmid sequence is constructed through enzyme digestion and connection, so that the AsCpf1 protein can be expressed in Escherichia coli in a recombinant mode.
The primer sequences used are shown in table 1:
TABLE 1
Primer name Primer sequence (5 '-3')
Cpf1-2NLS-FW gggaattgtgagcggataacaattcccc
SV40-RV cccactttacgtttctttttaggactgccctttttcttttttgcctggccgg
Cpf1-2NLS-RV cagtggtggtggtggtggtgctcgagggatcccactttacgtttctttttaggactgc
(3) Obtaining of AsCpf1 mutant plasmid
According to the analysis result of the AsCpf1 protein crystal structure, the inventor selects M806 site as target amino acid of site-directed mutagenesis, replaces original amino acid residue with unnatural amino acid AeF containing azide group, and then can take the former amino acid residue as raw material to react with DBCO modified crRNA in vitro to prepare covalent modified Cpf1-crRNA coupling complex (cCpf1), so as to solve the problem of low affinity of Cpf1 and the crRNA thereof and further improve the editing efficiency of Cpf1 system in T cells.
The inventor designs a primer capable of mutating a corresponding codon for coding the amino acid into TAG aiming at M806 position of AsCpf1, wherein the specific primer is shown in the following table.
Table 2: list of mutant primers
Primer name Primer sequence (5 '-3')
M806TAG-FW accggctgggagagaagtagctgaacaagaagctgaagg
M806TAG-RV ccttcagcttcttgttcagctacttctctcccagccggt
Using pET22b-AsCpf1 plasmid as a template, performing 18-cycle PCR amplification by using the primers and KOD enzyme, performing DpnI digestion on the obtained PCR product, transforming the obtained product into DH5 alpha strain, recovering the strain, coating the recovered strain on a plate with corresponding resistance, and picking single clone for sequencing the next day to obtain pET22b-AsCpf1-M806TAG plasmid
(4) Construction of site-directed mutated AsCpf1-M806TAG expressing Strain
And (2) transferring the helper plasmid pUltra-MjPolyRS (spectinomycin resistance) obtained in the step (1) and the pET22b-AsCpf1-M806TAG mutant plasmid (ampicillin resistance) obtained in the step (3) into Escherichia coli Ecoli. BL21(DE3) together in a chemical transformation mode, screening out a positive strain which is simultaneously transformed by two plasmids through a double-resistance plate (spectinomycin and ampicillin), and naming the positive strain as an expression strain BL21-pET22b-AsCpf1-M806 TAG. Meanwhile, a wild-type expression strain BL21-pET22b-AsCpf1-WT (screened by ampicillin resistance) was constructed as a control.
Example 2: expression purification and in vitro activity validation of chemically modified Cpf1(AzCpf1)
1: amino acids were synthesized by the inventors
The synthetic route is shown in figure 1, and the synthetic procedures are performed according to the literature.
2: expression of chemically modified Cpf1(AzCpf1) protein
The expression strain BL21-pET22b-AsCpf1-M806TAG obtained in step (4) of example 1 was cultured overnight in 2YT medium (containing spectinomycin and ampicillin) in a constant temperature shaker at 37 ℃ and 220 rpm. The next day, inoculating to fresh 2YT culture medium (containing spectinomycin and ampicillin) according to a ratio of 1:100, culturing in a constant temperature shaking table at 37 ℃ and 220rpm, adding 1mM AeF amino acid for continuous culture when the culture reaches early logarithmic growth, adding 0.2mM IPTG (Isopropyl thiogalactoside, Isopropyl beta-D-1-thiogalactopyranoside) when the culture reaches an OD value of 0.6-1.0, and collecting thalli after induced expression in a constant temperature shaking table at 18-20 ℃ and 220rpm for 16-18 hours. The positive control used in this expression experiment was strain BL21-pET22b-AsCpf1-WT, and the expression conditions were the same as for the mutant strain.
3: purification of chemically modified Cpf1(AzCpf1) proteins
1) Resuspending the collected thallus in step 2 with buffer (50mM Tris, pH8.0, 1M KCl), and ultrasonicating for 15min
2) Carrying out high-speed centrifugation on the ultrasonication product obtained in the step 1), and taking soluble protein in supernatant for protein purification
3) Purifying the supernatant obtained in the step 2) by using a Ni affinity column, and eluting by using high-concentration imidazole.
4) Displacing the purified product in the step 3) into a buffer PBS, further purifying by size exclusion chromatography, and collecting the target protein component by using a Superdex 20010/300 GL Incrase (GE healthcare) molecular sieve.
Through the purification, the wild-type and mutant proteins of AsCpf1 with SDS-PAGE grade purity of more than 95% can be obtained, and the results are shown in A in FIG. 2.
4: in vitro activity validation of chemically modified Cpf1(AzCpf1) protein
In order to verify whether the AzCpf1 purified in step 3 has the function of cutting double-stranded DNA under the guidance of CrRNA, the inventors verified the in vitro activity of AzCpf1 protein.
1) Obtaining double-stranded DNA template by in vitro activity verification
EGFP gene is used as a cutting target gene, and an EGFP gene segment containing a cutting target sequence is amplified through PCR and is used as an in vitro activity verification template. The primers used were as follows:
table 3:
primer name Primer sequence (5 '-3')
IVD-EGFP-FW catctgcaccaccggcaagc
IVD-EGFP-RV gacggcgctattcagatcctcttctgag
2) In vitro Activity verification CrRNA acquisition
CrRNA targeting the EGFP gene was obtained by in vitro transcription. Firstly, designing a complementary primer of a T7 promoter + CrRNA sequence, obtaining an in vitro transcription template in a primer gradient annealing mode, further transcribing the CrRNA by using an in vitro transcription kit, and purifying the CrRNA by using an RNA purification kit (Zymo).
The EGFP targeting sequence was cgtcgccgtccagctcgaccagg, and the complementary primer sequences used were as follows:
table 4:
name of primer Primer sequence (5 '-3')
Cpf1-crRNA-FW taatacgactcactataggaatttctactcttgtagatcgtcgccgtccagctcgaccagg
Cpf1-crRNA-RV cctggtcgagctggacggcgacgatctacaagagtagaaattcctatagtgagtcgtatta
The template DNA sequence is as follows:
catctgcaccaccggcaagctgcccgtgccctggcccaccctcgtgaccaccctgacctacggcgtgcagtgcttcagccgctaccccgaccacatgaagcagcacgacttcttcaagtccgccatgcccgaaggctacgtccaggagcgcaccatcttcttcaaggacgacggcaactacaagacccgcgccgaggtgaagttcgagggcgacaccctggtgaaccgcatcgagctgaagggcatcgacttcaaggaggacggcaacatcctggggcacaagctggagtacaactacaacagccacaacgtctatatcatggccgacaagcagaagaacggcatcaaggtgaacttcaagatccgccacaacatcgaggacggcagcgtgcagctcgccgaccactaccagcagaacacccccatcggcgacggccccgtgctgctgcccgacaaccactacctgagcacccagtccgccctgagcaaagaccccaacgagaagcgcgatcacatggtcctgctggagttcgtgaccgccgccgggatcactctcggcatggacgagctgtacaagtaagaattccaccacactggactagtggatccgagctcggtaccaagctttctagaacaaaaactcatctcagaagaggatctgaatagcgccgtc
the gradient annealing procedure used is shown in table 5:
table 5: primer gradient annealing program
Temperature of Time
95℃ 5min
95-85℃ -2℃/sec
85-25℃ -0.1℃/sec
4℃
3) Activity verification of AsCpf1 protein
Respectively diluting the wild-type protein and the mutant protein of AsCpf1 to 3 mu M, diluting CrRNA to 9 mu M, preparing a ribonucleoprotein complex according to the molar ratio of Cpf1 to CrRNA of 1:3, and preparing an in vitro cleavage system according to the table 3. The reaction system is then incubated at 37 ℃ for 1 hour and heat inactivated at 65 ℃ for 10min, and the reaction products are finally separated by electrophoresis on 1% agarose gel, and the result is shown in FIG. 2B, where AzCpf1 has cleavage activity consistent with WT Cpf1, which shows that the insertion of unnatural amino acids does not affect the recognition and cleavage activity of the protein on double-stranded DNA.
Table 6: in vitro cleavage reaction system
Figure BDA0003117757000000091
Example 3: covalent coupling and Activity evaluation of AzCpf1 with DBCO-modified RNA molecules
After introduction of AeF at the M806 site of ascipf 1, the azide group in AeF was covalently coupled to a DBCO-modified nucleic acid molecule by a copper-free click reaction, as demonstrated below by covalent coupling of AzCpf1 to a DBCO-modified RNA molecule and cleavage activity in vitro after coupling. In this validation, the EGFP target sequence was still cleaved as a double-stranded DNA template.
1: synthesis of DBCO-modified CrRNA
The WT-EGFP CrRNA and the DBCO-EGFP CrRNA of which the 5' -end is modified with a DBCO group are synthesized by Anhui general bio-corporation.
2: covalent coupling of AzCpf1 to DBCO-modified RNA molecules
Protein and CrRNA are mixed according to a molar ratio of 1:1.2, SDS-PAGE detection is carried out after 3-hour reaction at 4 ℃, and simultaneously, a ribonucleoprotein complex (RNP) after reaction is mixed and incubated with template double-stranded DNA in a buffer system according to the step 4 of the example 2 so as to verify the in vitro activity of the coupled complex. The results are shown in FIG. 3. According to the experimental result, the AzCpf1 and DBCO-CrRNA can realize almost complete covalent coupling at 4 ℃ within 3 hours, and the in vitro activity of the ribonucleoprotein complex after coupling is not influenced, so that the recognition and cleavage activity of the AsCpf1 protein on double-stranded DNA is not influenced by the insertion of unnatural amino acid AeF and the coupling of CrRNA. The Cpf1-CrRNA complex after point-mapping of CrRNA was designated as cCpf1(conjugated Cpf 1).
Example 4: in vivo gene editing efficiency evaluation of site-directed conjugation of cpcpf 1
The present inventors have selected four different cell lines and various target genes to assess the in vivo gene efficiency of site-directed conjugation of cpcpf 1.
1) The in vivo gene editing efficiency of 293T cells is evaluated by taking 293T-EGFP-Teton as a reporter cell line, and the specific process is as follows:
firstly, preparing a cCpf1 complex in vitro, preparing an RNP complex according to a Cpf1: CrRNA molar ratio of 1:1.2(60pmol:72pmol), incubating Cpf1 protein and CrRNA at room temperature for 5 minutes, placing at 4 ℃ for reaction for 3 hours to prepare cCpf1, performing cell electroporation after the reaction is finished, and preparing a wild-type WT Cpf1 complex as a control by the same method. Adherent 293T cells were aspirated off medium, washed once with PBS, digested with pancreatin, terminated with medium and counted in suspension. Take 1X 106Suspending the cells with 20 μ L electrotransfer buffer (Celetrix, used after mixing solution A and solution B in equal proportion before use), mixing RNP and cells, slowly adding into 20 μ L electric shock cup, performing electric shock according to optimized parameters (420V, 30ms, 1pulse), rapidly adding cells into preheated DMEM culture medium after electric shock is finished, and culturing in incubator (37 deg.C, 5% CO)2). After 24 hours, doxycycline hydrochloride solution was added to a final concentration of 10 μ g/ml. After 48 hours, the cells were digested and suspended, washed once with PBS and then subjected to flow analysis. The edited cells cannot generate EGFP fluorescence under the induction of DOX due to the cutting and the destruction of the EGFP gene, while the unedited cells can generate fluorescence, and the result of analyzing the flow type result is shown as A in FIG. 4, which shows that the cutting efficiency of the EGFP is improved from-20% to-70% by the conjugated cCpf1 compared with that of WT Cpf 1.
2) Evaluating the in vivo gene editing efficiency of K562 cells, and editing the gene locus of the targeted endogenous gene hHPRT1 by the following specific process:
firstly, preparing a cCpf1 complex in vitro, preparing an RNP complex according to a Cpf1: CrRNA molar ratio of 1:1.2(60pmol:72pmol), incubating Cpf1 protein and CrRNA at room temperature for 5 minutes, placing at 4 ℃ for reaction for 3 hours to prepare cCpf1, performing cell electroporation after the reaction is finished, and preparing a wild-type WT Cpf1 complex as a control by the same method. Counting K562 cells, 1.5X 106The cells were suspended in 20. mu.L of an electrotransfer buffer (Celetrix, used after mixing solution A and solution B at equal ratio before use), and RNP was mixed with the cells and then gradually suspendedSlowly adding into a 20 μ L electric shock cup, performing electric shock according to optimized parameters (380V, 30ms, 1pulse), rapidly adding cells into preheated RPMI-1640 culture medium after electric shock is finished, and culturing in an incubator (37 deg.C, 5% CO)2). Cells were harvested after 48 hours, and cell genome extraction was performed using a cell genome extraction kit (nuozin). Then, the extracted genome is used as a template, a locus fragment containing a target site is obtained by PCR amplification, and the gene editing efficiency is detected by a T7E1 mode. 200ng of the PCR product fragment was subjected to gradient annealing in T7E1 reaction buffer (Novozam), the procedure is shown in Table 2, T7E1 enzyme was added after the annealing was completed, the cleavage band was detected by agarose gel after incubation at 37 ℃ for 30 minutes, and the cleavage band was subjected to computational analysis by Tanon Gis. The results are shown in fig. 4, B, which shows that cpcf 1 can increase the genome cleavage efficiency from-10% to 44% compared to WT cpcf 1 after conjugation, and exhibits greater therapeutic potential for leukemia. Gene editing efficiency was 100 × (1-amount of cleaved band DNA/amount of total DNA in lane). The sequences of CrRNA used and primers used for genomic PCR are shown below:
TABLE 7
Name (R) Sequence (5 '-3')
hHPRT1 CrRNA gguuaaagaugguuaaaugau
Geno-hHPRT1-FW catggtacactcagcacggatgaaatg
Geno-hHPRT1-RV gctgttcaactatttcagccaacaagaagtg
3) The evaluation of the in vivo gene editing efficiency of the jurkat cell and the editing of the gene locus of the targeted endogenous gene hHPRT1 are the same as those of the K562 cell except that the specific process is different from the electrotransfer condition (the electrotransfer condition of the jurkat cell is 400V, 30ms and 1 pulse). The results of the evaluation of gene editing efficiency in vivo on jurkat cells are shown in fig. 4, C, and indicate that cpcf 1 showed twice the editing efficiency after conjugation compared to WT Cpf1, showing better potential for efficient T cell production.
4) The in vivo gene editing efficiency evaluation of NIH-3T3 cells, targeted endogenous gene mHPRT1 locus for editing, the specific process is as follows:
firstly, preparing a cCpf1 complex in vitro, preparing an RNP complex according to a Cpf1: CrRNA molar ratio of 1:1.2(60pmol:72pmol), incubating Cpf1 protein and CrRNA at room temperature for 5 minutes, placing at 4 ℃ for reaction for 3 hours to prepare cCpf1, performing cell electroporation after the reaction is finished, and preparing a wild-type WT Cpf1 complex as a control by the same method. Adherent NIH-3T3 cells were aspirated off the medium, washed once with PBS, digested with pancreatin, terminated with medium and counted in suspension. Take 1X 106Suspending the cells with 20 μ L electrotransfer buffer (Celetrix, used after mixing solution A and solution B in equal proportion before use), mixing RNP and cells, slowly adding into 20 μ L electric shock cup, performing electric shock according to optimized parameters (400V, 30ms, 1pulse), rapidly adding cells into preheated DMEM culture medium after electric shock is finished, and culturing in incubator (37 deg.C, 5% CO)2). Cells were harvested after 48 hours, and cell genome extraction was performed using a cell genome extraction kit (nuozin). The genome editing efficiency was detected and evaluated in accordance with step 2), and the editing efficiency was improved from-8% to-25% as shown in D in FIG. 4. The sequences of CrRNA used and primers used for genomic PCR are shown below:
TABLE 8
Name (R) Sequence (5 '-3')
Mouse HPRT1 CrRNA ggauguuaagagucccuaucu
Geno-mHPRT1-FW gttcaattcccagcaaccacatgttg
Geno-mHPRT1-RV gtatacacaaaatctcaccacaattcacacagag
Example 5: in vivo off-target efficiency assessment of site-directed conjugation of cpcf 1
The invention adopts the currently accepted strategy of Guide-seq and the strategy of PEM-seq reported in recent years to evaluate the safety of CRISPR/cCpf1 system gene editing. The Guide-seq is evaluated by drumhead medical treatment limited, the tracing of gene editing cutting sites is carried out by adding dsODN through homologous recombination, the enrichment of the cutting sites is realized by utilizing dsODN specific amplification, the recognition of the cutting sites is realized by utilizing a second-generation sequencing platform, and then the positions and the number of target sites and off-target sites are analyzed. Off-target sequencing results as shown in a in fig. 5, it can be seen that 1359 editing sites were detected in the target sequence and one off-target site was generated for wild-type Cpf1, whereas 2816 editing sites were detected in the target sequence and no off-target site was detected in the panel conjugated Cpf1, indicating that the cppf 1 system can improve gene editing efficiency without generating additional off-target effects.
To further detect possible chromosomal rearrangements or additional large fragment deletions by CRISPR/cpcpf 1, the inventors used PEM-Seq protocol to test the cleavage and amplify the sequence with specific primers as follows, followed by sequencing through a next-generation sequencing platform. Off-target sequencing results as shown in B in fig. 5, off-target at 1 site was detected using both wild-type Cpf1 and Cpf1, and no additional large deletion insertions or deletions were detected, indicating the safety and efficacy of the cppf 1 system.
Example 6: site-directed conjugation of AsCpf1 to crRNA evaluation of the level of homologous recombination in cells
In the embodiment, conserved gene HPRT genes in human cells are cut, 6 bp specific sequences containing 40bp homologous arms at the left and right are introduced at the cutting sites, the sequences are EcoR1 specific recognition sites, and if gene editing is successfully realized and accurate homologous recombination occurs, corresponding fragments can be specifically amplified through genomes, and then fragment enzyme digestion evaluation is carried out. The efficiency of EcoR1 cleavage can directly reflect the level of homologous recombination. Specifically, after incubating 40/60pmol AsCpf1 protein with the corresponding crRNA (1.2 fold equivalent) for 10min in vitro to form RNP, and reacting at 4 ℃ for 3 hr, 0.6 fold equivalent of single stranded donor DNA was added to form a complex for subsequent transfection. The transfection used was a CTX-1500A LE model electrotransfer apparatus, according to the instructions according to the buffer A and B equal proportion mixing configuration, for a single reaction using 20ul electrotransfer buffer heavy suspension 1x 106And (3) carrying out cell separation, namely adding the pre-prepared coupling compound into the cell suspension for electrotransformation under the condition of 420V for 30ms, immediately sucking out the cells after electrotransformation, and placing the cells in a preheated DMEM culture medium for continuous culture for 48 h. At 48 hours after transfection, cells were collected, the genome of the cells was extracted using a genome extraction kit provided by noximedium, and after PCR amplification of the target region, the amplified product was purified using a Cycle pure purification kit and then quantified. The evaluation of homologous recombination efficiency is carried out by enzyme digestion, the specific operation is as follows, sample adding is carried out according to 200 ng/reaction of PCR product, the enzyme digestion system is 10ul, 1ul CutSmart solution, 200ng template and 1ul EcoRI-HF enzyme are added, the mixture is supplemented to 10ul by water, the product is identified by 1% agarose gel electrophoresis after 1h reaction at 37 ℃, and the band is quantitatively analyzed. The results are shown in FIG. 6, where the RNP cut was performed at 40pmol and 60pmolThe coupling group (cCpf1) can be seen to be capable of obviously improving the homologous recombination efficiency by 7-8 times under the condition of the cut concentration, and the method for site-specific coupling of crRNA on RNP, which is created by the inventor, is proved to be capable of really improving the homologous recombination repair efficiency in gene editing.
The nucleic acid sequences used in this example are shown in the following table:
TABLE 9
Figure BDA0003117757000000121
Example 7: site-directed conjugation of cCpf1 in combination with AAV production of CAR-Jurkat cells and evaluation of cellular levels thereof
The invention adopts a Jurkat cell electrotransfer mode to target a TRAC locus for editing, and then delivers anti-CD19 CAR through adeno-associated virus (serotype 6), thereby realizing the site-specific integration and knock-in of CAR gene at the TRAC locus. The specific process is as follows:
1: production and purification of adeno-associated virus
The sequence of the integration delivered CAR gene used in the invention is shown in SEQ ID NO. 4. Wherein, the lower case underline is the homologous arm of TRAC gene, which is convenient for site-directed integration; lower case bold SFFV promoter to start expression of CAR gene; the capital bold is Myc-tag, which facilitates the detection of the expression of the CAR gene; the lower case italics are the bGH polyA signals; the upper case part is the gene sequence of anti-CD19 CAR. The gene sequence is synthesized by Anhui general biology company, then 5 'MluI and 3' RsrII restriction enzyme cutting sites are introduced at the tail ends through PCR primers, and the plasmid pAAV-SFFV-CD19CAR is obtained by cutting enzyme and connecting into a pAAV-MCS vector for packaging adeno-associated virus. The viral packaging helper plasmids pAAV-RC6 and pAAV-helper were purchased from Fenghui Bio Inc. AAV-293 cell lines for viral packaging were purchased from Nanjing Helhong Rui Biotech, Inc. The primers used for the construction of the pAAV-SFFV-CD19CAR plasmid are as follows:
watch 10
Name (R) Sequence (5 '-3')
Mlu1-FW ccatcactaggggttcctgcggccgcacgcgtgatgtaaggagctgctgtgacttgc
RsrII-RV cactaggggttcctgcggccgctcggtccggtgggttaatgagtgactgcgtgag
The AAV-293 cells were co-transfected with pAAV-SFFV-CD19CAR, pAAV-RC6, pAAV-helper in a mass ratio of 1:1:1 using PEI, virus was collected 72 hours after transfection, virus purification and titer were performed by Vigene Biosciences and named AAV-CD 19-CAR.
2: preparation of CAR-jurkat cells
Firstly, preparing a cCpf1 complex in vitro, preparing an RNP complex according to the molar ratio of Cpf1 to CrRNA of 1:1.2, incubating Cpf1 protein and CrRNA at room temperature for 5 minutes, reacting at 4 ℃ for 3 hours to prepare cCpf1, performing cell electrotransfer after the reaction is finished, and preparing a wild-type WT Cpf1 complex by the same method as a control. Jurkat cells were counted, 1.5X 106Suspending the cells with 20 μ L electrotransfer buffer (Celetrix, used after mixing solution A and solution B in equal proportion before use), mixing RNP and cells, slowly adding into 20 μ L electric shock cup, performing electric shock according to optimized parameters (400V, 30ms, 1pulse), rapidly adding cells into preheated RPMI-1640 culture medium after electric shock is finished, and culturing in incubator (37 deg.C, 5% CO)2). 4 hours after electrotransfer AAV-CD19-CAR at MOI of 1 × 106Jurkat cells were infected by addition to the medium. Cells were harvested 48 hours after electroporation, the genome extracted, and the knockout efficiency of the TRAC target site determined using the method described previously for T7E 1.
3: evaluation of production efficiency of CAR-jurkat cell
5 days after electroporation, the prepared CAR-jurkat cells were collected for flow assay to evaluate the efficiency of CAR-jurkat cell preparation. The specific process is as follows:
1) take 1X 106Individual cells were collected into 1.5ml Ep tubes for detection;
2) the cell culture medium was removed by centrifugation, the cells were resuspended with 200 μ L PBS + 2% FBS after one PBS wash, and blocked at 4 ℃ for 1 hour to reduce non-specific binding;
3) after blocking, the cells were centrifuged, resuspended in 100. mu.L PBS + 1% FBS, diluted 1:100, c-Mycmouse monoclonal antibody (Invitrogen) was added and incubated at 4 ℃ for 1.5 hours to allow primary antibody to bind to myc tag incubation;
4) after the primary antibody incubation was completed, the cells were washed twice with PBS to remove residual antibody as much as possible, then the cells were resuspended in 100. mu.L PBS, and Anti-mouse IgG (H + L), F (ab')2Fragment (Alexa) were added at a dilution of 1:1000
Figure BDA0003117757000000131
647Conjugate) (Cell Signaling Technology), incubating at 4 ℃ for 30 minutes in the absence of light to allow the secondary antibody to bind to the primary antibody;
5) after the incubation of the secondary antibody is finished, washing the cells twice by PBS, removing residual secondary antibody as much as possible to reduce the fluorescence background, and then re-suspending the cells by 100 mu L PBS;
5) the detection was carried out using a six-laser Flow Cytometer (CytoFLEX LX Flow Cytometer) with a detection channel of R660-APC, and the results were analyzed using analysis software CytExpert.
The flow analysis results are shown in FIG. 7, and show that the cCpf1 shows the improved efficiency of CAR-jurkat cell production compared with the WT Cpf1 group, and has the production efficiency as high as 80% when the cell is electrically transferred to 60pmol RNP. The CrRNA sequence and genomic PCR primers targeting the TRAC gene used in the experiment are shown below:
TABLE 11
Name (R) Sequence (5 '-3')
Human TRAC CrRNA gagtctctcagctggtacac
Geno-TRAC-FW cgagcagctggtttctaagatgc
Geno-TRAC-RV ctggactgccagaacaaggc
Example 8: evaluation of efficiency of integration at site-directed Gene integration CAR-Jurkat Gene level
The invention adopts an In-Out PCR mode to carry Out semi-quantitative evaluation on the efficiency of CAR site-specific integration In the TRAC locus. The specific process is as follows:
1: In-Out PCR primer design
Aiming at the integration of the TRAC locus, three specific primers of TRAC-1st-FW, CART-PCR-FW and TRAC-2st-RV are designed, wherein the primer TRAC-1st-FW is combined with a homologous arm at the 5 'end of the TRAC locus, the primer CART-PCR-FW is combined with a CAR gene integrated in a fixed point way, and the primer TRAC-2st-RV is combined with a homologous arm at the 3' end of the TRAC locus. The PCR primer sequences used In the In-Out PCR are shown below:
TABLE 12
Name (R) Sequence (5 '-3')
TRAC-1st-FW cccttgtccatcactggcat
CART-PCR-FW ggagtacgacgtgctggataag
TRAC-2st-RV gcacacccctcatctgactt
2: evaluation of Gene level integration efficiency
Jurkat cells obtained 5 days after the electroporation in step 3 of example 7 were taken, the genome of the cells was extracted using a genome extraction kit (Novozan), and three primers were mixed in the same tube of PCR reaction solution using the genomic DNA as a template to perform PCR reaction. If site-directed integration does not occur, only a band with the length of 843bp is obtained by amplification of primers TRAC-1st-FW and TRAC-2 st-RV; if the two primers are partially integrated at fixed points, there are 843bp bands obtained by amplification with the primers TRAC-1st-FW and TRAC-2st-RV and 1278bp bands obtained by amplification with the primers CART-PCR-FW and TRAC-2st-RV, respectively. The PCR reaction system is shown in Table 13.
TABLE 13 In-Out PCR reaction System
Genome template 50ng
Primer TRAC-1st-FW 1.5μL
Primer CART-PCR-FW 1.5μL
Primer TRAC-2st-RV 1.5μL
KOD OneTM PCR Master Mix(TOYOBO) 25μL
ddH2O Up to 50μL
After the PCR reaction is finished, electrophoresis is carried out by using 1% agarose gel according to the condition of 150V for 20min, and then analysis and quantification are carried out on a PCR band by using gel quantification software Tanon Gis, and the result is shown in figure 8, which shows that the group of cCpf1 has higher gene level integration efficiency than the group of wild-type Cpf1, and at the time of 30pmol RNP (low dose), the coupling complex cCpf1 has gene level integration efficiency equivalent to that of the high dose group, and has better CAR-jurkat cell preparation potential.
Example 9: site-directed conjugation of cCpf1 in combination with AAV production of CAR-T cells and evaluation of cellular levels thereof
It has been verified in examples 7 and 8 that the cpcf 1 has high production efficiency in combination with AAV in the production of CAR-jurkat cells, and subsequently the production effect of this method in the production of CAR-T cells was verified, as follows:
1: isolated culture of PBMC cells
Mononuclear lymphocytes in peripheral blood were isolated using Ficoll-Paque PLUS (GE Healthcare) and the procedure was as described by first diluting the blood sample by mixing it with a balanced salt solution in a volume ratio of 1:1, priming the 15ml tube with 3ml of Ficoll-Paque PLUS and carefully adding the diluted blood sample to the upper layer. Gradient centrifugation at 400 Xg for 30min at 20 deg.C, followed by careful aspiration of plasma layer and transfer of mononuclear cell layer to a new centrifuge tube, washing twice with balanced salt solution, and resuspension in T cell separation buffer (PBS without calcium and magnesium ions + 0.1% BSA +2mM EDTA) for further processing
2: isolation of primary T cells
Using DynabeadsTM UntouchedTMHuman T Cells Kit (Invitrogen) isolated primary T Cells from PBMC isolated in step 1 as follows:
1) taking 500 μ L PBMC (less than or equal to 5X 10)7Individual cells) were transferred to 15ml tubes;
2) adding 100 μ L of heat-inactivated fetal calf serum, and mixing to reduce nonspecific binding of antibody;
3) adding 100 mu L of allopody-mix, uniformly mixing, and incubating for 20min at 4 ℃;
4) adding 4ml of the T cell separation buffer solution in the step 1, uniformly mixing, and centrifuging at 350 Xg at 4 ℃ for 8 min;
5) resuspend the cells in 500. mu. L T cell isolation buffer, add 500. mu.L Dynabeads washed with T cell isolation buffer in advance, mix well and incubate at room temperature for 15 min;
6) adding 4ml of T cell separation buffer solution, uniformly mixing, placing the centrifugal tube on a magnetic frame, standing for 2 minutes, and transferring the supernatant into a new centrifugal tube to obtain the separated primary T cells.
3: stimulated culture of primary T cells
Primary T cells were cultured using XYbeads Human CD3/CD 28T Cell Activator (SAILY BIO) for stimulation. The primary T cells isolated in step 2 were counted, centrifuged to remove the T cell isolation buffer, and the cells were resuspended to a density of 8X 10 using T cell medium (X-VIVO + 10% heat inactivated FBS +30U/ml IL-2+5ng/ml IL-7+5ng/ml IL-15+ 1% ps)5Each cell/ml, was cultured in a six-well cell culture plate. According to the number of cells: the number of magnetic beads was 1:1, CD3/CD28 magnetic beads previously washed with T cell medium were added to the cells, and the cells were placed in a cell incubator (37 ℃ C., 5% CO)2) Culturing in the medium.
4: preparation of CAR-T cells
Similar to the preparation of CAR-jurkat cells, the cCpf1 complex was first prepared in vitro, RNP complex was prepared according to a molar ratio of Cpf1: CrRNA of 1:1.2(30pmol:42pmol), and Cpf1 protein was incubated with CrRNA at room temperatureAfter incubation for 5 minutes, the cells were incubated at 4 ℃ for 3 hours to prepare cCpf1, which was used for cell electroporation as a control in the same manner as wild-type WT Cpf1 complex. Taking T cells on the third day after stimulation for electrotransfer, firstly resuspending the T cells in a 15ml centrifuge tube, removing the magnetic beads in the culture solution by using a magnetic frame, repeating the process once to ensure that the magnetic beads are completely removed, and completely washing the magnetic beads by using PBS and storing the washed magnetic beads at 4 ℃ for later use. Counting primary T cells, taking 1.5X 106Suspending the cells with 20 μ L electrotransfer buffer (Celetrix, used after mixing solution A and solution B in equal proportion before use), mixing RNP and cells, slowly adding into 20 μ L electric shock cup, performing electric shock according to optimized parameters (420V, 20ms, 1pulse), rapidly adding cells into preheated T cell culture medium after electric shock is finished, and culturing in incubator (37 deg.C, 5% CO)2). 4 hours after electrotransfer AAV-CD19-CAR at MOI of 1 × 106The T cells were infected by addition to the medium. After 24 hours of electroporation, the rate of survival of T cells by electroporation and the cell density were recorded, and the CD3/CD28 magnetic beads previously kept ready were counted as cells: the number of magnetic beads was 1:1, and the cells were re-cultured in a re-stimulation manner. Cells were harvested 48 hours after electroporation, the genome extracted, and the knockout efficiency of the TRAC target site determined using the method described previously for T7E 1.
5: production evaluation of CAR-T cells
5 days after electroporation, the CAR-T cells prepared were collected for flow assay to evaluate the efficiency of CAR-T cell preparation. The specific process is as follows:
1) take 1X 106Collecting the cells into a 1.5ml Ep tube, and completely removing magnetic beads in the culture solution by using a magnetic frame so as not to influence flow detection;
2) the cell culture medium was removed by centrifugation, the cells were resuspended with 200 μ L PBS + 2% FBS after one PBS wash, and blocked at 4 ℃ for 1 hour to reduce non-specific binding;
3) after the blocking, the cells were centrifuged, resuspended in 100. mu.L PBS + 1% FBS, diluted 1:100, c-Mycmouse monoclonal antibody (Invitrogen) was added, and incubated at 4 ℃ for 1.5 hours to allow primary antibody to bind to myc tag incubation;
4) after the primary antibody incubation was completed, the cells were washed twice with PBS to remove residual antibody as much as possible, then the cells were resuspended in 100. mu.L PBS, and Anti-mouse IgG (H + L), F (ab')2Fragment (Alexa) were added at a dilution of 1:1000
Figure BDA0003117757000000151
647Conjugate) (Cell Signaling Technology), incubating at 4 ℃ for 30 minutes in the absence of light to allow the secondary antibody to bind to the primary antibody;
5) after the incubation of the secondary antibody is finished, washing the cells twice by PBS, removing residual secondary antibody as much as possible to reduce the fluorescence background, and then re-suspending the cells by 100 mu L PBS;
5) the detection was carried out using a six-laser Flow Cytometer (CytoFLEX LX Flow Cytometer) with a detection channel of R660-APC, and the results were analyzed using analysis software CytExpert.
As shown in FIG. 9B, the cCpf1 group increased the efficiency of CAR-T cell production from 30% to 53% compared to the wild-type Cpf1 group, and the immunological function of the produced CAR-T cells was evaluated.
Example 10: site-directed conjugation of cpcpf 1 in combination with AAV for immunological evaluation of CAR-T cells
The CD19 positive NALM6 (adult B-type acute lymphoblastic leukemia cell strain) is used for detecting the targeting killing function and the secretion of killer cytokines of the prepared CAR-T cells. LDH-Glo Using an LDH detection kitTMCytoxicity Assay (Promega) was used to detect the release of lactate dehydrogenase LDH in the culture medium after the death of NALM6 cells, and an ELISA detection kit (Invitrogen) was used to detect cytokine interferon gamma (IFN- γ) and tumor necrosis factor- α (TNF- α) secreted by CAR-T cells, as follows:
the prepared CAR-T cells were washed twice with RPMI-1640 cell culture medium (medium for NALM-6 cell culture) for use. The CAR-T cells and NALM-6 cells were counted separately and the two cells were co-incubated in 96-well plates at 1:10, i.e.1X 10 cells were added to each well4NALM-6 cells and 1X 105Each CAR-T cell is controlled by a T cell which is not modified by CAR, and each incubation group is provided with three diplopore platesAnd (6) rows. After incubation in an incubator at 37 ℃ for 24-48 hours, the culture supernatant was centrifuged and assayed for LDH and ELISA.
1: LDH release assay
The culture supernatant was diluted 1:100 with LDH storage buffer (200mM Tris-HCl, pH7.3+ 10% glycerol + 1% BSA), and 50. mu.L of the diluted solution was added to a 96-well plate for detection. An LDH detection reagent is prepared by using 50 mu L of Enzyme mix and 0.25 mu L of detection substrate according to each reaction in a 96-well plate, the LDH detection reagent is added into a sample to be detected, chemiluminescence is detected by using a microplate reader after incubation for 60min at room temperature, and the integration time is 1 ms. Cell killing efficiency of 100 × (experimental group LDH release-medium background)/(control group LDH release-medium background), cytotoxicity assay results for CAR-T cells as shown in fig. 10A, CAR-T cells prepared from wild-type Cpf1 group had a cell killing efficiency of 24.5%, whereas CAR-T cells prepared from coupling complex cpcf 1 group developed by the present inventors had a cell killing efficiency of 42.5%.
2: cytokine IFN-gamma and TNF-alpha secretion detection
In this experiment, Human IFN gamma Uncoated ELISA kit (Invitrogen) and Human TNF alpha Uncoated ELISA kit (Invitrogen) were used to detect the IFN-. gamma.and TNF-. alpha.content in the culture medium supernatant after co-incubation, respectively, and the cytokine content in the culture medium was quantified according to a standard curve. As shown in FIGS. 10B and 10C, it can be seen that the IFN-. gamma.and TNF-. alpha.release in the cCpf1 group was increased 2-fold and 1.5-fold, respectively, compared to the WT Cpf1 group.
Example 11: cCpf 1-based evaluation of multi-site knockout CAR-T cells
The inventor utilizes a cpcf 1 system to knock out four sites of TRAC, beta 2M, PD1 and CTLA4 of T cells at the same time, and utilizes AAV delivery to realize site-directed integration of CAR genes at the TRAC site, so as to successfully prepare universal T cells capable of breaking immunosuppression and knocking in CAR genes at a site, and as a result, as shown in fig. 11, it can be seen that the cpcf 1 group shows higher editing efficiency in double site editing, triple site editing and quadruple site editing compared with wild type.
The specific evaluation process is as follows:
1: preparation of RNP complexes targeting different sites
RNP complexes targeting different sites are prepared by reaction respectively, the RNP complexes are prepared according to the molar ratio of Cpf1 to CrRNA of 1:1.2, Cpf1 protein and CrRNA are incubated for 5 minutes at room temperature and then placed at 4 ℃ for reaction for 3 hours to prepare cCpf1, and wild-type WT Cpf1 complexes are prepared by the same method as a control.
2: preparation of multiple Gene edited CAR-T cells
Similar to the preparation of CAR-T cells in step 4 of example 8, 1.5X 10 cells were taken6Suspending each cell with 20 μ L electrotransfer buffer solution (Celetrix, mixing solution A and solution B at equal ratio before use), mixing RNPs in different multiple gene editing combinations, mixing total RNPs and cells, slowly adding into 20 μ L electric shock cup, performing electric shock according to optimized parameters (420V, 20ms, 1pulse), rapidly adding cells into preheated T cell culture medium after electric shock is finished, and culturing in incubator (37 deg.C, 5% CO)2). 4 hours after electrotransfer AAV-CD19-CAR at MOI of 1 × 106The cells were infected by addition to the medium. After 24 hours of electroporation, the rate of survival of T cells by electroporation and the cell density were recorded, and the CD3/CD28 magnetic beads previously kept ready were counted as cells: the number of magnetic beads was 1:1, and the cells were re-cultured in a re-stimulation manner. Cells were harvested 48 hours after electroporation, the genome extracted, and the gene level knockout efficiency of the TRAC target site determined using the method described previously for T7E 1. The CrRNA sequences and genomic PCR primers used in the experiments targeting the β 2M, PD1, CTLA4 genes are shown below:
TABLE 14
Name(s) Sequence (5 '-3')
Human β2M CrRNA ccgatattcctcaggtactc
Human PD1 CrRNA gcacgaagctctccgatgtg
Human CTLA4 CrRNA cctggagatgcatactcacacac
IVD-β2M-FW gaggaattatgagggaaagataccaagtcacgg
IVD-β2M-RV caaaggcctataccttcttgagatgttcgttcag
IVD-PD1-FW cagcagagacttctcaatgacattccagc
IVD-PD1-RV ggacagagatgccggtcaccattc
IVD-CLTA4-FW gctggaaaacagttgagagatggaggg
IVD-CLTA4-RV catgatggttagcactccagagcgag
3: evaluation of multiple Gene editing T cells
T cells after 5 days of electrotransformation in step 2 were characterized for efficiency of T cell multiplex gene editing by flow assay of β 2M, PD1, CTLA4 protein expression levels in the edited T cells. Firstly, 2 is multiplied by 106The individual cells were collected in 1.5ml Ep tubes and placed in culture using a magnetic holderThe magnetic beads are removed completely to avoid affecting flow detection, and the cells are divided into two parts, one part is used for detecting gene knockout efficiency, and the other part is used for detecting fixed point knock-in efficiency of CAR. Detection of CAR site-directed integration efficiency was performed using myc tag staining as described in example 8, step 5, and is not described herein. The flow detection process of gene knockout efficiency is as follows:
1) the cell culture medium was removed by centrifugation, the cells were resuspended with 200 μ L PBS + 2% FBS after one PBS wash, and blocked at 4 ℃ for 1 hour to reduce non-specific binding;
2) after blocking, the cells were centrifuged and resuspended in 80. mu.L PBS + 1% FBS and 5. mu.L of APC anti-human CD3 (Biolegged), 5. mu.L of FITC anti-human β 2-microrogobulin (Biolegged), 5. mu.L of PE anti-human CD279 (Biolegged), 5. mu.L of PE/cysteine 7 anti-human CD152 (Biolegged), approximately 100. mu.L of staining volume containing 1X 106Incubating the cells at 4 ℃ for 1 hour in the dark to allow the antibody to bind to the corresponding cell surface protein;
4) after the incubation is finished, washing the cells twice by PBS (phosphate buffer solution), removing residual antibodies as much as possible, and then resuspending the cells by 100 mu L of PBS;
5) detecting by using a six-laser Flow Cytometer (CytoFLEX LX Flow Cytometer), wherein detection channels are B525-FITC, Y585-PE, R660-APC and Y763-PC7, analyzing the total knockout efficiency of the T cells by using analysis software CytExpert, and if the four-site gene knockout efficiency is 100 multiplied by the cell number/total cell number of the APC-FITC-PE-PY7-, the knockout efficiencies of the rest sites are analogized.
The foregoing is only a preferred embodiment of the present invention, and it should be noted that it is obvious to those skilled in the art that various modifications and improvements can be made without departing from the principle of the present invention, and these modifications and improvements should also be considered as the protection scope of the present invention.
Sequence listing
<110> Beijing university
<120> use of chemically modified CRISPR/Cpf1 complex
<130> MP21006798
<160> 4
<170> SIPOSequenceListing 1.0
<210> 1
<211> 1342
<212> PRT
<213> AsCpf1(AsCpf1)
<400> 1
Met Thr Gln Phe Glu Gly Phe Thr Asn Leu Tyr Gln Val Ser Lys Thr
1 5 10 15
Leu Arg Phe Glu Leu Ile Pro Gln Gly Lys Thr Leu Lys His Ile Gln
20 25 30
Glu Gln Gly Phe Ile Glu Glu Asp Lys Ala Arg Asn Asp His Tyr Lys
35 40 45
Glu Leu Lys Pro Ile Ile Asp Arg Ile Tyr Lys Thr Tyr Ala Asp Gln
50 55 60
Cys Leu Gln Leu Val Gln Leu Asp Trp Glu Asn Leu Ser Ala Ala Ile
65 70 75 80
Asp Ser Tyr Arg Lys Glu Lys Thr Glu Glu Thr Arg Asn Ala Leu Ile
85 90 95
Glu Glu Gln Ala Thr Tyr Arg Asn Ala Ile His Asp Tyr Phe Ile Gly
100 105 110
Arg Thr Asp Asn Leu Thr Asp Ala Ile Asn Lys Arg His Ala Glu Ile
115 120 125
Tyr Lys Gly Leu Phe Lys Ala Glu Leu Phe Asn Gly Lys Val Leu Lys
130 135 140
Gln Leu Gly Thr Val Thr Thr Thr Glu His Glu Asn Ala Leu Leu Arg
145 150 155 160
Ser Phe Asp Lys Phe Thr Thr Tyr Phe Ser Gly Phe Tyr Glu Asn Arg
165 170 175
Lys Asn Val Phe Ser Ala Glu Asp Ile Ser Thr Ala Ile Pro His Arg
180 185 190
Ile Val Gln Asp Asn Phe Pro Lys Phe Lys Glu Asn Cys His Ile Phe
195 200 205
Thr Arg Leu Ile Thr Ala Val Pro Ser Leu Arg Glu His Phe Glu Asn
210 215 220
Val Lys Lys Ala Ile Gly Ile Phe Val Ser Thr Ser Ile Glu Glu Val
225 230 235 240
Phe Ser Phe Pro Phe Tyr Asn Gln Leu Leu Thr Gln Thr Gln Ile Asp
245 250 255
Leu Tyr Asn Gln Leu Leu Gly Gly Ile Ser Arg Glu Ala Gly Thr Glu
260 265 270
Lys Ile Lys Gly Leu Asn Glu Val Leu Asn Leu Ala Ile Gln Lys Asn
275 280 285
Asp Glu Thr Ala His Ile Ile Ala Ser Leu Pro His Arg Phe Ile Pro
290 295 300
Leu Phe Lys Gln Ile Leu Ser Asp Arg Asn Thr Leu Ser Phe Ile Leu
305 310 315 320
Glu Glu Phe Lys Ser Asp Glu Glu Val Ile Gln Ser Phe Cys Lys Tyr
325 330 335
Lys Thr Leu Leu Arg Asn Glu Asn Val Leu Glu Thr Ala Glu Ala Leu
340 345 350
Phe Asn Glu Leu Asn Ser Ile Asp Leu Thr His Ile Phe Ile Ser His
355 360 365
Lys Lys Leu Glu Thr Ile Ser Ser Ala Leu Cys Asp His Trp Asp Thr
370 375 380
Leu Arg Asn Ala Leu Tyr Glu Arg Arg Ile Ser Glu Leu Thr Gly Lys
385 390 395 400
Ile Thr Lys Ser Ala Lys Glu Lys Val Gln Arg Ser Leu Lys His Glu
405 410 415
Asp Ile Asn Leu Gln Glu Ile Ile Ser Ala Ala Gly Lys Glu Leu Ser
420 425 430
Glu Ala Phe Lys Gln Lys Thr Ser Glu Ile Leu Ser His Ala His Ala
435 440 445
Ala Leu Asp Gln Pro Leu Pro Thr Thr Leu Lys Lys Gln Glu Glu Lys
450 455 460
Glu Ile Leu Lys Ser Gln Leu Asp Ser Leu Leu Gly Leu Tyr His Leu
465 470 475 480
Leu Asp Trp Phe Ala Val Asp Glu Ser Asn Glu Val Asp Pro Glu Phe
485 490 495
Ser Ala Arg Leu Thr Gly Ile Lys Leu Glu Met Glu Pro Ser Leu Ser
500 505 510
Phe Tyr Asn Lys Ala Arg Asn Tyr Ala Thr Lys Lys Pro Tyr Ser Val
515 520 525
Glu Lys Phe Lys Leu Asn Phe Gln Met Pro Thr Leu Ala Ser Gly Trp
530 535 540
Asp Val Asn Lys Glu Lys Asn Asn Gly Ala Ile Leu Phe Val Lys Asn
545 550 555 560
Gly Leu Tyr Tyr Leu Gly Ile Met Pro Lys Gln Lys Gly Arg Tyr Lys
565 570 575
Ala Leu Ser Phe Glu Pro Thr Glu Lys Thr Ser Glu Gly Phe Asp Lys
580 585 590
Met Tyr Tyr Asp Tyr Phe Pro Asp Ala Ala Lys Met Ile Pro Lys Cys
595 600 605
Ser Thr Gln Leu Lys Ala Val Thr Ala His Phe Gln Thr His Thr Thr
610 615 620
Pro Ile Leu Leu Ser Asn Asn Phe Ile Glu Pro Leu Glu Ile Thr Lys
625 630 635 640
Glu Ile Tyr Asp Leu Asn Asn Pro Glu Lys Glu Pro Lys Lys Phe Gln
645 650 655
Thr Ala Tyr Ala Lys Lys Thr Gly Asp Gln Lys Gly Tyr Arg Glu Ala
660 665 670
Leu Cys Lys Trp Ile Asp Phe Thr Arg Asp Phe Leu Ser Lys Tyr Thr
675 680 685
Lys Thr Thr Ser Ile Asp Leu Ser Ser Leu Arg Pro Ser Ser Gln Tyr
690 695 700
Lys Asp Leu Gly Glu Tyr Tyr Ala Glu Leu Asn Pro Leu Leu Tyr His
705 710 715 720
Ile Ser Phe Gln Arg Ile Ala Glu Lys Glu Ile Met Asp Ala Val Glu
725 730 735
Thr Gly Lys Leu Tyr Leu Phe Gln Ile Tyr Asn Lys Asp Phe Ala Lys
740 745 750
Gly His His Gly Lys Pro Asn Leu His Thr Leu Tyr Trp Thr Gly Leu
755 760 765
Phe Ser Pro Glu Asn Leu Ala Lys Thr Ser Ile Lys Leu Asn Gly Gln
770 775 780
Ala Glu Leu Phe Tyr Arg Pro Lys Ser Arg Met Lys Arg Met Ala His
785 790 795 800
Arg Leu Gly Glu Lys Met Leu Asn Lys Lys Leu Lys Asp Gln Lys Thr
805 810 815
Pro Ile Pro Asp Thr Leu Tyr Gln Glu Leu Tyr Asp Tyr Val Asn His
820 825 830
Arg Leu Ser His Asp Leu Ser Asp Glu Ala Arg Ala Leu Leu Pro Asn
835 840 845
Val Ile Thr Lys Glu Val Ser His Glu Ile Ile Lys Asp Arg Arg Phe
850 855 860
Thr Ser Asp Lys Phe Phe Phe His Val Pro Ile Thr Leu Asn Tyr Gln
865 870 875 880
Ala Ala Asn Ser Pro Ser Lys Phe Asn Gln Arg Val Asn Ala Tyr Leu
885 890 895
Lys Glu His Pro Glu Thr Pro Ile Ile Gly Ile Asp Arg Gly Glu Arg
900 905 910
Asn Leu Ile Tyr Ile Thr Val Ile Asp Ser Thr Gly Lys Ile Leu Glu
915 920 925
Gln Arg Ser Leu Asn Thr Ile Gln Gln Phe Asp Tyr Gln Lys Lys Leu
930 935 940
Asp Asn Arg Glu Lys Glu Arg Val Ala Ala Arg Gln Ala Trp Ser Val
945 950 955 960
Val Gly Thr Ile Lys Asp Leu Lys Gln Gly Tyr Leu Ser Gln Val Ile
965 970 975
His Glu Ile Val Asp Leu Met Ile His Tyr Gln Ala Val Val Val Leu
980 985 990
Glu Asn Leu Asn Phe Gly Phe Lys Ser Lys Arg Thr Gly Ile Ala Glu
995 1000 1005
Lys Ala Val Tyr Gln Gln Phe Glu Lys Met Leu Ile Asp Lys Leu Asn
1010 1015 1020
Cys Leu Val Leu Lys Asp Tyr Pro Ala Glu Lys Val Gly Gly Val Leu
1025 1030 1035 1040
Asn Pro Tyr Gln Leu Thr Asp Gln Phe Thr Ser Phe Ala Lys Met Gly
1045 1050 1055
Thr Gln Ser Gly Phe Leu Phe Tyr Val Pro Ala Pro Tyr Thr Ser Lys
1060 1065 1070
Ile Asp Pro Leu Thr Gly Phe Val Asp Pro Phe Val Trp Lys Thr Ile
1075 1080 1085
Lys Asn His Glu Ser Arg Lys His Phe Leu Glu Gly Phe Asp Phe Leu
1090 1095 1100
His Tyr Asp Val Lys Thr Gly Asp Phe Ile Leu His Phe Lys Met Asn
1105 1110 1115 1120
Arg Asn Leu Ser Phe Gln Arg Gly Leu Pro Gly Phe Met Pro Ala Trp
1125 1130 1135
Asp Ile Val Phe Glu Lys Asn Glu Thr Gln Phe Asp Ala Lys Gly Thr
1140 1145 1150
Pro Phe Ile Ala Gly Lys Arg Ile Val Pro Val Ile Glu Asn His Arg
1155 1160 1165
Phe Thr Gly Arg Tyr Arg Asp Leu Tyr Pro Ala Asn Glu Leu Ile Ala
1170 1175 1180
Leu Leu Glu Glu Lys Gly Ile Val Phe Arg Asp Gly Ser Asn Ile Leu
1185 1190 1195 1200
Pro Lys Leu Leu Glu Asn Asp Asp Ser His Ala Ile Asp Thr Met Val
1205 1210 1215
Ala Leu Ile Arg Ser Val Leu Gln Met Arg Asn Ser Asn Ala Ala Thr
1220 1225 1230
Gly Glu Asp Tyr Ile Asn Ser Pro Val Arg Asp Leu Asn Gly Val Cys
1235 1240 1245
Phe Asp Ser Arg Phe Gln Asn Pro Glu Trp Pro Met Asp Ala Asp Ala
1250 1255 1260
Asn Gly Ala Tyr His Ile Ala Leu Lys Gly Gln Leu Leu Leu Asn His
1265 1270 1275 1280
Leu Lys Glu Ser Lys Asp Leu Lys Leu Gln Asn Gly Ile Ser Asn Gln
1285 1290 1295
Asp Trp Leu Ala Tyr Ile Gln Glu Leu Arg Asn Lys Arg Pro Ala Ala
1300 1305 1310
Thr Lys Lys Ala Gly Gln Ala Lys Lys Lys Lys Gly Ser Pro Lys Lys
1315 1320 1325
Lys Arg Lys Val Gly Ser Leu Glu His His His His His His
1330 1335 1340
<210> 2
<211> 1394
<212> PRT
<213> SpCas9(SpCas9)
<400> 2
Met Gly Phe Met Pro Lys Lys Lys Arg Lys Val Met Asp Lys Lys Tyr
1 5 10 15
Ser Ile Gly Leu Asp Ile Gly Thr Asn Ser Val Gly Trp Ala Val Ile
20 25 30
Thr Asp Glu Tyr Lys Val Pro Ser Lys Lys Phe Lys Val Leu Gly Asn
35 40 45
Thr Asp Arg His Ser Ile Lys Lys Asn Leu Ile Gly Ala Leu Leu Phe
50 55 60
Asp Ser Gly Glu Thr Ala Glu Ala Thr Arg Leu Lys Arg Thr Ala Arg
65 70 75 80
Arg Arg Tyr Thr Arg Arg Lys Asn Arg Ile Cys Tyr Leu Gln Glu Ile
85 90 95
Phe Ser Asn Glu Met Ala Lys Val Asp Asp Ser Phe Phe His Arg Leu
100 105 110
Glu Glu Ser Phe Leu Val Glu Glu Asp Lys Lys His Glu Arg His Pro
115 120 125
Ile Phe Gly Asn Ile Val Asp Glu Val Ala Tyr His Glu Lys Tyr Pro
130 135 140
Thr Ile Tyr His Leu Arg Lys Lys Leu Val Asp Ser Thr Asp Lys Ala
145 150 155 160
Asp Leu Arg Leu Ile Tyr Leu Ala Leu Ala His Met Ile Lys Phe Arg
165 170 175
Gly His Phe Leu Ile Glu Gly Asp Leu Asn Pro Asp Asn Ser Asp Val
180 185 190
Asp Lys Leu Phe Ile Gln Leu Val Gln Thr Tyr Asn Gln Leu Phe Glu
195 200 205
Glu Asn Pro Ile Asn Ala Ser Gly Val Asp Ala Lys Ala Ile Leu Ser
210 215 220
Ala Arg Leu Ser Lys Ser Arg Arg Leu Glu Asn Leu Ile Ala Gln Leu
225 230 235 240
Pro Gly Glu Lys Lys Asn Gly Leu Phe Gly Asn Leu Ile Ala Leu Ser
245 250 255
Leu Gly Leu Thr Pro Asn Phe Lys Ser Asn Phe Asp Leu Ala Glu Asp
260 265 270
Ala Lys Leu Gln Leu Ser Lys Asp Thr Tyr Asp Asp Asp Leu Asp Asn
275 280 285
Leu Leu Ala Gln Ile Gly Asp Gln Tyr Ala Asp Leu Phe Leu Ala Ala
290 295 300
Lys Asn Leu Ser Asp Ala Ile Leu Leu Ser Asp Ile Leu Arg Val Asn
305 310 315 320
Thr Glu Ile Thr Lys Ala Pro Leu Ser Ala Ser Met Ile Lys Arg Tyr
325 330 335
Asp Glu His His Gln Asp Leu Thr Leu Leu Lys Ala Leu Val Arg Gln
340 345 350
Gln Leu Pro Glu Lys Tyr Lys Glu Ile Phe Phe Asp Gln Ser Lys Asn
355 360 365
Gly Tyr Ala Gly Tyr Ile Asp Gly Gly Ala Ser Gln Glu Glu Phe Tyr
370 375 380
Lys Phe Ile Lys Pro Ile Leu Glu Lys Met Asp Gly Thr Glu Glu Leu
385 390 395 400
Leu Val Lys Leu Asn Arg Glu Asp Leu Leu Arg Lys Gln Arg Thr Phe
405 410 415
Asp Asn Gly Ser Ile Pro His Gln Ile His Leu Gly Glu Leu His Ala
420 425 430
Ile Leu Arg Arg Gln Glu Asp Phe Tyr Pro Phe Leu Lys Asp Asn Arg
435 440 445
Glu Lys Ile Glu Lys Ile Leu Thr Phe Arg Ile Pro Tyr Tyr Val Gly
450 455 460
Pro Leu Ala Arg Gly Asn Ser Arg Phe Ala Trp Met Thr Arg Lys Ser
465 470 475 480
Glu Glu Thr Ile Thr Pro Trp Asn Phe Glu Glu Val Val Asp Lys Gly
485 490 495
Ala Ser Ala Gln Ser Phe Ile Glu Arg Met Thr Asn Phe Asp Lys Asn
500 505 510
Leu Pro Asn Glu Lys Val Leu Pro Lys His Ser Leu Leu Tyr Glu Tyr
515 520 525
Phe Thr Val Tyr Asn Glu Leu Thr Lys Val Lys Tyr Val Thr Glu Gly
530 535 540
Met Arg Lys Pro Ala Phe Leu Ser Gly Glu Gln Lys Lys Ala Ile Val
545 550 555 560
Asp Leu Leu Phe Lys Thr Asn Arg Lys Val Thr Val Lys Gln Leu Lys
565 570 575
Glu Asp Tyr Phe Lys Lys Ile Glu Cys Phe Asp Ser Val Glu Ile Ser
580 585 590
Gly Val Glu Asp Arg Phe Asn Ala Ser Leu Gly Thr Tyr His Asp Leu
595 600 605
Leu Lys Ile Ile Lys Asp Lys Asp Phe Leu Asp Asn Glu Glu Asn Glu
610 615 620
Asp Ile Leu Glu Asp Ile Val Leu Thr Leu Thr Leu Phe Glu Asp Arg
625 630 635 640
Glu Met Ile Glu Glu Arg Leu Lys Thr Tyr Ala His Leu Phe Asp Asp
645 650 655
Lys Val Met Lys Gln Leu Lys Arg Arg Arg Tyr Thr Gly Trp Gly Arg
660 665 670
Leu Ser Arg Lys Leu Ile Asn Gly Ile Arg Asp Lys Gln Ser Gly Lys
675 680 685
Thr Ile Leu Asp Phe Leu Lys Ser Asp Gly Phe Ala Asn Arg Asn Phe
690 695 700
Met Gln Leu Ile His Asp Asp Ser Leu Thr Phe Lys Glu Asp Ile Gln
705 710 715 720
Lys Ala Gln Val Ser Gly Gln Gly Asp Ser Leu His Glu His Ile Ala
725 730 735
Asn Leu Ala Gly Ser Pro Ala Ile Lys Lys Gly Ile Leu Gln Thr Val
740 745 750
Lys Val Val Asp Glu Leu Val Lys Val Met Gly Arg His Lys Pro Glu
755 760 765
Asn Ile Val Ile Glu Met Ala Arg Glu Asn Gln Thr Thr Gln Lys Gly
770 775 780
Gln Lys Asn Ser Arg Glu Arg Met Lys Arg Ile Glu Glu Gly Ile Lys
785 790 795 800
Glu Leu Gly Ser Gln Ile Leu Lys Glu His Pro Val Glu Asn Thr Gln
805 810 815
Leu Gln Asn Glu Lys Leu Tyr Leu Tyr Tyr Leu Gln Asn Gly Arg Asp
820 825 830
Met Tyr Val Asp Gln Glu Leu Asp Ile Asn Arg Leu Ser Asp Tyr Asp
835 840 845
Val Asp His Ile Val Pro Gln Ser Phe Leu Lys Asp Asp Ser Ile Asp
850 855 860
Asn Lys Val Leu Thr Arg Ser Asp Lys Asn Arg Gly Lys Ser Asp Asn
865 870 875 880
Val Pro Ser Glu Glu Val Val Lys Lys Met Lys Asn Tyr Trp Arg Gln
885 890 895
Leu Leu Asn Ala Lys Leu Ile Thr Gln Arg Lys Phe Asp Asn Leu Thr
900 905 910
Lys Ala Glu Arg Gly Gly Leu Ser Glu Leu Asp Lys Ala Gly Phe Ile
915 920 925
Lys Arg Gln Leu Val Glu Thr Arg Gln Ile Thr Lys His Val Ala Gln
930 935 940
Ile Leu Asp Ser Arg Met Asn Thr Lys Tyr Asp Glu Asn Asp Lys Leu
945 950 955 960
Ile Arg Glu Val Lys Val Ile Thr Leu Lys Ser Lys Leu Val Ser Asp
965 970 975
Phe Arg Lys Asp Phe Gln Phe Tyr Lys Val Arg Glu Ile Asn Asn Tyr
980 985 990
His His Ala His Asp Ala Tyr Leu Asn Ala Val Val Gly Thr Ala Leu
995 1000 1005
Ile Lys Lys Tyr Pro Lys Leu Glu Ser Glu Phe Val Tyr Gly Asp Tyr
1010 1015 1020
Lys Val Tyr Asp Val Arg Lys Met Ile Ala Lys Ser Glu Gln Glu Ile
1025 1030 1035 1040
Gly Lys Ala Thr Ala Lys Tyr Phe Phe Tyr Ser Asn Ile Met Asn Phe
1045 1050 1055
Phe Lys Thr Glu Ile Thr Leu Ala Asn Gly Glu Ile Arg Lys Arg Pro
1060 1065 1070
Leu Ile Glu Thr Asn Gly Glu Thr Gly Glu Ile Val Trp Asp Lys Gly
1075 1080 1085
Arg Asp Phe Ala Thr Val Arg Lys Val Leu Ser Met Pro Gln Val Asn
1090 1095 1100
Ile Val Lys Lys Thr Glu Val Gln Thr Gly Gly Phe Ser Lys Glu Ser
1105 1110 1115 1120
Ile Leu Pro Lys Arg Asn Ser Asp Lys Leu Ile Ala Arg Lys Lys Asp
1125 1130 1135
Trp Asp Pro Lys Lys Tyr Gly Gly Phe Asp Ser Pro Thr Val Ala Tyr
1140 1145 1150
Ser Val Leu Val Val Ala Lys Val Glu Lys Gly Lys Ser Lys Lys Leu
1155 1160 1165
Lys Ser Val Lys Glu Leu Leu Gly Ile Thr Ile Met Glu Arg Ser Ser
1170 1175 1180
Phe Glu Lys Asn Pro Ile Asp Phe Leu Glu Ala Lys Gly Tyr Lys Glu
1185 1190 1195 1200
Val Lys Lys Asp Leu Ile Ile Lys Leu Pro Lys Tyr Ser Leu Phe Glu
1205 1210 1215
Leu Glu Asn Gly Arg Lys Arg Met Leu Ala Ser Ala Gly Glu Leu Gln
1220 1225 1230
Lys Gly Asn Glu Leu Ala Leu Pro Ser Lys Tyr Val Asn Phe Leu Tyr
1235 1240 1245
Leu Ala Ser His Tyr Glu Lys Leu Lys Gly Ser Pro Glu Asp Asn Glu
1250 1255 1260
Gln Lys Gln Leu Phe Val Glu Gln His Lys His Tyr Leu Asp Glu Ile
1265 1270 1275 1280
Ile Glu Gln Ile Ser Glu Phe Ser Lys Arg Val Ile Leu Ala Asp Ala
1285 1290 1295
Asn Leu Asp Lys Val Leu Ser Ala Tyr Asn Lys His Arg Asp Lys Pro
1300 1305 1310
Ile Arg Glu Gln Ala Glu Asn Ile Ile His Leu Phe Thr Leu Thr Asn
1315 1320 1325
Leu Gly Ala Pro Ala Ala Phe Lys Tyr Phe Asp Thr Thr Ile Asp Arg
1330 1335 1340
Lys Arg Tyr Thr Ser Thr Lys Glu Val Leu Asp Ala Thr Leu Ile His
1345 1350 1355 1360
Gln Ser Ile Thr Gly Leu Tyr Glu Thr Arg Ile Asp Leu Ser Gln Leu
1365 1370 1375
Gly Gly Asp Pro Lys Lys Lys Arg Lys Val Leu Glu His His His His
1380 1385 1390
His His
<210> 3
<211> 1263
<212> PRT
<213> LbCpf1(LbCpf1)
<400> 3
Met Ser Lys Leu Glu Lys Phe Thr Asn Cys Tyr Ser Leu Ser Lys Thr
1 5 10 15
Leu Arg Phe Lys Ala Ile Pro Val Gly Lys Thr Gln Glu Asn Ile Asp
20 25 30
Asn Lys Arg Leu Leu Val Glu Asp Glu Lys Arg Ala Glu Asp Tyr Lys
35 40 45
Gly Val Lys Lys Leu Leu Asp Arg Tyr Tyr Leu Ser Phe Ile Asn Asp
50 55 60
Val Leu His Ser Ile Lys Leu Lys Asn Leu Asn Asn Tyr Ile Ser Leu
65 70 75 80
Phe Arg Lys Lys Thr Arg Thr Glu Lys Glu Asn Lys Glu Leu Glu Asn
85 90 95
Leu Glu Ile Asn Leu Arg Lys Glu Ile Ala Lys Ala Phe Lys Gly Asn
100 105 110
Glu Gly Tyr Lys Ser Leu Phe Lys Lys Asp Ile Ile Glu Thr Ile Leu
115 120 125
Pro Glu Phe Leu Asp Asp Lys Asp Glu Ile Ala Leu Val Asn Ser Phe
130 135 140
Asn Gly Phe Thr Thr Ala Phe Thr Gly Phe Phe Asp Asn Arg Glu Asn
145 150 155 160
Met Phe Ser Glu Glu Ala Lys Ser Thr Ser Ile Ala Phe Arg Cys Ile
165 170 175
Asn Glu Asn Leu Thr Arg Tyr Ile Ser Asn Met Asp Ile Phe Glu Lys
180 185 190
Val Asp Ala Ile Phe Asp Lys His Glu Val Gln Glu Ile Lys Glu Lys
195 200 205
Ile Leu Asn Ser Asp Tyr Asp Val Glu Asp Phe Phe Glu Gly Glu Phe
210 215 220
Phe Asn Phe Val Leu Thr Gln Glu Gly Ile Asp Val Tyr Asn Ala Ile
225 230 235 240
Ile Gly Gly Phe Val Thr Glu Ser Gly Glu Lys Ile Lys Gly Leu Asn
245 250 255
Glu Tyr Ile Asn Leu Tyr Asn Gln Lys Thr Lys Gln Lys Leu Pro Lys
260 265 270
Phe Lys Pro Leu Tyr Lys Gln Val Leu Ser Asp Arg Glu Ser Leu Ser
275 280 285
Phe Tyr Gly Glu Gly Tyr Thr Ser Asp Glu Glu Val Leu Glu Val Phe
290 295 300
Arg Asn Thr Leu Asn Lys Asn Ser Glu Ile Phe Ser Ser Ile Lys Lys
305 310 315 320
Leu Glu Lys Leu Phe Lys Asn Phe Asp Glu Tyr Ser Ser Ala Gly Ile
325 330 335
Phe Val Lys Asn Gly Pro Ala Ile Ser Thr Ile Ser Lys Asp Ile Phe
340 345 350
Gly Glu Trp Asn Val Ile Arg Asp Lys Trp Asn Ala Glu Tyr Asp Asp
355 360 365
Ile His Leu Lys Lys Lys Ala Val Val Thr Glu Lys Tyr Glu Asp Asp
370 375 380
Arg Arg Lys Ser Phe Lys Lys Ile Gly Ser Phe Ser Leu Glu Gln Leu
385 390 395 400
Gln Glu Tyr Ala Asp Ala Asp Leu Ser Val Val Glu Lys Leu Lys Glu
405 410 415
Ile Ile Ile Gln Lys Val Asp Glu Ile Tyr Lys Val Tyr Gly Ser Ser
420 425 430
Glu Lys Leu Phe Asp Ala Asp Phe Val Leu Glu Lys Ser Leu Lys Lys
435 440 445
Asn Asp Ala Val Val Ala Ile Met Lys Asp Leu Leu Asp Ser Val Lys
450 455 460
Ser Phe Glu Asn Tyr Ile Lys Ala Phe Phe Gly Glu Gly Lys Glu Thr
465 470 475 480
Asn Arg Asp Glu Ser Phe Tyr Gly Asp Phe Val Leu Ala Tyr Asp Ile
485 490 495
Leu Leu Lys Val Asp His Ile Tyr Asp Ala Ile Arg Asn Tyr Val Thr
500 505 510
Gln Lys Pro Tyr Ser Lys Asp Lys Phe Lys Leu Tyr Phe Gln Asn Pro
515 520 525
Gln Phe Met Gly Gly Trp Asp Lys Asp Lys Glu Thr Asp Tyr Arg Ala
530 535 540
Thr Ile Leu Arg Tyr Gly Ser Lys Tyr Tyr Leu Ala Ile Met Asp Lys
545 550 555 560
Lys Tyr Ala Lys Cys Leu Gln Lys Ile Asp Lys Asp Asp Val Asn Gly
565 570 575
Asn Tyr Glu Lys Ile Asn Tyr Lys Leu Leu Pro Gly Pro Asn Lys Met
580 585 590
Leu Pro Lys Val Phe Phe Ser Lys Lys Trp Met Ala Tyr Tyr Asn Pro
595 600 605
Ser Glu Asp Ile Gln Lys Ile Tyr Lys Asn Gly Thr Phe Lys Lys Gly
610 615 620
Asp Met Phe Asn Leu Asn Asp Cys His Lys Leu Ile Asp Phe Phe Lys
625 630 635 640
Asp Ser Ile Ser Arg Tyr Pro Lys Trp Ser Asn Ala Tyr Asp Phe Asn
645 650 655
Phe Ser Glu Thr Glu Lys Tyr Lys Asp Ile Ala Gly Phe Tyr Arg Glu
660 665 670
Val Glu Glu Gln Gly Tyr Lys Val Ser Phe Glu Ser Ala Ser Lys Lys
675 680 685
Glu Val Asp Lys Leu Val Glu Glu Gly Lys Leu Tyr Met Phe Gln Ile
690 695 700
Tyr Asn Lys Asp Phe Ser Asp Lys Ser His Gly Thr Pro Asn Leu His
705 710 715 720
Thr Met Tyr Phe Lys Leu Leu Phe Asp Glu Asn Asn His Gly Gln Ile
725 730 735
Arg Leu Ser Gly Gly Ala Glu Leu Phe Met Arg Arg Ala Ser Leu Lys
740 745 750
Lys Glu Glu Leu Val Val His Pro Ala Asn Ser Pro Ile Ala Asn Lys
755 760 765
Asn Pro Asp Asn Pro Lys Lys Thr Thr Thr Leu Ser Tyr Asp Val Tyr
770 775 780
Lys Asp Lys Arg Phe Ser Glu Asp Gln Tyr Glu Leu His Ile Pro Ile
785 790 795 800
Ala Ile Asn Lys Cys Pro Lys Asn Ile Phe Lys Ile Asn Thr Glu Val
805 810 815
Arg Val Leu Leu Lys His Asp Asp Asn Pro Tyr Val Ile Gly Ile Asp
820 825 830
Arg Gly Glu Arg Asn Leu Leu Tyr Ile Val Val Val Asp Gly Lys Gly
835 840 845
Asn Ile Val Glu Gln Tyr Ser Leu Asn Glu Ile Ile Asn Asn Phe Asn
850 855 860
Gly Ile Arg Ile Lys Thr Asp Tyr His Ser Leu Leu Asp Lys Lys Glu
865 870 875 880
Lys Glu Arg Phe Glu Ala Arg Gln Asn Trp Thr Ser Ile Glu Asn Ile
885 890 895
Lys Glu Leu Lys Ala Gly Tyr Ile Ser Gln Val Val His Lys Ile Cys
900 905 910
Glu Leu Val Glu Lys Tyr Asp Ala Val Ile Ala Leu Glu Asp Leu Asn
915 920 925
Ser Gly Phe Lys Asn Ser Arg Val Lys Val Glu Lys Gln Val Tyr Gln
930 935 940
Lys Phe Glu Lys Met Leu Ile Asp Lys Leu Asn Tyr Met Val Asp Lys
945 950 955 960
Lys Ser Asn Pro Cys Ala Thr Gly Gly Ala Leu Lys Gly Tyr Gln Ile
965 970 975
Thr Asn Lys Phe Glu Ser Phe Lys Ser Met Ser Thr Gln Asn Gly Phe
980 985 990
Ile Phe Tyr Ile Pro Ala Trp Leu Thr Ser Lys Ile Asp Pro Ser Thr
995 1000 1005
Gly Phe Val Asn Leu Leu Lys Thr Lys Tyr Thr Ser Ile Ala Asp Ser
1010 1015 1020
Lys Lys Phe Ile Ser Ser Phe Asp Arg Ile Met Tyr Val Pro Glu Glu
1025 1030 1035 1040
Asp Leu Phe Glu Phe Ala Leu Asp Tyr Lys Asn Phe Ser Arg Thr Asp
1045 1050 1055
Ala Asp Tyr Ile Lys Lys Trp Lys Leu Tyr Ser Tyr Gly Asn Arg Ile
1060 1065 1070
Arg Ile Phe Arg Asn Pro Lys Lys Asn Asn Val Phe Asp Trp Glu Glu
1075 1080 1085
Val Cys Leu Thr Ser Ala Tyr Lys Glu Leu Phe Asn Lys Tyr Gly Ile
1090 1095 1100
Asn Tyr Gln Gln Gly Asp Ile Arg Ala Leu Leu Cys Glu Gln Ser Asp
1105 1110 1115 1120
Lys Ala Phe Tyr Ser Ser Phe Met Ala Leu Met Ser Leu Met Leu Gln
1125 1130 1135
Met Arg Asn Ser Ile Thr Gly Arg Thr Asp Val Asp Phe Leu Ile Ser
1140 1145 1150
Pro Val Lys Asn Ser Asp Gly Ile Phe Tyr Asp Ser Arg Asn Tyr Glu
1155 1160 1165
Ala Gln Glu Asn Ala Ile Leu Pro Lys Asn Ala Asp Ala Asn Gly Ala
1170 1175 1180
Tyr Asn Ile Ala Arg Lys Val Leu Trp Ala Ile Gly Gln Phe Lys Lys
1185 1190 1195 1200
Ala Glu Asp Glu Lys Leu Asp Lys Val Lys Ile Ala Ile Ser Asn Lys
1205 1210 1215
Glu Trp Leu Glu Tyr Ala Gln Thr Ser Val Lys His Lys Arg Pro Ala
1220 1225 1230
Ala Thr Lys Lys Ala Gly Gln Ala Lys Lys Lys Lys Gly Ser Pro Lys
1235 1240 1245
Lys Lys Arg Lys Val Ser Ser Leu Glu His His His His His His
1250 1255 1260
<210> 4
<211> 3706
<212> DNA
<213> AAV Delivery donor (AAV Delivery donor)
<400> 4
gatgtaagga gctgctgtga cttgctcaag gccttatatc gagtaaacgg tagtgctggg 60
gcttagacgc aggtgttctg atttatagtt caaaacctct atcaatgaga gagcaatctc 120
ctggtaatgt gatagatttc ccaacttaat gccaacatac cataaacctc ccattctgct 180
aatgcccagc ctaagttggg gagaccactc cagattccaa gatgtacagt ttgctttgct 240
gggccttttt cccatgcctg cctttactct gccagagtta tattgctggg gttttgaaga 300
agatcctatt aaataaaaga ataagcagta ttattaagta gccctgcatt tcaggtttcc 360
ttgagtggca ggccaggcct ggccgtgaac gttcactgaa atcatggcct cttggccaag 420
attgatagct tgtgcctgtc cctgagtccc agtccatcac gagcagctgg tttctaagat 480
gctatttccc gtataaagca tgagaccgtg acttgccagc cccacagagc cccgcccttg 540
tccatcactg gcatctggac tccagcctgg gttggggcaa agagggaaat gagatcatgt 600
cctaaccctg atcctcttgt cccacagata tccagaaccc tgaccctgcc gtaacgccat 660
tttgcaaggc atggaaaaat accaaaccaa gaatagagaa gttcagatca agggcgggta 720
catgaaaata gctaacgttg ggccaaacag gatatctgcg gtgagcagtt tcggccccgg 780
cccggggcca agaacagatg gtcaccgcag tttcggcccc ggcccgaggc caagaacaga 840
tggtccccag atatggccca accctcagca gtttcttaag acccatcaga tgtttccagg 900
ctcccccaag gacctgaaat gaccctgcgc cttatttgaa ttaaccaatc agcctgcttc 960
tcgcttctgt tcgcgcgctt ctgcttcccg agctctataa aagagctcac aacccctcac 1020
tcggcgcgcc agtcctccga cagactgagt cgcccgggtt aattaagcca ccatggccct 1080
gcctgtgacc gcactgctgc tgcccctggc cctgctgctg cacgctgctc gacccgacat 1140
tcagatgact cagacaacaa gctccctgtc cgcctctctg ggcgacagag tgaccatcag 1200
ctgccgggcc tctcaggata tcagcaagta tctgaactgg taccagcaga agcctgacgg 1260
cacagtgaag ctgctgatct atcacacctc cagactgcac tctggagtgc caagcaggtt 1320
cagcggatcc ggatctggca cagactactc cctgaccatc tctaacctgg agcaggagga 1380
tatcgccaca tatttctgtc agcagggcaa tacactgcca tacacctttg gcggcggcac 1440
aaagctggag atcacccggg cagacgcagc accaaccgtg tctatctttc ccccttctag 1500
caacggcgga ggaggatccg gaggaggagg atctggcggc ggcggcagcg aggtgaagct 1560
gcaggagagc ggaccaggcc tggtggcacc cagccagtcc ctgtctgtga catgcaccgt 1620
gtccggcgtg tctctgcctg attacggcgt gtcttggatc aggcagccac caaggaaggg 1680
cctggagtgg ctgggcgtga tctggggcag cgagacaaca tactataata gcgccctgaa 1740
gtccagactg accatcatca aggacaacag caagtcccag gtgttcctga agatgaacag 1800
cctgcagaca gacgataccg ccatctacta ttgcgccaag cactactatt acggcggcag 1860
ctatgccatg gattactggg gccagggcac atccgtgacc gtgtcctcta tcgatgagca 1920
gaagctgatc agcgaggagg atctgaagcc aaccacaacc cctgcaccaa ggcctccaac 1980
accagcacct accatcgcct ctcagcctct gagcctgcgg cccgaggcct gtaggcccgc 2040
agcaggcggc gccgtgcaca cacgcggcct ggacttcgcc tgcgattttt gggtgctggt 2100
ggtggtggga ggcgtgctgg cctgttattc cctgctggtg accgtggcct tcatcatctt 2160
ttgggtgcgc agcaagcgga gccggctgct gcactctgac tacatgaaca tgacaccaag 2220
acggcccgga ccaacccgga agcactatca gccttacgcc ccacctaggg atttcgcagc 2280
atatcggtcc aagagaggca ggaagaagct gctgtacatc ttcaagcagc cctttatgcg 2340
ccctgtgcag acaacccagg aggaggacgg ctgcagctgt cggttcccag aggaggagga 2400
gggaggatgt gagctgcgcg tgaagttttc tcggagcgcc gatgcacctg catatcagca 2460
gggacagaat cagctgtaca acgagctgaa tctgggcagg cgcgaggagt acgacgtgct 2520
ggataagagg agaggccggg accccgagat gggaggcaag cctaggcgca agaacccaca 2580
ggagggcctg tataatgagc tgcagaagga caagatggcc gaggcctaca gcgagatcgg 2640
catgaaggga gagcggagaa ggggcaaggg acacgatggc ctgtatcagg gcctgtccac 2700
agccaccaag gacacctacg atgccctgca catgcaggcc ctgccaccaa ggtaagaatt 2760
ctgcagatat ccagcacagt ggcggccgct cgagtctaga gggcccgttt aaacccgctg 2820
atcagcctcg actgtgcctt ctagttgcca gccatctgtt gtttgcccct cccccgtgcc 2880
ttccttgacc ctggaaggtg ccactcccac tgtcctttcc taataaaatg aggaaattgc 2940
atcgcattgt ctgagtaggt gtcattctat tctggggggt ggggtggggc aggacagcaa 3000
gggggaggat tgggaagaca atagcaggca tgctggggat gcggtgggct ctatggtaaa 3060
tccagtgaca agtctgtctg cctattcacc gattttgatt ctcaaacaaa tgtgtcacaa 3120
agtaaggatt ctgatgtgta tatcacagac aaaactgtgc tagacatgag gtctatggac 3180
ttcaagagca acagtgctgt ggcctggagc aacaaatctg actttgcatg tgcaaacgcc 3240
ttcaacaaca gcattattcc agaagacacc ttcttcccca gcccaggtaa gggcagcttt 3300
ggtgccttcg caggctgttt ccttgcttca ggaatggcca ggttctgccc agagctctgg 3360
tcaatgatgt ctaaaactcc tctgattggt ggtctcggcc ttatccattg ccaccaaaac 3420
cctcttttta ctaagaaaca gtgagccttg ttctggcagt ccagagaatg acacgggaaa 3480
aaagcagatg aagagaaggt ggcaggagag ggcacgtggc ccagcctcag tctctccaac 3540
tgagttcctg cctgcctgcc tttgctcaga ctgtttgccc cttactgctc ttctaggcct 3600
cattctaagc cccttctcca agttgcctct ccttatttct ccctgtctgc caaaaaatct 3660
ttcccagctc actaagtcag tctcacgcag tcactcatta acccac 3706

Claims (10)

1. The chemically modified CRISPR/Cpf1 complex is prepared from Cpf1 family protein and DBCO modified crRNA; wherein the Cpf1 family protein is AsCpf1, the M806 site of the protein is AeF, and the amino acid sequence of the AsCpf1 protein is shown as SEQ ID NO. 1.
2. Use according to claim 1, wherein the cells are stem cells, embryonic cells, somatic cells and/or immune cells.
3. The use according to claim 2, wherein the cells are NIH-3T3 cells, 293T cells, K562 cells, Jurkat cells, T cells, NK cells or Macrophage cells.
4. A method of gene editing of a cell of non-therapeutic interest, comprising: transforming the chemically modified CRISPR/Cpf1 complex of claim 1 into a cell, and culturing to obtain a gene-edited cell; the chemically modified CRISPR/Cpf1 complex is prepared from a Cpf1 family protein and DBCO modified crRNA; wherein the Cpf1 family protein is AsCpf1, the M806 site of the protein is AeF, and the amino acid sequence of the AsCpf1 protein is shown as SEQ ID NO. 1.
5. The method of gene editing according to claim 4, wherein the transformation method is electrical transformation or chemical transformation.
6. The method of gene editing according to claim 4 or 5, wherein said gene editing is further combined with a plasmid vector, lentivirus or adenovirus-mediated gene transformation method.
7. Use of a chemically modified CRISPR/Cpf1 complex in the manufacture of a medicament for the treatment of a disease; the chemically modified CRISPR/Cpf1 complex is prepared from a Cpf1 family protein and DBCO modified crRNA; wherein the Cpf1 family protein is AsCpf1, the M806 site of the protein is AeF, and the amino acid sequence of the AsCpf1 protein is shown as SEQ ID NO. 1.
8. The use according to claim 7, wherein the disease comprises: immune system diseases or tumors.
9. The use of claim 7, wherein the treatment comprises: autologous cell therapy or allogeneic cell therapy.
10. A medicament for the treatment of a disease comprising a chemically modified CRISPR/Cpf1 complex;
the chemically modified CRISPR/Cpf1 complex is prepared from a Cpf1 family protein and DBCO modified crRNA; wherein the Cpf1 family protein is AsCpf1, the M806 site of the protein is AeF, and the amino acid sequence of the AsCpf1 protein is shown as SEQ ID NO. 1.
CN202110668136.XA 2021-06-16 2021-06-16 Use of chemically modified CRISPR/Cpf1 complex Active CN113355362B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110668136.XA CN113355362B (en) 2021-06-16 2021-06-16 Use of chemically modified CRISPR/Cpf1 complex

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110668136.XA CN113355362B (en) 2021-06-16 2021-06-16 Use of chemically modified CRISPR/Cpf1 complex

Publications (2)

Publication Number Publication Date
CN113355362A CN113355362A (en) 2021-09-07
CN113355362B true CN113355362B (en) 2022-06-07

Family

ID=77534692

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110668136.XA Active CN113355362B (en) 2021-06-16 2021-06-16 Use of chemically modified CRISPR/Cpf1 complex

Country Status (1)

Country Link
CN (1) CN113355362B (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114807230A (en) * 2022-06-06 2022-07-29 朱文敏 Method for knocking out human mesenchymal stem cell TET2 gene by using CRISPR-Cas9 system

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107312761A (en) * 2017-07-18 2017-11-03 江苏溥博生物科技有限公司 A kind of AsCpf1 mutant proteins, encoding gene, recombinant expression carrier and preparation method and application
CN110402287A (en) * 2016-11-22 2019-11-01 综合Dna技术公司 CRISPR/Cpf1 system and method
CN110452893A (en) * 2019-08-19 2019-11-15 南方医科大学 A kind of building and its application of high-fidelity CRISPR/AsCpf1 mutant

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017181107A2 (en) * 2016-04-16 2017-10-19 Ohio State Innovation Foundation Modified cpf1 mrna, modified guide rna, and uses thereof
US11845929B2 (en) * 2016-07-08 2023-12-19 Ohio State Innovation Foundation Modified nucleic acids, hybrid guide RNAs, and uses thereof
BR112019021719A2 (en) * 2017-04-21 2020-06-16 The General Hospital Corporation CPF1 VARIANT (CAS12A) WITH CHANGED PAM SPECIFICITY
AU2018370588B2 (en) * 2017-11-21 2022-09-08 Genkore Co. Ltd. Composition for Genome Editing Using Crispr/CPF1 System and Use Thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110402287A (en) * 2016-11-22 2019-11-01 综合Dna技术公司 CRISPR/Cpf1 system and method
CN107312761A (en) * 2017-07-18 2017-11-03 江苏溥博生物科技有限公司 A kind of AsCpf1 mutant proteins, encoding gene, recombinant expression carrier and preparation method and application
CN110452893A (en) * 2019-08-19 2019-11-15 南方医科大学 A kind of building and its application of high-fidelity CRISPR/AsCpf1 mutant

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
"Highly specific chimeric DNA-RNA guided genome editing with enhanced CRISPR-Cas12a system";Hanseop Kim 等;《BioRxiv》;20210905;全文 *
"Improving the efficiency of CRISPR-Cas12a-based genome editing with site-specific covalent Cas12a-crRNA conjugates";Xinyu Ling 等;《Molecular Cell》;20211118;全文 *
"Improving the efficiency of precise genome editing with site-specific Cas9-oligonucleotide conjugates";Xinyu Ling 等;《Science Advances》;20200408;全文 *

Also Published As

Publication number Publication date
CN113355362A (en) 2021-09-07

Similar Documents

Publication Publication Date Title
EP3674328B1 (en) Chimeric antigen receptor (car) binding to bcma, and uses thereof
EP3219800A1 (en) A transposon-based transfection system for primary cells
KR20200015939A (en) Expression of Novel Cell Tags
KR20210043562A (en) ROR-1 specific chimeric antigen receptor and uses thereof
JP7235391B2 (en) artificially engineered immune cells
KR20190130608A (en) Compositions and Methods for Immune Oncology
WO2019080538A1 (en) Chimeric antigen receptor, nk cell modified by same, coding dna, mrna, expression vector, preparation method and application
WO2019113132A1 (en) Modified lymphocytes
KR20220115602A (en) Methods and compositions for regulated armor of cells
AU2016373365A1 (en) Transposon system, kit comprising the same, and uses thereof
CN110923255A (en) Chimeric antigen receptor targeting BCMA and CD19 and uses thereof
CN113355362B (en) Use of chemically modified CRISPR/Cpf1 complex
CN111848819A (en) MSLN-targeted chimeric antigen receptor and application thereof
CN114729320B (en) Compositions, methods and uses for reprogramming cells to dendritic cell type 2 capable of presenting antigen
WO2020103777A1 (en) Interleukin 21 protein (il21) mutant and use thereof
CN116083435A (en) Antitumor application based on humanized LIGHT truncated mutant protein
CN112538462B (en) Cell membrane for rapid amplification of NK cells and application thereof
WO2022022113A1 (en) Chimeric antigen receptor and use thereof
CN114907485B (en) Chimeric antigen receptor using endogenous protein molecules to replace single domain antibodies
CN113754756B (en) An antibody that recognizes HLA-A0229-38TCR and uses thereof
CN114591963B (en) sgRNA and method for constructing GM-CSF (-) cell by using same
CN116731155B (en) Human papillomavirus-specific T cell receptor, cells expressing same and uses thereof
CN113801228B (en) CD 38-targeted single-chain antibody, fully human chimeric antigen receptor, preparation method and application
WO2024050551A2 (en) Compositions and methods for in vivo expression of chimeric antigen receptors
CN116284409A (en) GPC3CAR-T cells and their use in the preparation of a medicament for the treatment of cancer

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant