CN114685676B - Recombinant protein, expression method, purification method and application thereof - Google Patents

Recombinant protein, expression method, purification method and application thereof Download PDF

Info

Publication number
CN114685676B
CN114685676B CN202011580950.8A CN202011580950A CN114685676B CN 114685676 B CN114685676 B CN 114685676B CN 202011580950 A CN202011580950 A CN 202011580950A CN 114685676 B CN114685676 B CN 114685676B
Authority
CN
China
Prior art keywords
rsv
recombinant protein
purifying
protein
liquid
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
CN202011580950.8A
Other languages
Chinese (zh)
Other versions
CN114685676A (en
Inventor
朱传凤
瓦晓霞
马超
李雄雄
傅生芳
王婉
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.)
Lanzhou Institute Of Biological Products Co ltd
Original Assignee
Lanzhou Institute Of Biological Products 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 Lanzhou Institute Of Biological Products Co ltd filed Critical Lanzhou Institute Of Biological Products Co ltd
Priority to CN202011580950.8A priority Critical patent/CN114685676B/en
Publication of CN114685676A publication Critical patent/CN114685676A/en
Application granted granted Critical
Publication of CN114685676B publication Critical patent/CN114685676B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • 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
    • A61P31/14Antivirals for RNA viruses
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56983Viruses
    • 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
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/036Fusion polypeptide containing a localisation/targetting motif targeting to the medium outside of the cell, e.g. type III secretion
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18511Pneumovirus, e.g. human respiratory syncytial virus
    • C12N2760/18522New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18511Pneumovirus, e.g. human respiratory syncytial virus
    • C12N2760/18534Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/005Assays involving biological materials from specific organisms or of a specific nature from viruses
    • G01N2333/08RNA viruses
    • G01N2333/115Paramyxoviridae, e.g. parainfluenza virus
    • G01N2333/135Respiratory syncytial virus

Abstract

The invention relates to a recombinant protein, an expression method, a purification method and application thereof. The recombinant protein is the F protein of RSV, namely RSV-F, the nucleotide sequence of the RSV-F is the sequence table SEQ ID No.1, and the amino acid sequence of the RSV-F protein is the sequence table SEQ ID No.2. The expression method of the recombinant protein comprises the following steps: 1) Amplification of RSV-F coding fragments; 2) Constructing subclone plasmids containing F protein according to the amplified RSV-F coding fragment; 3) Constructing eukaryotic expression plasmids of RSV-F according to the subclone plasmids; 4) Eukaryotic expression plasmids transfected with the RSV-F were cultured in HEK293E cells in medium. Purifying the recombinant protein by an anion exchange column, a cation exchange column and a gel filtration chromatographic column. The purified RSV-F recombinant protein has immunogenicity and can be used for the development of RSV subunit vaccine or the immunodiagnosis of RSV infection.

Description

Recombinant protein, expression method, purification method and application thereof
Technical Field
The invention belongs to the technical field of biology, and particularly relates to a recombinant protein, an expression method, a purification method and application thereof.
Background
Respiratory syncytial virus (respiratory syncytial virus, RSV) is the most prominent causative agent of acute lower respiratory illness in infants and young children. Children under 3400 and ten thousand years old, about 340 ten thousand hospitalizations, 16-20 ten thousand deaths, suffer RSV infection worldwide each year, and 99% of the deaths of RSV infection occur in low-and medium-income developing countries. Infants within 24 months are infected almost once with RSV, and all children under 3 years of age are infected with RSV and repeatedly infected multiple times throughout life. Worldwide, RSV causes mortality in infants under 1 year of age up to 6.7%, the second cause of mortality outside malaria. In addition, the elderly and immunocompromised patients are also susceptible to RSV infection, with the disease burden associated with RSV being comparable to that associated with influenza in non-epidemic seasons. Ribavirin is an antiviral drug used for the treatment of RSV disease, but its efficacy is uncertain, costly and difficult to administer, limited to severely symptomatic patients. Palivizumab is a monoclonal antibody for passively preventing RSV disease, which can effectively reduce the hospitalization rate caused by RSV, but is expensive and needs to be injected monthly, and is only currently applied to infants with high risk of RSV disease in developed countries. Severe RSV disease burden there is an urgent need to develop RSV vaccines, for which RSV vaccines are listed by the world health organization as one of the most developed vaccines worldwide.
The F protein is glycoprotein on the surface of respiratory syncytial virus, and belongs to type I glycoprotein. In the intracellular synthesis of inactive precursor F0, F0 is cleaved by furin during maturation to form F2 (aa 1-109), P27 (aa 110-136) and F1 (aa 137-574), F2 forms a heterodimer with F1 as disulfide, i.e., monomers of F protein, and the three monomers assemble to form a trimer. Functional F proteins on the respiratory syncytial virus envelope exist in an unstable pre-fusion conformation that folds to form a stable post-fusion conformation under the initiation of factors not yet identified. The F protein mediates fusion of the viral and host cell membranes, which is necessary for viral entry into the cell. The sequences of the F proteins of the two subtypes of RSV are highly conserved, and the homology is as high as more than 90%; monoclonal antibodies to protein F have been shown to reduce the incidence of severe RSV disease in infants at high risk, and have been approved for the prevention of RSV disease in infants, so protein F is the primary target antigen for developing vaccines.
The F protein is envelope protein, and can be obtained by culturing and splitting the virus in vitro, but the yield is lower; and is prone to losing its activity during purification of the F protein. To develop F protein vaccines, new methods of F protein preparation and purification must be established.
Disclosure of Invention
The invention provides a recombinant protein, an expression method, a purification method and application thereof.
A recombinant protein which is prepared from the recombinant protein,
the recombinant protein is F protein of RSV, namely RSV-F;
the nucleotide sequence of the RSV-F is a sequence table SEQ ID No.1.
Further, the method comprises the steps of,
the amino acid sequence of the RSV-F protein is SEQ ID No.2 of the sequence table.
Further, the method comprises the steps of,
a method of expressing a recombinant protein, the method comprising:
(1) Amplification of RSV-F coding fragments;
(2) Constructing subclone plasmids containing F protein according to the amplified RSV-F coding fragment;
(3) Constructing eukaryotic expression plasmids of RSV-F according to the subclone plasmids;
(4) Eukaryotic expression plasmids transfected with the RSV-F were cultured in HEK293E cells in medium.
Further, the method comprises the steps of,
the amplification of the RSV-F coding fragment takes an optimized RSV-F nucleotide sequence as a template, and the RSV-F fragment is amplified by PCR.
Further, the method comprises the steps of,
the RSV-F nucleotide sequence is nt94-nt1563 of the sequence table SEQ ID No.1.
Further, the method comprises the steps of,
the construction of the subclone plasmid containing the F protein is to connect the F protein with a pGEM-Teasy vector, thereby constructing a subclone plasmid pTe-F containing the F protein.
Further, the method comprises the steps of,
the eukaryotic expression plasmid of the RSV-F is constructed by respectively carrying out enzyme digestion and purification on the pTe-F and the expression vector pSEC and then connecting the pTe-F and the expression vector pSEC.
Further, the method comprises the steps of,
the eukaryotic expression plasmid of the RSV-F is transfected into HEK293E cells by a Lipofectamine2000 or PEI method;
the culture medium is a serum-free culture medium.
A method of purifying a recombinant protein, the method comprising:
(1) Amplification of RSV-F coding fragments;
(2) Constructing subclone plasmids containing F protein according to the amplified RSV-F coding fragment;
(3) Constructing eukaryotic expression plasmids of RSV-F according to the subclone plasmids;
(4) Transfecting eukaryotic expression plasmids of the RSV-F in HEK293E cells and culturing in a culture medium;
(5) Collecting supernatant RSV-F of the culture solution;
(6) Purifying the collected supernatant RSV-F by Capto Q anion exchange column;
(7) Purifying the RSV-F purified in the step (6) by a Sepharose sp hp cation exchange column;
(8) And (3) purifying the RSV-F purified in the step (7) by a Surperdex 200 gel filtration chromatographic column.
Further, the method comprises the steps of,
the anion exchange column is used for purifying, a liquid A equilibrium chromatographic column and a chromatographic system are used for loading concentrated supernatant containing target protein diluted by liquid A, and flow-through liquid is collected;
the A liquid is NaH 2 PO 4 ,Na 2 HPO 4 ,pH 7.0。
Further, the method comprises the steps of,
the anion exchange column is purified by using a B liquid equilibrium chromatographic column and a chromatographic system, and linear gradient elution is carried out by using a C liquid;
the solution B is a citric acid-sodium citrate buffer solution, and the pH value is 6.0; the solution C is NaCl.
Further, the method comprises the steps of,
the gel filtration chromatographic column is purified by using a D liquid balance chromatographic column and a chromatographic system, and then the D liquid is used for eluting to remove trace impurity proteins;
the solution D is Tris buffer solution, naCl, tween 20 and pH 7.4.
Further, the method comprises the steps of,
the purified RSV-F recombinant protein has immunogenicity and can be used for the development of RSV subunit vaccine or the immunodiagnosis of RSV infection.
The invention has the following advantages:
(1) The recombinant protein expressed RSV-F only comprises the extracellular region of the F protein, and does not comprise the transmembrane region and extracellular region of the F protein;
(2) The secretion signal peptide is adopted to replace the protein signal peptide, so that the expressed protein is soluble protein and is secreted into a culture medium, and the later protein purification is facilitated;
(3) A serum-free suspension culture system is adopted, so that the amplification of the culture scale and the protein purification are convenient;
(4) The expressed recombinant protein is purified in three steps to obtain high-purity RSV-F recombinant protein, has immunogenicity, and can be used for the research and development of RSV subunit vaccine or the immunodiagnosis of RSV infection;
(5) The RSV-F recombinant protein realizes secretion, solubility and high-level expression of the recombinant protein.
Additional features and advantages of the invention will be set forth in the description which follows, and in part will be obvious from the description, or may be learned by practice of the invention. The objectives and other advantages of the invention may be realized and attained by the structure particularly pointed out in the written description and claims hereof as well as the appended drawings.
Drawings
In order to more clearly illustrate the embodiments of the present invention or the technical solutions in the prior art, a brief description will be given below of the drawings that are needed in the embodiments or the prior art descriptions, and it is obvious that the drawings in the following description are some embodiments of the present invention.
FIG. 1 shows electrophoresis results of gene amplification products of RSV-F recombinant proteins according to an embodiment of the present invention;
FIG. 2 shows the results of the subcloning of the recombinant protein plasmid for RSV-F (EcoRI) according to an embodiment of the present invention;
FIG. 3 shows the results of the restriction enzyme assay (Age I/Kpn I) of eukaryotic expression plasmids for RSV-F recombinant proteins according to an embodiment of the invention: 1, pSEC;2, pSEC-F;
FIG. 4 shows SDS-PAGE (left) and Westernblot (right) identification of RSV-F recombinant proteins according to an embodiment of the invention: 1, pSEC-F;2, pSEC;
FIG. 5 shows an anion exchange chromatography of RSV-F recombinant protein according to an embodiment of the invention;
FIG. 6 shows a RSV-F recombinant protein cation exchange chromatography diagram according to an embodiment of the invention;
FIG. 7 shows a gel filtration chromatogram of RSV-F recombinant protein according to an embodiment of the invention;
FIG. 8 shows SDS-PAGE (left) and Westernblot (right) results of purified RSV-F recombinant proteins according to an embodiment of the invention;
FIG. 9 shows the induced anti-serum titers following immunization of mice with RSV-F recombinant proteins according to an embodiment of the invention.
Detailed Description
For the purpose of making the objects, technical solutions and advantages of the embodiments of the present invention more apparent, the technical solutions of the embodiments of the present invention will be clearly and completely described below with reference to the accompanying drawings in the embodiments of the present invention, and it is apparent that the described embodiments are some embodiments of the present invention, but not all embodiments of the present invention. All other embodiments, which can be made by those skilled in the art based on the embodiments of the invention without making any inventive effort, are intended to be within the scope of the invention.
1. Materials, reagents and instruments
The main materials are as follows:
and (3) cells: human embryonic kidney cells HEK293E
Main reagents, instruments:
EasApolymerase is available from Agilent corporation;
DNA markers, benchMark Prestained Protein Ladder and Bioluninescence Protein Ladder were purchased from Takara corporation;
EcoRI, age I, kpn I were purchased from NEB corporation;
QIAquick Gel Extraction kit, QIAprep Spin Miniprep Kit and Endo-free Plasmid Maxi Kit are purchased from QIAGEN;
amp, X-gal, IPTG were purchased from Beijing Ding national company;
polypepton is available from Japanese pharmaceutical Co;
yeast extract was purchased from OXOID corporation;
coli Top10 was purchased from Thermofiser, inc., saved by the applicant; na (Na) 2 HPO 4 .12H 2 O、NaH 2 PO 4 .2H 2 O, naCl, naOH, tween-20, etc. are available from national pharmaceutical group chemical company, inc.;
trizma@base, lumi-LightWestern BlottingSubstrate was purchased from Sigma corporation;
mouse monoclone [2F7] to RSV (ab 43812), goat Anti-Mouse IgG H & L (HRP) (ab 205719) are available from Abcam corporation;
FBS is purchased from excel corporation;
freeform F17 Expression Medium is available from Gibco corporation;
pen Strep, 0.05% Trypsin-EDTA, opti-MEM, DMEM from Gibco Lipofectamine2000 from Invitrogen;
PEI was purchased from Sigma company;
capto Q, sepharose sp hp, superdex 200 from GE company;
ultrafiltration concentrate columns were purchased from Millipore Ireland Ltd.
The materials, reagents and experimental equipment related to the embodiment of the invention are all commercial products conforming to the technical field of bioengineering unless otherwise specified.
EXAMPLE 1 expression method of RSV-F recombinant protein
1. Amplification of RSV-F-encoding fragments
The amplification of the RSV-F coding fragment takes the optimized RSV-F nucleotide sequence as a template, and the RSV-F fragment is amplified by PCR;
in particular, the method comprises the steps of,
the RSV-F nucleotide sequence is nt94 th to nt1563 th of a sequence table SEQ ID No. 1;
in particular, the method comprises the steps of,
taking an optimized sequence of a natural fusion protein gene coding sequence of a Long strain of wild RSV as a template, and designing the following primers according to the optimized sequence:
F-f GAAACCGGTCAGAATATCACCGAGGAGTTC(Age I)
F-r CTTGGTACCTCATTACAGCAGCTCATCGGACTTCC(KpnI)
amplifying the gene sequence of the extracellular region (AA 26-AA 512) of the RSV-F protein without signal peptide, namely the gene open reading frame nt94-nt1557;
in particular, the method comprises the steps of,
reaction conditions for PCR: pre-denaturation at 94 ℃ for 2min, denaturation at 94 ℃ for 30s, renaturation at 56 ℃ for 30s, extension at 72 ℃ for 1.5min,30 cycles, and extension at 72 ℃ for 10min;
the PCR reaction system is as follows:
reagent(s) Dosage (uL)
Stencil (pTe-F, 20 ng/ul) 1
dNTP(10mM) 4
EasyA polymerase 0.5
10xEasyA buffer 5
Forward primer (10 uM) 2
Reverse primer (10 uM) 2
Double distilled water 35.5
Total volume of 50
The result of electrophoresis of the amplified product is shown in FIG. 1, and FIG. 1 shows that the PCR amplified fragment has a length of 1488bp.
2. Construction of subclone plasmids containing F fragments
The construction of the subclone plasmid containing the F fragment is to connect the F fragment with a pGEM-Teasy vector so as to construct a subclone plasmid pTe-F containing the F fragment;
in particular, the method comprises the steps of,
the eukaryotic expression plasmid of the RSV-F is constructed by respectively carrying out enzyme digestion and purification on the pTe-F and an expression vector pSEC and then connecting;
after the PCR product is recovered and purified by gel, the PCR product is connected with pGEM-Teasy to construct a subclone vector pTe-F containing RSV gene fragments, and the EcoRI digestion result shows that the sum of the sizes of the two fragments is equivalent to the size of the insert fragment, as shown in figure 2. The sequence of the inserted fragment is 100% homologous with the target gene sequence.
3. Construction of eukaryotic expression plasmids for RSV-F
The eukaryotic expression plasmid of the RSV-F is constructed by respectively carrying out enzyme digestion and purification on the pTe-F and the expression vector pSEC, and the enzyme digestion identification result is shown in figure 3.
In particular, the method comprises the steps of,
the subclone plasmid of RSV-F and the empty vector pSEC of eukaryotic expression plasmid are digested by Age I/Kpn I, and after electrophoresis, the target fragment and vector arm are recovered and purified by gel, and then the eukaryotic expression plasmid of RSV-F (pSEC-F) is constructed by ligation and transformation. The sequence of the inserted fragment is 100% homologous with the target gene sequence. Eukaryotic expression plasmids were prepared in large quantities using Endo-free Plasmid Maxi Kit, see the Endo-free Plasmid Maxi Kit Specification (Endo-free Plasmid Maxi Kit from QIAGEN).
4. Transfecting eukaryotic expression plasmid of RSV-F in HEK293E cells, and culturing in a culture medium
The eukaryotic expression plasmid of the RSV-F is transfected into HEK293E cells by a Lipofectamine2000 or PEI method, namely, the transfection reagent adopted is Lipofectamine2000 or PEI;
in particular, the method comprises the steps of,
at a cell concentration of 1X 10 6 Transfecting eukaryotic expression plasmids of RSV-F in HEK293E cells under transfection conditions of cells/mL, cell sap, transfection reagent Lipofectamine2000 or PEI and RSV-F at 1.5mL-2mL/1 μg;
in particular, the method comprises the steps of,
the culture medium is a serum-free culture medium;
in particular, the method comprises the steps of,
reach 2X 10 at cell density 6 At the time of cell/ml, the ratio was 0.5X10 6 cells/ml were inoculated into F17 medium containing 2% glutamine and placed at 37℃in 5% CO 2 Culturing the cells in a shaker at 225 rpm;
in particular, the method comprises the steps of,
collecting culture supernatant on day 5 after transfection;
in particular, the method comprises the steps of,
concentrating by a concentration column for 4 times, concentrating the supernatant of the culture solution, and further centrifuging at 500xg for 10min at 4 ℃ to collect the culture solution.
Experiments show that: the expression plasmid was transfected into HEK293E cells using Lipofectamine2000, see Lipofectamine2000 instructions, followed by optimization of transfected cell density and recombinant protein harvest time:
mu.g pSEC-Fecto and 40. Mu.l of upper transfection reagent were mixed, 30ml HEK293E cells of different densities were transfected, and recombinant proteins were harvested at different times after transfection, results tableMing Dang cell density of 1X 10 6 At the time of cells/ml, the recombinant protein harvested on day 5 has higher protein expression amount.
Harvesting the recombinant protein of interest in the cell culture supernatant: collecting the cell culture solution in a centrifuge tube, centrifuging at 500xg for 10min at 4 ℃, and collecting the cell culture solution supernatant; adding into ultrafiltration concentration column for 4 times, and storing the concentrated solution in refrigerator at 4deg.C for purifying recombinant protein.
Further, the method comprises the steps of,
SDS-PAGE and Western blot analysis of the RSV-F recombinant protein, the identification results are shown in FIG. 4.
In particular, the method comprises the steps of,
taking 100 mu l of supernatant concentrate of the culture solution, respectively adding SDS-PAGE loading buffer solution, and boiling for SDS-PAGE and Western blot analysis;
in particular, the method comprises the steps of,
samples were spotted on two different gels, one of which was stained with SDS-PAGE followed by direct coomassie brilliant blue staining and destaining with destaining solution. The other block was subjected to Western blot analysis using 1: goat anti-RSV polyclonal antibody at 1000-fold dilution;
the secondary antibody is prepared by using HRP to mark goat anti-mouse IgG, adding the developing solution prepared by equal volumes of liquid A and liquid B, dropwise adding the developing solution onto a PVDF film for developing color, and placing the film in a gel imager for photographing.
Successful expression of the RSV-F recombinant protein was confirmed by SDS-PAGE and Western blot analysis, and the identification result is shown in FIG. 4.
EXAMPLE 2 purification method of RSV-F recombinant protein
The recombinant protein of RSV-F expressed in example 1 was purified by the following steps:
1. anion exchange purification: purification was performed using Capto Q anion exchange column, using 15 column volumes of binding buffer A (0.04M NaH) 2 PO 4 ,0.06M Na 2 HPO 4 pH 7.0) equilibrated column; then diluting the concentrated solution of the culture solution supernatant containing the target protein by using a buffer solution A, and adjusting the pH of the sample to be the same as the pH of the buffer solution; loading with loading pump, setting loading volume (500 ml), flow rate (3 ml/mi)n), pre-column pressure (0.3 MPa), column pressure difference (0.2 MPa) and the like, so that a sample flows through the column under a set condition, and flow-through liquid is collected for SDS-PAGE analysis. The chromatographic chart is shown in FIG. 5, and the electrophoretic chart is shown in FIG. 8.
2. Cation exchange purification: purifying with Sepharose sp hp cation exchange column, and balancing column with buffer B (citric acid-sodium citrate, pH 6.0) with flow rate of 10ml/min; concentrating the collected flow-through liquid, diluting with a buffer solution B, and regulating the pH of the sample; loading the sample by a loading pump, and setting parameters such as loading volume (200 ml), flow rate (2 ml/min), column front pressure (0.3 MPa), column pressure difference (0.2 MPa) and the like, so that the sample flows through the column under set conditions; after the sample loading is finished, carrying out linear gradient elution by using a buffer solution C (1M NaCl), wherein the ion concentration is 1% -100%, and eluting by using 10 times of column volume; the elution peaks were collected and subjected to SDS-PAGE and Western Blot analysis to obtain positive eluate fractions. The chromatographic chart is shown in FIG. 6, and the electrophoretic chart is shown in FIG. 8.
3. Gel filtration and purification: purifying with Superdex 200 (10/300) gel filtration chromatographic column, and balancing column with buffer D (Tris, naCl, tween 20, pH 7.4) to 1.5 times column volume at flow rate of 0.45ml/min; concentrating the components containing the target sample obtained by cation exchange, and taking 500ml for loading; eluting with buffer D (TBS pH 7.4) at a flow rate of 0.45ml/min for 1.5 times column volume, collecting each eluting peak, and performing SDS-PAGE and Western Blot analysis to obtain positive eluate. The chromatographic chart is shown in FIG. 7, and the electrophoretic chart is shown in FIG. 8.
EXAMPLE 3 analysis of purity of RSV-F recombinant protein
1. Protein content determination by BCA method
Standard protein BSA (2 mg/ml) was diluted with PBS to prepare standard series of 0, 25, 125, 250, 500, 750 and 1000. Mu.g/ml. 4ul of each series of standard substances and purified samples of recombinant proteins after proper dilution are taken and added into standard substance wells of a 96-well plate, 200ml of BCA working solution is added into each well, and the wells are placed at room temperature for 5min, and absorbance at 595nm is measured by an enzyme-labeled instrument. A standard curve (y= 1.2787 x-0.516) was drawn with the standard concentration and absorbance, where y is the protein concentration and x is the absorbance, and the sample concentration was calculated.
2. Protein purity analysis using a gray scale scanner
Purifying the expressed target protein, performing SDS-PAGE on the target protein obtained after three steps of purification, and analyzing the purity of the expressed protein by using a gray-scale scanner, wherein the purities of the three batches of expressed proteins are 92.2%, 96.7% and 98.7% respectively.
EXAMPLE 4 analysis of immunogenicity of RSV-F recombinant proteins
To confirm immunogenicity of the RSV-F recombinant protein, 36 BALB/C females of 6 weeks old were selected and divided into 6 groups of 6, 6 each, and the mice were vaccinated with TBS,1ug F (aluminum adjuvant), 5ug F (aluminum adjuvant), FI-RSV (1.78 mg) on days 0 and 28, respectively, and venous blood was collected in the tail of the mice on days 14, 28, 42 and 49, respectively, after vaccination. Antigen-specific IgG antibody titers in serum samples by ELISA according to standard procedures. Purified inactivated rsvp long was embedded in 96-well plates and incubated overnight at 4 ℃. Serum samples were diluted in blocking buffer at a gradient of 1:10 initial serum dilution, after which they were diluted in 2-fold series to different concentrations. There were also blank (serum dilution), negative control (1:100 diluted non-immune animal serum) and positive serum (1:6000 diluted murine anti-RSV serum), 100 μl of the above anti-serum dilution was added to the ELISA plate per well and incubated at 37℃for 1 hour. A horseradish peroxidase (HRP) -conjugated anti-mouse IgG (1:2000) secondary antibody was used. 3,3A, 5A-tetramethylbenzidine (TMB, BD Opt EIATM, BD Biosciences, ON) was used as a substrate for HRP. Mu.l of 1M H are added to each well 2 SO 4 The reaction was terminated. Absorbance at 450nm was measured using a microplate reader (Molecular Devices, USA) and the absorbance results are shown in figure 9. The results show that RSV-F has high immunogenicity, the immune response is dose dependent, and the aluminium adjuvant has the effect of enhancing the immune response.
Although the invention has been described in detail with reference to the foregoing embodiments, it will be understood by those of ordinary skill in the art that: the technical scheme described in the foregoing embodiments can be modified or some technical features thereof can be replaced by equivalents; such modifications and substitutions do not depart from the spirit and scope of the technical solutions of the embodiments of the present invention.
Sequence listing
<110> all Limited for Lanzhou biological research
<120> a recombinant protein, and expression method, purification method and use thereof
<160> 2
<170> SIPOSequenceListing 1.0
<210> 1
<211> 1563
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 1
atggggatcc ttcccagccc tgggatgcct gcgctgctct ccctcgtgag ccttctctcc 60
gtgctgctga tgggttgcgt agctgaaacc ggtcagaata tcaccgagga gttctaccag 120
tccacatgtt ctgccgtgag caagggctac ctgagcgccc tgaggacagg atggtatacc 180
tctgtgatca caatcgagct gagcaacatc aaggagaaca agtgcaatgg caccgacgcc 240
aaggtgaagc tgatcaacca ggagctggat aagtacaaga atgccgtgac agagctgcag 300
ctgctgatgc agtccaccac agccgccaac aatcgggccc ggagagagct gccacggttc 360
atgaactata ccctgaacaa taccaagaag acaaatgtga ccctgagcaa gaagaggaag 420
aggcgcttcc tgggctttct gctgggagtg ggatccgcca tcgcctctgg catcgccgtg 480
tccaaggtgc tgcacctgga gggcgaggtg aacaagatca agagcgccct gctgtccacc 540
aacaaggccg tggtgtctct gagcaatggc gtgtctgtgc tgacaagcaa ggtgctggac 600
ctgaagaatt atatcgataa gcagctgctg cccatcgtga acaagcagtc ctgtaggatc 660
tctaatatcg agaccgtgat cgagttccag cagaagaaca ataggctgct ggagatcaca 720
cgcgagtttt ctgtgaacgc cggcgtgacc acacctgtga gcacctacat gctgacaaat 780
agcgagctgc tgtccctgat caacgacatg ccaatcacca atgatcagaa gaagctgatg 840
tctaacaatg tgcagatcgt gcgccagcag tcctattcta tcatgagcat catcaaggag 900
gaggtgctgg cctacgtggt gcagctgcca ctgtatggcg tgatcgacac cccctgctgg 960
aagctgcaca catcccctct gtgcaccaca aacaccaagg agggctctaa tatctgcctg 1020
acccggacag acagaggctg gtactgtgat aacgccggca gcgtgtcctt ctttccccag 1080
gccgagacct gcaaggtgca gagcaaccgg gtgttctgtg acaccatgaa ttctctgaca 1140
ctgccaagcg aggtgaacct gtgcaatgtg gacatcttta atcccaagta tgattgtaag 1200
atcatgacat ctaagaccga cgtgagcagc agcgtgatca ccagcctggg cgccatcgtg 1260
tcctgctacg gcaagacaaa gtgtaccgcc tccaacaaga atagaggcat catcaagaca 1320
ttctccaacg gctgcgacta cgtgagcaac aagggcgtgg ataccgtgtc cgtgggcaac 1380
acactgtact atgtgaacaa gcaggagggc aagtctctgt acgtgaaggg cgagcccatc 1440
atcaacttct atgaccccct ggtgttccct agcgacgagt ttgatgcctc tatcagccag 1500
gtgaacgaga agatcaatca gagcctggcc tttatcagga agtccgatga gctgctgtaa 1560
tga 1563
<210> 1
<211> 519
<212> PRT
<213> Artificial sequence (Artificial Sequence)
<400> 1
Met Gly Ile Leu Pro Ser Pro Gly Met Pro Ala Leu Leu Ser Leu Val
1 5 10 15
Ser Leu Leu Ser Val Leu Leu Met Gly Cys Val Ala Glu Thr Gly Gln
20 25 30
Asn Ile Thr Glu Glu Phe Tyr Gln Ser Thr Cys Ser Ala Val Ser Lys
35 40 45
Gly Tyr Leu Ser Ala Leu Arg Thr Gly Trp Tyr Thr Ser Val Ile Thr
50 55 60
Ile Glu Leu Ser Asn Ile Lys Glu Asn Lys Cys Asn Gly Thr Asp Ala
65 70 75 80
Lys Val Lys Leu Ile Asn Gln Glu Leu Asp Lys Tyr Lys Asn Ala Val
85 90 95
Thr Glu Leu Gln Leu Leu Met Gln Ser Thr Thr Ala Ala Asn Asn Arg
100 105 110
Ala Arg Arg Glu Leu Pro Arg Phe Met Asn Tyr Thr Leu Asn Asn Thr
115 120 125
Lys Lys Thr Asn Val Thr Leu Ser Lys Lys Arg Lys Arg Arg Phe Leu
130 135 140
Gly Phe Leu Leu Gly Val Gly Ser Ala Ile Ala Ser Gly Ile Ala Val
145 150 155 160
Ser Lys Val Leu His Leu Glu Gly Glu Val Asn Lys Ile Lys Ser Ala
165 170 175
Leu Leu Ser Thr Asn Lys Ala Val Val Ser Leu Ser Asn Gly Val Ser
180 185 190
Val Leu Thr Ser Lys Val Leu Asp Leu Lys Asn Tyr Ile Asp Lys Gln
195 200 205
Leu Leu Pro Ile Val Asn Lys Gln Ser Cys Arg Ile Ser Asn Ile Glu
210 215 220
Thr Val Ile Glu Phe Gln Gln Lys Asn Asn Arg Leu Leu Glu Ile Thr
225 230 235 240
Arg Glu Phe Ser Val Asn Ala Gly Val Thr Thr Pro Val Ser Thr Tyr
245 250 255
Met Leu Thr Asn Ser Glu Leu Leu Ser Leu Ile Asn Asp Met Pro Ile
260 265 270
Thr Asn Asp Gln Lys Lys Leu Met Ser Asn Asn Val Gln Ile Val Arg
275 280 285
Gln Gln Ser Tyr Ser Ile Met Ser Ile Ile Lys Glu Glu Val Leu Ala
290 295 300
Tyr Val Val Gln Leu Pro Leu Tyr Gly Val Ile Asp Thr Pro Cys Trp
305 310 315 320
Lys Leu His Thr Ser Pro Leu Cys Thr Thr Asn Thr Lys Glu Gly Ser
325 330 335
Asn Ile Cys Leu Thr Arg Thr Asp Arg Gly Trp Tyr Cys Asp Asn Ala
340 345 350
Gly Ser Val Ser Phe Phe Pro Gln Ala Glu Thr Cys Lys Val Gln Ser
355 360 365
Asn Arg Val Phe Cys Asp Thr Met Asn Ser Leu Thr Leu Pro Ser Glu
370 375 380
Val Asn Leu Cys Asn Val Asp Ile Phe Asn Pro Lys Tyr Asp Cys Lys
385 390 395 400
Ile Met Thr Ser Lys Thr Asp Val Ser Ser Ser Val Ile Thr Ser Leu
405 410 415
Gly Ala Ile Val Ser Cys Tyr Gly Lys Thr Lys Cys Thr Ala Ser Asn
420 425 430
Lys Asn Arg Gly Ile Ile Lys Thr Phe Ser Asn Gly Cys Asp Tyr Val
435 440 445
Ser Asn Lys Gly Val Asp Thr Val Ser Val Gly Asn Thr Leu Tyr Tyr
450 455 460
Val Asn Lys Gln Glu Gly Lys Ser Leu Tyr Val Lys Gly Glu Pro Ile
465 470 475 480
Ile Asn Phe Tyr Asp Pro Leu Val Phe Pro Ser Asp Glu Phe Asp Ala
485 490 495
Ser Ile Ser Gln Val Asn Glu Lys Ile Asn Gln Ser Leu Ala Phe Ile
500 505 510
Arg Lys Ser Asp Glu Leu Leu
515

Claims (9)

1. A method for purifying recombinant protein is characterized in that,
the recombinant protein is RSV-F protein; the amino acid sequence of the RSV-F protein is the amino acid sequence of the 32 th to 519 th positions in a sequence table SEQ ID No. 2;
the method comprises the following steps:
(1) Amplification of RSV-F coding fragments;
(2) Constructing subclone plasmids containing F protein according to the amplified RSV-F coding fragment;
(3) Constructing eukaryotic expression plasmids of RSV-F according to the subclone plasmids;
(4) Transfecting eukaryotic expression plasmids of the RSV-F in HEK293E cells and culturing in a culture medium;
(5) Collecting supernatant RSV-F of the culture solution;
(6) Purifying the collected supernatant RSV-F by Capto Q anion exchange column;
(7) Purifying the RSV-F purified in the step (6) by a Sepharose sp hp cation exchange column;
(8) And (3) purifying the RSV-F purified in the step (7) by a Surperdex 200 gel filtration chromatographic column.
2. The method for purifying a recombinant protein according to claim 1, wherein,
the amplification of the RSV-F coding fragment takes an optimized RSV-F nucleotide sequence as a template, and the RSV-F fragment is amplified by PCR.
3. The method for purifying a recombinant protein according to claim 1, wherein,
the RSV-F nucleotide sequence is nt94-nt1563 of the sequence table SEQ ID No.1.
4. The method for purifying a recombinant protein according to claim 1, wherein,
the construction of the subclone plasmid containing the F protein is to connect the F protein with a pGEM-Teasy vector, thereby constructing a subclone plasmid pTe-F containing the F protein.
5. The method for purifying a recombinant protein according to claim 1, wherein,
the eukaryotic expression plasmid of the RSV-F is constructed by respectively carrying out enzyme digestion and purification on the pTe-F and the expression vector pSEC and then connecting the pTe-F and the expression vector pSEC.
6. The method for purifying a recombinant protein according to claim 1, wherein,
the eukaryotic expression plasmid of the RSV-F is transfected into HEK293E cells by a Lipofectamine2000 or PEI method;
the culture medium is a serum-free culture medium.
7. The method for purifying a recombinant protein according to claim 1, wherein,
the anion exchange column is used for purifying, a liquid A equilibrium chromatographic column and a chromatographic system are used for loading concentrated supernatant containing target protein diluted by liquid A, and flow-through liquid is collected;
the A liquid is NaH 2 PO 4 ,Na 2 HPO 4 ,pH 7.0。
8. The method for purifying a recombinant protein according to claim 1, wherein,
the cation exchange column is purified by using a B liquid equilibrium chromatographic column and a chromatographic system, and linear gradient elution is carried out by using a C liquid;
the solution B is a citric acid-sodium citrate buffer solution, and the pH value is 6.0; the solution C is NaCl.
9. The method for purifying a recombinant protein according to claim 1, wherein,
the gel filtration chromatographic column is purified by using a D liquid balance chromatographic column and a chromatographic system, and then the D liquid is used for eluting to remove trace impurity proteins;
the solution D is Tris buffer solution, naCl, tween 20 and pH 7.4.
CN202011580950.8A 2020-12-28 2020-12-28 Recombinant protein, expression method, purification method and application thereof Active CN114685676B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202011580950.8A CN114685676B (en) 2020-12-28 2020-12-28 Recombinant protein, expression method, purification method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202011580950.8A CN114685676B (en) 2020-12-28 2020-12-28 Recombinant protein, expression method, purification method and application thereof

Publications (2)

Publication Number Publication Date
CN114685676A CN114685676A (en) 2022-07-01
CN114685676B true CN114685676B (en) 2024-02-13

Family

ID=82130780

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202011580950.8A Active CN114685676B (en) 2020-12-28 2020-12-28 Recombinant protein, expression method, purification method and application thereof

Country Status (1)

Country Link
CN (1) CN114685676B (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113462656B (en) * 2021-03-24 2022-09-30 兰州生物制品研究所有限责任公司 Human three-type parainfluenza virus cold-adaptation temperature-sensitive strain and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102028943A (en) * 2010-12-08 2011-04-27 中国人民解放军军事医学科学院微生物流行病研究所 Respiratory syncytial virus-like particle vaccine and preparation method thereof
CN105214080A (en) * 2009-07-15 2016-01-06 诺华股份有限公司 RSV F protein compositions and its manufacture method
CN108738312A (en) * 2015-12-23 2018-11-02 辉瑞公司 Rsv f protein mutant

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009145606A1 (en) * 2008-05-28 2009-12-03 U-Protein Express B.V. Method for the production of proteins or protein fragments

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105214080A (en) * 2009-07-15 2016-01-06 诺华股份有限公司 RSV F protein compositions and its manufacture method
CN102028943A (en) * 2010-12-08 2011-04-27 中国人民解放军军事医学科学院微生物流行病研究所 Respiratory syncytial virus-like particle vaccine and preparation method thereof
CN108738312A (en) * 2015-12-23 2018-11-02 辉瑞公司 Rsv f protein mutant

Also Published As

Publication number Publication date
CN114685676A (en) 2022-07-01

Similar Documents

Publication Publication Date Title
Lasky et al. Neutralization of the AIDS retrovirus by antibodies to a recombinant envelope glycoprotein
Limonta-Fernández et al. An engineered SARS-CoV-2 receptor-binding domain produced in Pichia pastoris as a candidate vaccine antigen
CN107973850B (en) Recombinant classical swine fever virus E2 protein swine-derived monoclonal antibody, and preparation method and application thereof
CN114106164B (en) Monoclonal antibody for resisting novel coronavirus S protein and application thereof
CN110551187B (en) Chemically synthesized H7N9 avian influenza virus NA protein extracellular region antigen segment, preparation method and application
CN115838433A (en) Beta coronavirus polymer antigen, preparation method and application thereof
CN114685676B (en) Recombinant protein, expression method, purification method and application thereof
CN112390863A (en) Modified new coronavirus Spike protein extracellular domain and application thereof
KR102128453B1 (en) NS1 protein of yellow fever virus, monoclonal antibody specifically binding thereto, and uses thereof
WO2019100777A1 (en) Peptibody multi-epitope vaccine for inhibiting tumor angiogenesis, and application thereof
CN116747298B (en) Varicella-zoster virus vaccine and preparation method and application thereof
CN116836243A (en) F protein mutant before fusion of respiratory syncytial virus and application thereof
CN108383897A (en) Expression and its application of the VZV glycoprotein in Pichia pastoris
US8318662B2 (en) Protein increasing cell infectivity of herpes simplex virus and use thereof
EP4238983A1 (en) Vaccine composition for prevention or treatment of sars-coronavirus-2 infection
EP3545971A9 (en) Reconstituted rsv antigen
KR20210027606A (en) Pentamer-based recombinant protein vaccine platform and expressing system there of
Zhang et al. Identification of a linear epitope on the capsid protein of classical swine fever virus
CN108503696A (en) A kind of zika virus subunit vaccine of yeast cell to express
TW202216736A (en) Method for producing surface protein antigen of varicella zoster virus
US10766930B2 (en) Fusion proteins and use thereof for preparing vaccines
EA031404B1 (en) Vaccine for the prophylaxis and treatment of rotavirus infection with a fusion protein as active agent (variants)
CN105085638A (en) KSHV virus vIRF4 DNA binding domain and polyclonal antibody thereof, and preparation method of polyclonal antibody
WO2018175560A1 (en) Nanoparticle immunogens to elicit responses against the influenza receptor binding site on the hemagglutinin head domain
US20180360946A1 (en) Dengue virus glycoprotein e diii variants and uses thereof

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