CN113388042B - Recombinant protein, recombinant expression vector, recombinant cell and NK cell activated magnetic bead as well as preparation method and application thereof - Google Patents

Recombinant protein, recombinant expression vector, recombinant cell and NK cell activated magnetic bead as well as preparation method and application thereof Download PDF

Info

Publication number
CN113388042B
CN113388042B CN202110715596.3A CN202110715596A CN113388042B CN 113388042 B CN113388042 B CN 113388042B CN 202110715596 A CN202110715596 A CN 202110715596A CN 113388042 B CN113388042 B CN 113388042B
Authority
CN
China
Prior art keywords
cell
cells
recombinant
magnetic beads
expression vector
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
CN202110715596.3A
Other languages
Chinese (zh)
Other versions
CN113388042A (en
Inventor
焦顺昌
张嵘
袁翰
吕鹏敏
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.)
Beijing Dingcheng Taiyuan Biotechnology Co ltd
Original Assignee
Beijing Dingcheng Taiyuan Biotechnology Co ltd
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 Beijing Dingcheng Taiyuan Biotechnology Co ltd filed Critical Beijing Dingcheng Taiyuan Biotechnology Co ltd
Priority to CN202110715596.3A priority Critical patent/CN113388042B/en
Publication of CN113388042A publication Critical patent/CN113388042A/en
Application granted granted Critical
Publication of CN113388042B publication Critical patent/CN113388042B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K17/00Carrier-bound or immobilised peptides; Preparation thereof
    • C07K17/14Peptides being immobilised on, or in, an inorganic carrier
    • 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
    • C12N15/66General methods for inserting a gene into a vector to form a recombinant vector using cleavage and ligation; Use of non-functional linkers or adaptors, e.g. linkers containing the sequence for a restriction endonuclease
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • C12N5/0681Cells of the genital tract; Non-germinal cells from gonads
    • C12N5/0682Cells of the female genital tract, e.g. endometrium; Non-germinal cells from ovaries, e.g. ovarian follicle cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/20Fusion polypeptide containing a tag with affinity for a non-protein ligand
    • C07K2319/22Fusion polypeptide containing a tag with affinity for a non-protein ligand containing a Strep-tag
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2302Interleukin-2 (IL-2)
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2315Interleukin-15 (IL-15)
    • 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
    • C12N2510/02Cells for production

Landscapes

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

Abstract

The invention provides a recombinant protein, a recombinant expression vector, a recombinant cell, an NK cell activation magnetic bead, a preparation method and an application thereof, and relates to the technical field of NK cell activation propagation. The invention fuses the single-chain antibody and the Streptag2 label, has simple structure, ensures that the antibody expressed by the recombinant expression vector or the recombinant cell can be directly assembled with the magnetic bead without biotinylation modification, and the assembled magnetic bead can be directly used as an amplification stimulator of the NK cell, thereby effectively amplifying the NK cell. The NK cell activating magnetic bead has the binding capacity of more than 97 percent and high specificity; the killing efficiency of the NK cell line obtained by activating and culturing the NK cell activated magnetic beads is not obviously different from that of the NK cell activated and cultured by a feeder cell method.

Description

Recombinant protein, recombinant expression vector, recombinant cell and NK cell activated magnetic bead as well as preparation method and application thereof
Technical Field
The invention belongs to the technical field of NK cell activation propagation, and particularly relates to a recombinant protein, a recombinant expression vector, a recombinant cell, an NK cell activation magnetic bead, a preparation method and an application thereof.
Background
Natural killer cells (NK) are an innate lymphocyte that can spontaneously kill tumor cells without antigen stimulation or prior sensitization, playing a key role in antiviral and antitumor immunity. The immunophenotypic characteristics are mainly CD3-CD56+Is a subpopulation of lymphocytes that do not express a T cell receptor nor a B cell receptor. NK cells can kill tumor cells or virally infected cells through a variety of pathways, including direct cytotoxicity (both natural and antibody-dependent cell-mediated cytotoxicity) and indirect effects (such as cytokine production and interaction with adaptive immunity). Because of its broad spectrum and high lethality, NK cells have a very promising application prospect and are expected to become a new strategy for adoptive immunotherapy for treating tumors, and therefore, it is very necessary to develop a method for efficiently and stably amplifying NK cells in vitro.
NK cells account for about 5-10% of lymphocytes in blood, and sufficient cell number and cell purity in cell therapy are important factors of the effect of NK cells in killing tumor cells, but the proliferation of NK cells is weak, so that sufficient cells cannot be provided for administration, and clinical requirements cannot be met, so that most of treatment schemes depending on NK cells are in an early development stage, which becomes the biggest problem in clinical application. At present, the method with the highest NK cell in-vitro activation efficiency is a method of adding a feeder cell for inactivating K562 feeder cells, the positive rate of NK cells after 2-3 weeks of culture can be up to more than 90%, but inactivated feeder cells are difficult to remove, and the treatment level is difficult to achieve. In recent two years, some recombinant cytokine culture schemes are also available on the market, most of the recombinant cytokine culture schemes can stably achieve about 40-50% of positive rate, but a large amount of T cells can proliferate in the culture process, influence the subsequent use effect, possibly bring the risk of acute graft-versus-host disease (GVHD), and achieve more than 80% of positive rate, or the amplification efficiency is general, or the individual difference is serious.
Disclosure of Invention
In view of the above, the present invention is directed to a recombinant protein, a recombinant expression vector, a recombinant cell, and an NK cell-activated magnetic bead, and a preparation method and an application thereof, wherein the recombinant protein or an antibody obtained from the recombinant expression vector and the recombinant cell can be directly assembled with the magnetic bead, and the assembled magnetic bead can be directly used as an NK cell amplification stimulator to efficiently amplify NK cells.
In order to achieve the above object, the present invention provides the following technical solutions:
the invention provides a recombinant protein, the structure of which comprises a single-chain antibody NM3E2And Streptag 2.
Preferably, in the single chain antibody NM3E2The N terminal of the (B) is connected with an IL21 signal peptide, and the C terminal is connected with IgG1 to form IL21-NM3E2IgG1, the IL21-NM3E2The encoding gene of IgG1 comprises the nucleotide sequence shown in SEQ ID NO. 4.
Preferably, the coding gene of the IL21 signal peptide comprises a nucleotide sequence shown in SEQ ID NO. 1; single chain antibody NM3E2The coding gene of (A) comprises a nucleotide sequence shown in SEQ ID NO. 2.
Preferably, the encoding gene of Streptag2 includes the nucleotide sequence shown in SEQ ID NO. 3.
The invention also provides a recombinant expression vector for expressing the recombinant protein, and a basic vector of the recombinant expression vector comprises a eukaryotic expression vector pFUSE-hIgG 1-Fc.
The invention also provides a construction method of the recombinant expression vector, which comprises the following steps: (1) adding a coding gene sequence of Streptag2 between an Fc gene and a stop codon TGA of a eukaryotic expression vector pFUSE-hIgG1-Fc to obtain a vector pFUSE-hIgG1-Fc-Streptag 2;
(2) IL21-NM3E2The coding gene of IgG1 is inserted between the Nco I and BgII enzyme cutting sites of the vector pFUSE-hIgG1-Fc-Streptag2 to obtain the recombinant expression vector; the IL21-NM3E2The encoding gene of IgG1 comprises the nucleotide sequence shown in SEQ ID NO. 4.
The invention also provides a recombinant engineering cell for expressing the recombinant protein, and the basic cell of the recombinant engineering cell comprises an expiCHO cell.
The invention also provides a preparation method of the NK cell activated magnetic bead, which comprises the following steps: and inducing the recombinant engineering cells to generate recombinant proteins, purifying and reacting with activated magnetic beads to obtain NK cell activated magnetic beads.
Preferably, the ratio of the number of activated magnetic beads to the purified recombinant protein per 150. mu.L of the reaction system is 1X 107The method comprises the following steps: 15 μ g.
The invention also provides the NK cell activated magnetic bead prepared by the preparation method.
The invention also provides application of the NK cell activation magnetic bead in-vitro culture of NK cells.
The invention also provides a method for culturing NK cells in vitro, which comprises the following steps: mixing human peripheral blood mononuclear cells and the NK cell activated magnetic beads according to the number ratio of 1:1, and then culturing;
the culture medium for culture takes Human Lymphocytes Serum Free Media culture medium as a basic culture medium and also comprises: 1000IU/mL of cell culture factor IL-2, 10ng/mL of cell culture factor IL-15 and 10 percent of fetal bovine serum FBS.
Preferably, the culture conditions comprise: 37 ℃ and 5% CO2And (4) concentration.
The invention also provides the NK cell line cultured by the method.
The invention also provides application of the NK cell line in preparation of a medicine for killing tumor cells or virus-infected cells.
The invention provides a recombinant protein, which is characterized in that a single-chain antibody and a Streptag2 label are fused, the structure is simple, the recombinant expression vector or the antibody expressed by recombinant cells can be directly assembled with magnetic beads without biotinylation modification, and the assembled magnetic beads can be directly used as an amplification stimulator of NK cells, so that the NK cells can be effectively amplified. In the embodiment of the invention, the binding condition of the magnetic beads and the recombinant protein is detected by adopting flow cytometry, the binding capacity is up to more than 97%, the specificity of the magnetic beads and the recombinant protein is high, and the magnetic beads and the recombinant protein can be used for in-vitro activation experiments of NK cells. The embodiment of the invention utilizes peripheral blood mononuclear cells (PBMC cells) and the assembled magnetic beads to carry out in-vitro activation and amplification on NK cells, pure NK cells are amplified 7 times after in-vitro activation culture for 14 days, and other cells (such as T cells) are also amplified in the activation culture process because cells used for activation initiation are PBMC cells. Meanwhile, the detection result of the flow cytometry shows that the classical NK cell (namely CD 3) is cultured for 14 days-CD56+Cell population) of 63.6%, the proportion of pure NK cells (CD 3)-CD16+CD56+Cell population) was 73.1%. The embodiment of the invention also performs an in-vitro killing experiment on the obtained NK cell line, the cell culture lasts for 7 days, and after killing for 4 hours, the effective-to-target ratio is 2: at 1, the killing efficiency of the experimental group is 77.04%; the effective target ratio is 1: the killing efficiency of the experimental group was 46.95% at 1. Therefore, the NK cell line obtained by the invention has obvious killing activity on K-562-luc2 cells. In conclusion, the scheme of the invention can obtain a large amount of NK cells with remarkable killing activity, and can be used for clinically killing tumor cells or viral infectionThe cells promote the clinical application and development of NK cells.
Drawings
FIG. 1 is a structural diagram of NK cell activated magnetic beads obtained by the present invention;
FIG. 2 is an SDS-PAGE electrophoresis of recombinant proteins;
FIG. 3 is a diagram showing the flow analysis of the binding effect of magnetic beads and antibodies;
FIG. 4 shows the cell counts of PBMC cells cultured and sampled on days 1, 2, 3, 6, 10 and 14;
FIG. 5 shows the growth of PBMC cells after culture, under a microscope on days 1, 2, 3, 6, 10 and 14, wherein A: cell culture 1 day picture; b: cell culture 2 days; c: cell culture 3 days; d: cell culture 6 days; e: cell culture 10 days; f: cell culture 14 days;
FIG. 6 shows the results of the flow cytometry for detecting cell phenotype;
FIG. 7 is a graph showing the results of the in vitro cell killing experiment on day 7;
FIG. 8 shows the results of the GraphPadprism software analysis of comparative example 1;
FIG. 9 shows the results of the flow cytometry test of comparative example 2;
FIG. 10 is a graph showing the results of the in vitro cell killing experiments in comparative example 3 and example 2.
Detailed Description
The invention provides a recombinant protein, which comprises a single-chain antibody NM3E2And Streptag 2.
The invention is in the single-chain antibody NM3E2The N end of the polypeptide is preferably also connected with an IL21 signal peptide, and the coding gene of the IL21 signal peptide comprises a nucleotide sequence shown in SEQ ID NO. 1; in the single-chain antibody NM3E2Preferably, the C-terminus of (A) is further linked to IgG1 to form IL21-NM3E2IgG 1. In the present invention, the IL21 signal peptide is preferably derived from human, and can promote the secretory expression of the recombinant protein in mammalian cells. In the present invention, the recombinant protein is also referred to as recombinant NM3E2Streptag2 protein. Single chain antibody NM of the invention3E2The coding gene of (1) preferably comprises the nucleotide sequence shown in SEQ ID NO. 2. In order to make IL21-NM3E2IgG can be coupled directly to magnetic beads using Streptag2 with IL21-NM3E2IgG, the encoding gene of Streptag2 preferably comprises the nucleotide sequence shown in SEQ ID NO. 3. The gene sequence of IL21 is not particularly limited in the invention, and is preferably shown in SEQ ID NO. 1. The invention is directed to said IL21-NM3E2The gene encoding IgG1 is not particularly limited, and is preferably synthesized by Kingzhi Biotech Co., Ltd. In the present invention, the nucleotide sequences are all 5 'to 3' unless otherwise specified.
The invention also provides a recombinant expression vector for expressing the recombinant protein, and a basic vector of the recombinant expression vector comprises a eukaryotic expression vector pFUSE-hIgG 1-Fc.
The eukaryotic expression vector pFUSE-hIgG1-Fc is a transient expression vector of mammalian cell protein, and the source of the basic vector is not particularly limited in the invention, and is preferably purchased from Invivogen company. The invention names the recombinant expression vector NM3E2Streptag2 eukaryotic expression vector.
The invention also provides a construction method of the recombinant expression vector, which comprises the following steps: (1) adding a coding gene sequence of Streptag2 between an Fc gene and a stop codon TGA of a eukaryotic expression vector pFUSE-hIgG1-Fc to obtain a vector pFUSE-hIgG1-Fc-Streptag 2;
(2) IL21-NM3E2The coding gene of IgG1 is inserted between the Nco I and BgII enzyme cutting sites of the vector pFUSE-hIgG1-Fc-Streptag2 to obtain the recombinant expression vector; the IL21-NM3E2The encoding gene of IgG1 comprises the nucleotide sequence shown in SEQ ID NO. 4.
According to the invention, a coding gene sequence of Streptag2 is added between an Fc gene of a eukaryotic expression vector pFUSE-hIgG1-Fc and a stop codon TGA to obtain a vector pFUSE-hIgG1-Fc-Streptag 2. The invention preferably entrusts Jinwei Zhi Biotech Co Ltd to modify an expression vector pFUSE-hIgG1-Fc, adds Streptag2 nucleotide sequence between Fc gene and termination codon TGA, and obtains the vector pFUSE-hIgG1-Fc-Streptag 2.
After obtaining the carrier pFUSE-hIgG1-Fc-Streptag2, the invention adds IL21-NM3E2The coding gene of IgG1 is inserted between the Nco I and BgII enzyme cutting sites of the vector pFUSE-hIgG1-Fc-Streptag2 to obtain the recombinant expression vector.
The present invention preferably uses the above IL21-NM3E2The coding gene of IgG1 is synthesized and directionally cloned to pFUSE-hIgG1-Fc-Streptag2 expression vector after NcoI and BgII linearization treatment, and the recombinant expression vector is obtained.
The invention uses the recombinant expression vector to transform DH5 alpha, and then obtains the recombinant plasmid NM after the plasmid is extracted3E2Streptag2 inpFUSE-hIgGle3-Fc。
The invention also provides a recombinant engineering cell for expressing the recombinant protein, and the basic cell of the recombinant engineering cell comprises an expiCHO cell.
The invention preferably utilizes a transfection method to construct the recombinant engineering cell, and specifically comprises the following steps: and transforming the recombinant expression vector into escherichia coli DH5 alpha, and selecting a plurality of clones for sequencing identification. The correctly sequenced recombinants (recombinant plasmids) were then placed in plasmids and used for transfection of expichcho cells (purchased from Thermo Fisher Scientific).
The transfection according to the invention preferably comprises electroporation, more preferably by means of electroporation with reference to the instructions of the electrotransformation apparatus Celetrix-CTX-1500A.
The invention also provides a method for expressing recombinant protein by using the recombinant engineering cell, which preferably comprises the following steps: after the electrotransfer is finished, the cell liquid in the electric shock tube is transferred to a pre-added 3mL solution
Figure BDA0003135035790000061
CHO CD04 Medium complete Medium (C.) (II)
Figure BDA0003135035790000062
CHO CD04 Medium basal Medium + 1% GlutaMAX from QuaCell Corp.) in six-well platesPlacing the six-hole plate at 37 deg.C and 5% CO2The cell culture chamber (purchased from Thermo Fisher Scientific Co., Ltd.) was used to culture the cells after electroporation at a shaker rotation speed of 105rpm, and after 5 days, the culture supernatant was collected and examined for the expression of the target protein. After obtaining the recombinant protein, the invention preferably further comprises protein purification, and more preferably, the protein purification is carried out by using a ProteinA affinity chromatography column.
The invention also provides a preparation method of the NK cell activated magnetic bead, which comprises the following steps: and inducing the recombinant engineering cells to generate recombinant proteins, purifying and reacting with activated magnetic beads to obtain NK cell activated magnetic beads.
The induction according to the present invention is preferably the same as the above-described method for expressing a recombinant protein, and will not be described herein. In the present invention, it is preferable that the protein is purified by the protein affinity column and then the purified protein obtained is reacted with activated magnetic beads. The ratio of the number of activated magnetic beads to the purified recombinant protein in the reaction system of 150. mu.L is preferably 1X 107The method comprises the following steps: 15 μ g.
In the present invention, the method for activating magnetic beads preferably includes: the stock solution of magnetic beads (Strep-Tactin mag. Microbeads, available from IBA) was shaken on a vortex shaker for 30s to obtain a solution containing about 1X 10 beads7And (3) placing the stock solution of the magnetic beads in a 1.5mLEP tube, adding 1mLBinding Buffer, blowing, uniformly mixing, carefully abandoning the supernatant on a magnetic frame, and repeatedly using the Bindingbuffer for cleaning once.
The reaction time of the present invention is preferably 1 hour, and the working concentration of the recombinant protein is at least 100. mu.g/mL when the reaction is carried out. After the reaction is finished, the method preferably further comprises washing once with a Binding Buffer, resuspending antibody-coupled magnetic beads (or called NK cell activated magnetic beads) in 1mL of 1 × PBS solution, counting and calculating the concentration of the antibody-coupled magnetic beads, and storing in a refrigerator at 4 ℃ for later use. The NK cell activation magnetic bead has the binding capacity of more than 97 percent and high specificity through flow cytometry detection, and can be used for in vitro culture and amplification of NK cells.
The invention also provides the NK cell activated magnetic bead prepared by the preparation method.
The structure of the NK cell-activating magnetic bead of the present invention is preferably as shown in FIG. 1.
The invention also provides application of the NK cell activation magnetic bead in-vitro culture of NK cells.
The invention also provides a method for culturing NK cells in vitro, which comprises the following steps: mixing human peripheral blood mononuclear cells (PBMC cells) and the NK cell activated magnetic beads according to the number ratio of 1:1, and culturing;
the culture medium for culture takes Human Lymphocytes Serum Free Media culture medium (LY-08 culture medium for short) as a basal culture medium and also comprises: 1000IU/mL of cell culture factor IL-2, 10ng/mL of cell culture factor IL-15 and 10 percent of fetal bovine serum FBS.
In the present invention, the source of the PBMC cells is not particularly limited, and the PBMC cells can be extracted by themselves, but in the present embodiment, human PBMC cells are preferably extracted by collecting peripheral blood of the elbow vein of a volunteer and separating the blood by density gradient centrifugation using a Ficoll lymphocyte separation medium (purchased from TBD).
The present invention preferably uses the culture medium to resuspend the PBMC cells and adjust the cell density to 1.5X 106mL, following human PBMC cells: NK cell activating magnetic beads ═ 1:1, inoculating the human PBMC cells and the NK cell activated magnetic beads into a six-hole plate, uniformly mixing, and placing the six-hole plate at the temperature of 37 ℃ and 5% CO2The cell culture box of (2) is used for carrying out in-vitro amplification culture. The invention carries out complete liquid change after the culture for 48 hours, then carries out liquid supplement or liquid change treatment every other day, each liquid supplement or liquid change culture medium contains the cell culture factor, and the cell density is maintained at 1 x 106/mL。
The invention also provides the NK cell line cultured by the method.
The invention adopts flow cytometry to detect the NK cell line, and shows that the classical NK cell (namely CD 3) is cultured for 14 days-CD56+Cell population) of 63.6%, the proportion of pure NK cells (CD 3)-CD16+CD56+Cell population) was 73.1%, and the amplification effect was good.
The invention also provides application of the NK cell line in preparation of a medicine for killing tumor cells or virus-infected cells.
The NK cell line has strong killing capacity on tumor cells, and has no obvious difference from the killing efficiency of NK cells activated and cultured by a feeder cell method.
The following examples are provided to illustrate a recombinant protein, a recombinant expression vector, a recombinant cell and an NK cell-activated magnetic bead, and methods for preparing and using the same, but they should not be construed as limiting the scope of the present invention.
Example 1
One, NM3E2Construction of Streptag2 eukaryotic expression vector
The expression vector was modified to pFUSE-hIgG1-Fc-Streptag2 by Kingzhi Biotech, Inc. by adding Streptag2 nucleotide sequence (SEQ ID NO.3) between the Fc gene of expression vector pFUSE-hIgG1-Fc (purchased from Invivogen) and the stop codon TGA.
Synthesis of IL21-NM by Jinzhi Biotechnology Ltd3E2-IgG1(SEQ ID NO.4)。
Synthesizing IL21-NM from whole gene3E2IgG1 was directionally cloned into pFUSE-hIgG1-Fc-Streptag2 expression vector linearized with Nco I and BgIIL, transformed into E.coli DH 5. alpha. and several clones were picked for sequencing identification. The correctly sequenced recombinants (recombinant plasmids) were then placed in plasmids and used for transfection of expichcho cells (purchased from Thermo Fisher Scientific).
By sequencing, IL21-NM3E2The full-length gene sequence of IgG is identical to that of SEQ ID NO.4, and is consistent with the expectation.
II, recombination NM3E2Expression and purification of Streptag2 protein
1. Recombinant NM3E2Expression of Streptag2 protein
expiCHO cells were counted and 2X 10 cells were taken7Individual expihcho cells were used for plasmid transfection. Following an electropolymerization Buffer 103 partA:transfection Buffer was prepared at a ratio of 1:1 (available from Celetrix corporation) for electrophoresis Buffer 103part B, and recombinant plasmid NM was taken3E2Streptag2 inpFSUSE-hIgGle 3-Fc, adding the prepared transfection buffer solution and the prepared recombinant plasmid into the cell sediment, uniformly mixing, transferring into a 120 μ L electric shock tube (purchased from Celetrix company), operating with reference to the instruction of an electrotransfer Celetrix-CTX-1500A during electric shock, transferring the cell liquid in the electric shock tube into 3mL of the electric shock tube after the electric shock is finished
Figure BDA0003135035790000082
CHO CD04 Medium complete Medium (C.) (II)
Figure BDA0003135035790000081
CHO CD04 Medium basal Medium + 1% GlutaMAX from QuaCell corporation) were placed in a six-well plate at 37 ℃ with 5% CO2The cell culture chamber (purchased from Thermo Fisher Scientific Co., Ltd.) was used to culture the cells after electroporation at a shaker rotation speed of 105rpm, and after 5 days, the culture supernatant was collected and examined for the expression of the target protein.
2. Recombinant NM3E2Purification of Streptag2 protein
Adding a buffer solution of 20mM PB and 200mM NaCl into the cell culture supernatant after transfection to adjust the pH to 7.5;
protein purification chromatography column: ProteinA affinity chromatography column (available from GE Healthcare, column volume 1.0 mL);
buffer a (buffera): PBS, pH 7.4;
buffer b (bufferb): 0.1M Glycine, pH 3.0;
buffer c (buffer c): 0.1M Glycine, pH 2.7.
And (3) purification process: the protein affinity column was pretreated with buffer A using AKTAexplorer 100 type protein purification system (available from GE Healthcare), and the culture supernatant collected above was loaded after adjusting pH to 7.5 with 20mM PB buffer and 200mM NaCl, and the effluent was collected. After the sample loading is finished, the column is equilibrated by using at least 1.5mL of Buffer A, after the equilibration, the column is respectively eluted by using Buffer B and Buffer C, the target protein eluent is collected (1% of 1M Tris is required to be added in advance to a collection tube of the eluent, the pH value of the eluent is neutralized by 8.0, and the final concentration of Tris is about 10mM), and finally, the concentrated dialysis is carried out to the Buffer PBS.
The results show that the final purified recombinant NM3E2The Streptag2 protein was analyzed by SDS-PAGE electrophoresis, the SDS-PAGE electrophoresis under reducing and non-reducing conditions is shown in FIG. 2, and after purification by protein affinity chromatography column, the recombinant NM protein3E2The purity of Streptag2 protein is as high as 95%.
Preparation of magnetic beads
The stock solution of magnetic beads (Strep-Tactin mag. Microbeads, available from IBA) was shaken on a vortex shaker for 30s to obtain a solution containing about 1X 10 beads7Adding 1mL Binding Buffer into the stock solution of the magnetic beads in a 1.5mL EP tube, blowing and uniformly mixing, carefully discarding supernatant on a magnetic frame, and repeatedly using the Binding Buffer to clean once; adding the recombinant NM purified by a ProteinA affinity chromatography column3E2Streptag2 protein into activated magnetic beads, mixing, reacting at room temperature for 1h, wherein the recombinant NM is added3E2The working concentration of Streptag2 protein is at least 100 μ g/mL; washing once by using Binding Buffer, resuspending the antibody-coupled magnetic beads in 1mL of 1 XPBS solution, counting and calculating the concentration of the antibody-coupled magnetic beads (the structure is shown in figure 1), and storing in a refrigerator at 4 ℃ for later use.
Taking the magnetic beads coupled with the antibody in a flow tube, and detecting the magnetic beads and NM by adopting flow cytometry3E2Binding of Streptag2 recombinant proteins. The flow cytometry analysis results are shown in FIG. 3, which shows that the magnetic beads Strep-Tactin mag. Microbeads and recombinant NM of IBA3E2The binding capacity of Streptag2 protein is up to 97%, and the specificity of both is high.
Example 2
In vitro amplification culture of NK cells
Collecting peripheral blood of elbow vein of volunteer, separating and extracting human PBMC cells by density gradient centrifugation with Ficoll lymphocyte separation medium (purchased from TBD), adding fetal bovine serum FBS (purchased from gibco) and IL-2Human lymphocyes Serum Free Media (LY-08) (available from Gilles Biotech, hereinafter abbreviated as LY-08 culture medium) for (Human recombinant interleukin-2 for injection, available from Erlu pharmaceutical Co.) and IL-15 (available from novoprotein Co.) were resuspended, followed by cell counting, and the cell density was adjusted to 1.5X 106mL, following human PBMC cells: magnetic beads are 1:1, inoculating the PBMC cells and the magnetic beads into a six-hole plate, uniformly mixing, and placing the six-hole plate at the temperature of 37 ℃ and 5% CO2The cell culture box of (2) is used for carrying out in-vitro amplification culture. Wherein the working concentration of the added cell culture factor IL-2 is 1000IU/mL, the working concentration of IL-15 is 10ng/mL, and the working concentration of the fetal bovine serum FBS is 10%. Completely changing the culture medium after 48h, and supplementing or changing the culture medium every other day, wherein the cell culture factor is contained in each supplemented or changed culture medium, and the cell density is maintained at 1 × 106mL, with cell counting. Then at 0d, 7d and 14d, respectively, taking the cells, performing cell phenotype detection by using flow cytometry, and counting cell proliferation times, wherein the cell phenotype detects the classical NK cell phenotype (namely CD 3)-CD56+The cell population of (a).
As shown in FIGS. 4-6, the pure NK cells were amplified 7 times after 14 days of in vitro activation culture, and other cells (such as T cells) were also amplified during the activation culture process because the cells used for the activation initiation are PBMC cells. Meanwhile, the detection result of the flow cytometry shows that the classical NK cell (namely CD 3) is cultured for 14 days-CD56+Cell population) of 63.6%, the proportion of pure NK cells (CD 3)-CD16+CD56+Cell population) was 73.1%.
Example 3
In vitro killing experiment of NK cells
NK cells cultured for 7 days in example 2 were collected and effector cell density was adjusted to 4X 106Perml and 2X 106Per mL; the target cells, human myeloid leukemia cell K-562-luc2 (purchased from ATCC company,
Figure BDA0003135035790000111
CCL-243-LUC2TM) And adjusting the density of the target cells to 2X 106mL, i.e. effective target ratio of 2: 1 and 1: 1; according to different effective target ratios, respectively adding a total of 200 mu L of effector cells and target cells with different concentrations into a round-bottom 96-well plate, and dividing the effector cells and the target cells into two groups:
target cell group: 100 μ L of target cells +100 μ L of LY-08 complete medium, 3 multiple wells per group;
experimental groups: mu.L of target cells + 100. mu.L of NK cells cultured for 7 days in example 1, 3 duplicate wells per group;
placing the two groups of mixed cells at 37 deg.C and 5% CO2Cultured for 4h in a cell culture box and utilizes One-LiteTMThe Luciferaaseassay system kit (from Vazyme) detected the remaining K-562-luc2 cells and the killing efficiency of NK cells was calculated according to the formula (results are expressed as mean of 3 replicates).
The killing efficiency calculation formula is as follows: killing efficiency ═ 100% for (1-experimental/target cell).
After the cells are cultured for 7 days, the results of in vitro killing experiments are shown in figure 7, and after 4 hours of killing, the effective-to-target ratio is 2: at 1, the killing efficiency of the experimental group is 77.04%; the effective target ratio is 1:1, the killing efficiency of the experimental group is 46.95%, and the NK cells activated and cultured by the invention have obvious killing activity on K-562-luc2 cells.
Comparative example 1
Antibody-coupled magnetic beads and recombinant NM3E2Effect of Streptag2 protein formulation
1) Recombinant NM3E2Dilution of Streptag2 protein
Recombination of NM3E2Streptag2 protein was diluted using a gradient method according to 1: 2 was diluted down with 1 x PBS solution for 10 concentrations, the highest concentration being the original concentration, i.e.: 284. mu.g/mL, stored at 4 ℃ in a refrigerator protected from light for later use.
2) Flow cytometry analysis
Taking equivalent magnetic bead Strep-Tactinmag. Microbeads, and respectively adding the recombinant NM with different concentrations3E2The Streptag2 protein was coupled, i.e.: mixing magnetic bead stock solution (Strep-Tactin mag. micro)beads) was oscillated on a vortex oscillator for 30s, which was taken to include about 1X 107Adding 1mLBinding Buffer into a 1.5mL EP tube to blow and mix uniformly, carefully abandoning supernatant on a magnetic frame, and repeatedly using the Binding Buffer to clean once; adding the above different concentrations of recombinant NM3E2Mixing Streptag2 protein into activated magnetic beads, and reacting at room temperature for 1 h; washing once by using Binding Buffer, adding a flow-type secondary antibody PE anti-human IgG Fc (purchased from BioLegend), and incubating for 20min at room temperature in a dark place; taking antibody-coupled magnetic beads in a flow tube, and detecting the magnetic beads and NM (nanometer) with different concentrations by adopting flow cytometry3E2Binding of Streptag2 recombinant proteins.
3) Data processing and analysis
Data analysis was performed using FlowJo V10 software and GraphPad Prism software.
Analysis of the data by FlowJo V10 software (table 1) and GraphPad Prism software (fig. 8) after detection by flow cytometry showed that recombinant NM was present at different concentrations3E2Microbeads of the Streptag2 protein, the highest binding ability and sensitivity to the magnetic beads Strep-Tactin mag of IBA, was the original concentration of recombinant NM3E2Streptag2 protein, i.e. added recombinant NM3E2The working concentration of Streptag2 protein was 284 μ g/mL, followed by a concentration of 1: 2 equal dilution of the first two gradients, the three concentrations of beads and recombinant protein were all more specific, therefore, in antibody recombination NM3E2Recombinant NM added when Streptag2 protein was coupled to magnetic beads3E2The working concentration of Streptag2 protein was at least 100. mu.g/mL.
TABLE 1FlowJo V10 software analysis results
Concentration (μ g/mL) Geometric mean (PE-A)
284 13876
142 13060
71 15046
35.5 12871
17.75 8166
8.875 2847
4.4375 387
2.21875 59.8
1.109375 34.7
0.5546875 31.4
0.27734375 6036
0.138671875 6552
Comparative example 2
The problem of the proportion of magnetic beads to cells during the activation of NK cells
1) Preparation of magnetic beads
Shaking the stock solution of magnetic beads (Strep-Tactin mag. Microbeads) on a vortex oscillator for 30s to obtain a solution containing about 1 × 107Adding 1mL Binding Buffer into the stock solution of the magnetic beads in a 1.5mL EP tube, blowing and uniformly mixing, carefully discarding supernatant on a magnetic frame, and repeatedly using the Binding Buffer to clean once; addition of recombinant NM3E2Streptag2 protein into activated magnetic beads, mixing, reacting at room temperature for 1h, wherein the recombinant NM is added3E2The working concentration of Streptag2 protein is at least 100 μ g/mL; washing once by using Binding Buffer, resuspending the antibody-coupled magnetic beads in 1mL of 1 × PBS solution, counting and calculating the concentration of the antibody-coupled magnetic beads, and storing in a refrigerator at 4 ℃ for later use.
2) Extraction of human PBMC cells
Collecting peripheral blood of elbow vein of volunteer, separating and extracting human PBMC cell by density gradient centrifugation with Ficoll lymphocyte separation solution, suspending with LY-08 culture medium containing fetal bovine serum FBS, IL-2 and IL-15, counting cells, and adjusting cell density to 1.5 × 106and/mL. Wherein the working concentration of the added cell culture factor IL-2 is 1000IU/mL, the working concentration of IL-15 is 10ng/mL, and the working concentration of the fetal bovine serum FBS is 10%.
3) Inoculation of
Respectively taking 6 × 1062, 3 x 106And 1.2X 107Antibody-coupled magnetic beads and 6X 106Inoculation of human PBMC cells was performed according to magnetic bead: human PBMC cells ═ 1: 1. 1: 2 and 2: 1, inoculating the PBMC cells and the magnetic beads coupled with the antibody into a six-hole plate, uniformly mixing, and placing the six-hole plate at the temperature of 37 ℃ and 5% CO2The cell culture box of (2) is used for carrying out in-vitro amplification culture. Completely changing the culture medium after 48h, and supplementing or changing the culture medium every other day, wherein the cell culture factor is contained in each supplemented or changed culture medium, and the cell density is maintained at 1 × 106/mL。
4) Flow cytometry detection
Are respectively at the firstAt 0d and 7d, taking cells, performing cell phenotype detection by flow cytometry, and counting cell proliferation times, wherein the cell phenotype detects the classical NK cell phenotype (namely CD 3)-CD56+The cell population of (a).
The results of cell culture for 7 days and cell phenotype detection by flow cytometry are shown in FIG. 9, when the magnetic beads: human PBMC cells ═ 1:1 hour, can guarantee the stable activation effect of NK cell under the quantity of certain magnetic bead and antibody, consequently when carrying out the activation experiment of NK cell in vitro, the proportion of magnetic bead and human PBMC cell is 1: the comprehensive effect is best when 1.
Comparative example 3
Comparison of the trophoblast method for NK cell activation with the method described in example 2 (trophoblast-free method)
1) NK cell activation method (trophoblast method):
collecting peripheral blood of elbow vein of volunteer, separating and extracting human PBMC cell by density gradient centrifugation with Ficoll lymphocyte separation solution, suspending with LY-08 culture medium containing fetal bovine serum FBS, IL-2 and IL-15, counting cells, and adjusting cell density to 1 × 106and/mL. Wherein the working concentration of the added cell culture factor IL-2 is 1000IU/mL, the working concentration of IL-15 is 10ng/mL, and the working concentration of the fetal bovine serum FBS is 10%.
Following human PBMC cells: trophoblast K562/IL21[ human Chronic myelogenous leukemia cell (interleukin 21 gene modification) from national laboratory cell resource sharing platform, resource No. 3111C0001CCC000457]2: 1 ratio of human PBMC and feeder cells K562/IL21 were inoculated into a T25 cell culture flask (purchased from Corning Corp.), mixed well and then the T25 cell culture flask was placed at 37 ℃ with 5% CO2The cell culture box of (2) is used for carrying out in-vitro amplification culture. 7d was supplemented with feeder cells K562/IL21 in an amount of one half of the number of NK cells at that time. Absolutely static state is ensured in the first 48h of cell culture, complete liquid change is carried out after 48h, liquid supplement or liquid change treatment is carried out every other day, the cell culture factors are contained in each liquid supplement or liquid change culture medium, and the cell density is maintained at 1 × 106/mL。
2) NK cell activation method (non-feeder cell method):
the specific implementation procedure is the same as in example 2.
3) In vitro killing experiment of NK cells:
taking effector cells, namely NK cells cultured for 7 days by a feeder cell method and a non-feeder cell method, and adjusting the density of various effector cells to 4 × 106Perml and 2X 106Per mL; taking target cells, i.e. human myeloid leukemia cell K-562-luc2, and adjusting the target cell density to 2X 106mL, i.e. effective target ratio of 2: 1 and 1: 1; according to different effective target ratios, respectively adding a total of 200 mu L of effector cells and target cells with different concentrations into a round-bottom 96-well plate, and totally dividing into three groups:
target cell group: 100 μ L of target cells +100 μ L of LY-08 complete medium, 3 multiple wells per group;
experimental groups: 100 mu L of target cells and 100 mu L of NK cells cultured for 7 days by a non-feeder cell method, and each group is provided with 3 multiple wells;
control group: culturing 7-day NK cells by a target cell + feeder cell method in 100 mu L, and making 3 multiple holes in each group;
placing the three groups of mixed cells at 37 deg.C and 5% CO2The cell culture box of (1) was cultured for 4 hours, the remaining K-562-luc2 cells were detected using the One-Lit (TM) Luciferase Assay system kit, and the killing efficiency of NK cells was calculated according to the formula (the results are expressed as the mean of 3 replicate wells).
The killing efficiency calculation formula is as follows: killing efficiency ═ 100% for (1-experimental/target cell).
After killing for 4h, substituting into a formula to calculate the killing efficiency as shown in table 2 and fig. 10, wherein the effective target ratio is 2: at 1, the killing efficiency of the examples and the comparative examples is 77.04% and 76.60%, respectively; the effective target ratio is 1: at 1, the killing efficiencies of the examples and comparative examples were 46.95% and 37.95%, respectively. Therefore, the killing efficiency of NK cells activated and cultured by the non-feeder cell method of the invention is not obviously different from that of NK cells activated and cultured by the feeder cell method in the comparative example.
TABLE 2 killing effect of in vitro cell experiment
Figure BDA0003135035790000151
The foregoing is only a preferred embodiment of the present invention, and it should be noted that, for those skilled in the art, various modifications and decorations can be made without departing from the principle of the present invention, and these modifications and decorations should also be regarded as the protection scope of the present invention.
Sequence listing
<110> Beijing ancient cooking peptide source Biotechnology Ltd
<120> recombinant protein, recombinant expression vector, recombinant cell and NK cell activated magnetic bead, and preparation method and application thereof
<160> 4
<170> SIPOSequenceListing 1.0
<210> 1
<211> 60
<212> DNA
<213> Homo sapiens
<400> 1
atgtacagga tgcaactcct gtcttgcatt gcactaagtc ttgcacttgt cacgaattcg 60
<210> 2
<211> 1395
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 2
atggaggtgc agctggtgga gagcggaggc ggagtggtga ggcccggcgg atctctgagg 60
ctgagctgcg ctgcctccgg cttcaccttc gacgactacg gcatgagctg ggtgagacaa 120
gcccccggca agggactgga gtgggtgagc ggaatcaact ggaacggagg ctccaccggc 180
tacgctgaca gcgtgaaggg aagatttacc atctctagag acaacgccaa gaactctctg 240
tatctgcaga tgaacagcct cagagccgag gatacagccg tctactactg tgccagagga 300
aggtctctgc tgtttgacta ctggggccaa ggcacactgg tcacagtgag cagaggaggc 360
ggcggaagcg gaggaggagg atccggcggc ggaggctcca gcgagctcac acaagacccc 420
gccgtgagcg tggctctggg ccagaccgtg agaatcacat gccaaggcga ttctctgagg 480
agctactacg ccagctggta ccagcagaag cccggccaag cccccgtgct ggtcatctac 540
ggcaagaaca atagaccttc cggcattccc gacagattca gcggcagctc cagcggcaac 600
acagcctctc tgacaatcac cggcgcccaa gccgaagacg aggccgacta ctactgcaac 660
tctagagaca gctccggcaa ccacgtggtg tttggaggcg gcaccaagct caccgtgctg 720
agatctgtgg agtgcccacc ttgcccagca ccacctgtgg caggaccttc agtcttcctc 780
ttccccccaa aacccaagga caccctcatg atctcccgga cccctgaggt cacatgcgtg 840
gtggtggacg tgagccacga agaccctgag gtcaagttca actggtacgt ggacggcgtg 900
gaggtgcata atgccaagac aaagccgcgg gaggagcagt acaacagcac gtaccgtgtg 960
gtcagcgtcc tcaccgtcct gcaccaggac tggctgaatg gcaaggagta caagtgcaag 1020
gtctccaaca aaggcctccc atcctccatc gagaaaacca tctccaaagc caaagggcag 1080
ccccgagaac cacaggtgta caccctgccc ccatcccggg aggagatgac caagaaccag 1140
gtcagcctga cctgcctggt caaaggcttc tatcccagcg acatcgccgt ggagtgggag 1200
agcaatgggc agccggagaa caactacaag accacgcctc ccgtgctgga ctccgacggc 1260
tccttcttcc tctacagcaa gctcaccgtg gacaagagca ggtggcagca ggggaacgtc 1320
ttctcatgct ccgtgatgca tgaggctctg cacaaccact acacgcagaa gagcctctcc 1380
ctgtctccgg gtaaa 1395
<210> 3
<211> 87
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 3
tggtcccatc cgcaatttga gaaggggggc ggatctggag ggggcagtgg aggttccgct 60
tggagccacc cccagttcga aaagtga 87
<210> 4
<211> 1542
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 4
atgtacagga tgcaactcct gtcttgcatt gcactaagtc ttgcacttgt cacgaattcg 60
atggaggtgc agctggtgga gagcggaggc ggagtggtga ggcccggcgg atctctgagg 120
ctgagctgcg ctgcctccgg cttcaccttc gacgactacg gcatgagctg ggtgagacaa 180
gcccccggca agggactgga gtgggtgagc ggaatcaact ggaacggagg ctccaccggc 240
tacgctgaca gcgtgaaggg aagatttacc atctctagag acaacgccaa gaactctctg 300
tatctgcaga tgaacagcct cagagccgag gatacagccg tctactactg tgccagagga 360
aggtctctgc tgtttgacta ctggggccaa ggcacactgg tcacagtgag cagaggaggc 420
ggcggaagcg gaggaggagg atccggcggc ggaggctcca gcgagctcac acaagacccc 480
gccgtgagcg tggctctggg ccagaccgtg agaatcacat gccaaggcga ttctctgagg 540
agctactacg ccagctggta ccagcagaag cccggccaag cccccgtgct ggtcatctac 600
ggcaagaaca atagaccttc cggcattccc gacagattca gcggcagctc cagcggcaac 660
acagcctctc tgacaatcac cggcgcccaa gccgaagacg aggccgacta ctactgcaac 720
tctagagaca gctccggcaa ccacgtggtg tttggaggcg gcaccaagct caccgtgctg 780
agatctgtgg agtgcccacc ttgcccagca ccacctgtgg caggaccttc agtcttcctc 840
ttccccccaa aacccaagga caccctcatg atctcccgga cccctgaggt cacatgcgtg 900
gtggtggacg tgagccacga agaccctgag gtcaagttca actggtacgt ggacggcgtg 960
gaggtgcata atgccaagac aaagccgcgg gaggagcagt acaacagcac gtaccgtgtg 1020
gtcagcgtcc tcaccgtcct gcaccaggac tggctgaatg gcaaggagta caagtgcaag 1080
gtctccaaca aaggcctccc atcctccatc gagaaaacca tctccaaagc caaagggcag 1140
ccccgagaac cacaggtgta caccctgccc ccatcccggg aggagatgac caagaaccag 1200
gtcagcctga cctgcctggt caaaggcttc tatcccagcg acatcgccgt ggagtgggag 1260
agcaatgggc agccggagaa caactacaag accacgcctc ccgtgctgga ctccgacggc 1320
tccttcttcc tctacagcaa gctcaccgtg gacaagagca ggtggcagca ggggaacgtc 1380
ttctcatgct ccgtgatgca tgaggctctg cacaaccact acacgcagaa gagcctctcc 1440
ctgtctccgg gtaaatggtc ccatccgcaa tttgagaagg ggggcggatc tggagggggc 1500
agtggaggtt ccgcttggag ccacccccag ttcgaaaagt ga 1542

Claims (4)

1. A preparation method of NK cell activated magnetic beads is characterized by comprising the following steps: inducing the recombinant engineering cells to generate recombinant proteins, purifying and reacting with activated magnetic beads to obtain NK cell activated magnetic beads; the magnetic beads are Strep-Tactin mag.
The basic cells of the recombinant engineering cells comprise expiCHO cells, the genome of the recombinant engineering cells comprises a recombinant expression vector, and the construction method of the recombinant expression vector comprises the following steps:
(1) adding a coding gene sequence of Streptag2 between an Fc gene and a stop codon TGA of a eukaryotic expression vector pFUSE-hIgG1-Fc to obtain a vector pFUSE-hIgG1-Fc-Streptag 2;
(2) IL21-NM3E2The coding gene of IgG1 is inserted between the Nco I and BgII enzyme cutting sites of the vector pFUSE-hIgG1-Fc-Streptag2 to obtain the recombinant expression vector, and the IL21-NM3E2The encoding gene of the-IgG 1 is a nucleotide sequence shown in SEQ ID NO. 4.
2. NK cell-activated magnetic beads prepared by the method according to claim 1.
3. Use of the NK cell activating magnetic bead according to claim 2 for in vitro culturing NK cells.
4. A method for culturing NK cells in vitro, comprising the steps of: mixing human peripheral blood mononuclear cells and the NK cell activated magnetic beads of claim 2in a number ratio of 1:1, and culturing;
the culture medium for culture takes Human Lymphocytes Serum Free Media culture medium as a basic culture medium and also comprises: 1000IU/mL of cell culture factor IL-2, 10ng/mL of cell culture factor IL-15 and 10 percent of fetal bovine serum FBS.
CN202110715596.3A 2021-06-28 2021-06-28 Recombinant protein, recombinant expression vector, recombinant cell and NK cell activated magnetic bead as well as preparation method and application thereof Active CN113388042B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110715596.3A CN113388042B (en) 2021-06-28 2021-06-28 Recombinant protein, recombinant expression vector, recombinant cell and NK cell activated magnetic bead as well as preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110715596.3A CN113388042B (en) 2021-06-28 2021-06-28 Recombinant protein, recombinant expression vector, recombinant cell and NK cell activated magnetic bead as well as preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN113388042A CN113388042A (en) 2021-09-14
CN113388042B true CN113388042B (en) 2022-02-01

Family

ID=77624051

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110715596.3A Active CN113388042B (en) 2021-06-28 2021-06-28 Recombinant protein, recombinant expression vector, recombinant cell and NK cell activated magnetic bead as well as preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN113388042B (en)

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103756963A (en) * 2012-12-13 2014-04-30 上海柯莱逊生物技术有限公司 Method used for in vitro proliferation of NK cells
CN104357390A (en) * 2014-10-15 2015-02-18 深圳源正细胞医疗技术有限公司 Method for simultaneous and efficient amplification of CD<3+>CD<56+>CIK cells and CD<3->CD<56+>NK cells
CN105238754A (en) * 2015-11-20 2016-01-13 赵顺英 Method for in vitro culture of high-proliferation and high-mortality NK cells
CN107354133A (en) * 2017-08-31 2017-11-17 银丰生物工程集团有限公司 Magnetic bead is coupled method and the application of the amplification in vitro NK cells of a variety of stimulates the proteins
CN108250303A (en) * 2016-12-19 2018-07-06 南京金斯瑞生物科技有限公司 Single domain antibody fusion protein and its application
CN109666640A (en) * 2019-01-14 2019-04-23 武汉睿健医药科技有限公司 The method of the external pure culture of natural killer cells
CN111690606A (en) * 2020-05-14 2020-09-22 厚朴生物科技(苏州)有限公司 In-vitro activation and amplification of human natural killer cells and killing rate detection method
CN112608896A (en) * 2020-12-30 2021-04-06 广州瑞铂茵健康科技有限公司 NK cell culture method and application thereof
CN113293132A (en) * 2021-05-19 2021-08-24 江苏豪科生物工程有限公司 NK cell in-vitro amplification system and culture method
CN113493766A (en) * 2021-09-07 2021-10-12 依科赛生物科技(太仓)有限公司 NK cell in-vitro culture method for improving cell amplification multiple

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20220136455A (en) * 2014-04-23 2022-10-07 주노 쎄러퓨티크스 인코퍼레이티드 Methods for isolating, culturing, and genetically engineering immune cell populations for adoptive therapy
JP7337773B2 (en) * 2017-07-29 2023-09-04 ジュノー セラピューティクス インコーポレイテッド Reagents for expanding cells expressing recombinant receptors

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103756963A (en) * 2012-12-13 2014-04-30 上海柯莱逊生物技术有限公司 Method used for in vitro proliferation of NK cells
CN104357390A (en) * 2014-10-15 2015-02-18 深圳源正细胞医疗技术有限公司 Method for simultaneous and efficient amplification of CD<3+>CD<56+>CIK cells and CD<3->CD<56+>NK cells
CN105238754A (en) * 2015-11-20 2016-01-13 赵顺英 Method for in vitro culture of high-proliferation and high-mortality NK cells
CN108250303A (en) * 2016-12-19 2018-07-06 南京金斯瑞生物科技有限公司 Single domain antibody fusion protein and its application
CN107354133A (en) * 2017-08-31 2017-11-17 银丰生物工程集团有限公司 Magnetic bead is coupled method and the application of the amplification in vitro NK cells of a variety of stimulates the proteins
CN109666640A (en) * 2019-01-14 2019-04-23 武汉睿健医药科技有限公司 The method of the external pure culture of natural killer cells
CN111690606A (en) * 2020-05-14 2020-09-22 厚朴生物科技(苏州)有限公司 In-vitro activation and amplification of human natural killer cells and killing rate detection method
CN112608896A (en) * 2020-12-30 2021-04-06 广州瑞铂茵健康科技有限公司 NK cell culture method and application thereof
CN113293132A (en) * 2021-05-19 2021-08-24 江苏豪科生物工程有限公司 NK cell in-vitro amplification system and culture method
CN113493766A (en) * 2021-09-07 2021-10-12 依科赛生物科技(太仓)有限公司 NK cell in-vitro culture method for improving cell amplification multiple

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
A multi-Fc-species system for recombinant antibody production;Moutel 等;《BMC Biotechnology》;20090226;第9卷(第14期);第3页Figure 1图注 *
A new format of bispecific antibody: highly efficient heterodimerization, expression and tumor cell lysis;Xie 等;《Journal of Immunological Methods》;20041119;第296卷(第1-2期);第96页左栏、第98页右栏第1段 *
CD16单抗联合细胞因子体外扩增的NK细胞对K562杀伤功能研究;李雪莲 等;《生物化工》;20170825;第3卷(第4期);第30-36页 *
Direct immobilization of functional single-chain variable fragment antibodies (scFvs) onto a polystyrene plate by genetic fusion of a polystyrene-binding peptide (PS-tag);Kumada 等;《Anal Bioanal Chem》;20090813;第395卷;第760页左栏第2段 *
Isolation and characterization of an anti-CD16 single-chain Fv fragment and construction of an anti-HER2/neu/anti-CD16 bispecific scFv that triggers CD16-dependent tumor cytolysis;McCall 等;《Molecular Immunology》;19990531;第36卷(第7期);第433页摘要、第440页Table 2 *
一种融合抗体ScFv-Fc通用表达载体的构建;王丁丁 等;《中国生物工程杂志》;20110815;第31卷(第8期);第110-117页 *

Also Published As

Publication number Publication date
CN113388042A (en) 2021-09-14

Similar Documents

Publication Publication Date Title
CN110464841B (en) Immunity-enhancing pharmaceutical composition and application thereof
KR20170100660A (en) New artificial nucleic acid molecule
MXPA00007889A (en) Optimization of immunomodulatory properties of genetic vaccines.
CN113527510A (en) Fusion protein molecule and preparation method and application thereof
CN108264567A (en) A kind of Chimeric antigen receptor and cell for identifying CD19 positive tumors
CN111150841B (en) Active immune regulation particle and preparation method and application thereof
JP4870432B2 (en) Method for producing cytotoxic lymphocytes
CN111848819A (en) MSLN-targeted chimeric antigen receptor and application thereof
CN112625137B (en) Human interleukin 10-Fc fusion protein and medical application thereof
CN113061192B (en) PDL1 fusion protein with high affinity to PD-1 receptor and application thereof as T cell inhibitor
CN113388042B (en) Recombinant protein, recombinant expression vector, recombinant cell and NK cell activated magnetic bead as well as preparation method and application thereof
CN117797250A (en) Individuation mRNA composition, vector, mRNA vaccine and application thereof
EP1812459B1 (en) Method for the mass production of multimeric mannose binding lectin
CN107987172A (en) A kind of peptibody polyepitope vaccines for suppressing neonate tumour blood vessel and its application
CN110964118A (en) Bispecific fusion antibody and application thereof in tumor immunotherapy
CN103265637A (en) Recombinant porcine interleukin 4-Fc fusion protein as well as coding gene and expression method thereof
JP4741906B2 (en) Method for producing lymphocytes
CN110079539B (en) Preparation method of prostatic acid phosphatase/granulocyte-macrophage colony stimulating factor
CN103509100B (en) A kind of IL-1 R antagonist mutant
CN106432497B (en) Utilize the method for safflower suspension cell production CD20 antibody
AU2021461372A1 (en) Long-acting recombinant human interleukin-2 fusion protein, and preparation method therefor and application thereof
CN112646044B (en) TFF2-Fc fusion protein and high-efficiency expression production method thereof
CN114292337B (en) Soluble NK-CAR fusion protein, preparation method and application thereof in mediated immune cell targeted tumor cell killing medicament
CN112175905A (en) Method for efficiently knocking out CD96 gene in NK cell
CN111349615A (en) Method for preparing cell over expressing exogenous gene

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