CN117106814A - Soluble recombinant dengue 2 virus ED III protein and preparation method and application thereof - Google Patents

Soluble recombinant dengue 2 virus ED III protein and preparation method and application thereof Download PDF

Info

Publication number
CN117106814A
CN117106814A CN202311096010.5A CN202311096010A CN117106814A CN 117106814 A CN117106814 A CN 117106814A CN 202311096010 A CN202311096010 A CN 202311096010A CN 117106814 A CN117106814 A CN 117106814A
Authority
CN
China
Prior art keywords
protein
virus
ediii
dengue
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.)
Pending
Application number
CN202311096010.5A
Other languages
Chinese (zh)
Inventor
赵肃清
马梓婷
郭锦年
蒋璐
苏静怡
韩泽林
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.)
Guangdong University of Technology
Original Assignee
Guangdong University of Technology
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 Guangdong University of Technology filed Critical Guangdong University of Technology
Priority to CN202311096010.5A priority Critical patent/CN117106814A/en
Publication of CN117106814A publication Critical patent/CN117106814A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/70Vectors or expression systems specially adapted for E. coli
    • 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • C07K16/1081Togaviridae, e.g. flavivirus, rubella virus, hog cholera virus
    • 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
    • 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
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6854Immunoglobulins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/20Fusion polypeptide containing a tag with affinity for a non-protein ligand
    • C07K2319/21Fusion polypeptide containing a tag with affinity for a non-protein ligand containing a His-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
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/24011Flaviviridae
    • C12N2770/24111Flavivirus, e.g. yellow fever virus, dengue, JEV
    • C12N2770/24122New 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
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/24011Flaviviridae
    • C12N2770/24111Flavivirus, e.g. yellow fever virus, dengue, JEV
    • C12N2770/24134Use 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
    • C12RINDEXING SCHEME ASSOCIATED WITH SUBCLASSES C12C - C12Q, RELATING TO MICROORGANISMS
    • C12R2001/00Microorganisms ; Processes using microorganisms
    • C12R2001/01Bacteria or Actinomycetales ; using bacteria or Actinomycetales
    • C12R2001/185Escherichia
    • C12R2001/19Escherichia coli
    • 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
    • G01N2333/18Togaviridae; Flaviviridae
    • G01N2333/183Flaviviridae, e.g. pestivirus, mucosal disease virus, bovine viral diarrhoea virus, classical swine fever virus (hog cholera virus) or border disease virus
    • G01N2333/185Flaviviruses or Group B arboviruses, e.g. yellow fever virus, japanese encephalitis, tick-borne encephalitis, dengue
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2469/00Immunoassays for the detection of microorganisms
    • G01N2469/20Detection of antibodies in sample from host which are directed against antigens from microorganisms

Landscapes

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

Abstract

The application discloses a soluble recombinant dengue-2 virus EDIII protein and a preparation method and application thereof, and aims to provide the soluble recombinant dengue-2 virus EDIII protein which is efficiently expressed in a soluble form, so that fusion protein has complete biological activity and the advantages of strong specificity and high affinity, and the preparation method of the protein sequentially comprises the following steps: screening and detecting a dengue 2 virus EDIII protein virus high-specificity amino acid sequence and a corresponding coding gene fragment, wherein the amino acid sequence is shown as SEQ ID NO.1, and the coding gene fragment is shown as SEQ ID NO. 2; carrying out codon optimization on the high specificity encoding gene fragment of the dengue 2 virus EDIII protein, wherein the optimized sequence is shown as SEQ ID NO. 3; designing PCR amplification primers, and amplifying coding genes through PCR; constructing a target protein expression vector: inserting the EDIII protein coding target gene sequence into an expression vector cold shock expression vector to construct and obtain a target EDIII-pCold-TF protein expression vector; the EDIII-pCold-TF protein expression vector obtained in the step (a) is transformed into host cell escherichia coli, and the expression host cell is cultured to generate recombinant protein.

Description

Soluble recombinant dengue 2 virus ED III protein and preparation method and application thereof
Technical Field
The application relates to the technical field of genetic engineering, in particular to a soluble recombinant dengue 2 virus E protein domain III, and also relates to a preparation method and application of the soluble recombinant dengue 2 virus E protein domain III.
Background
Dengue is currently the most widely prevalent and most serious insect-borne infectious disease, an acute infectious disease caused by dengue virus (DENV) infection. Dengue virus belongs to the flaviviridae genus of flaviviridae family, and the main vectors and transmission vehicles are aedes albopictus and aedes aegypti, which are mainly divided into four serotypes of dengue 1-4 (DENV 1, DENV2, DENV3 and DENV 4), with type 2 transmission being the most widespread. Dengue is primarily prevalent in tropical and subtropical regions in early stages, and as global climate warms, the process of urbanization increases and international population frequencies increase, dengue spread and popularity further increases, and has expanded into non-tropical regions. Dengue has become a serious public health problem due to its rapid spread, high incidence, general susceptibility to the population and its high mortality. It places a heavy economic burden on both government and individuals. Dengue fever has been one of the infections that is being controlled with great importance in our country. Dengue disease progresses rapidly, from mild to severe within days, and the optimal window for diagnosis of dengue infection is usually from the onset of fever to 10 days after infection, so it is important to develop a rapid and accurate diagnostic tool for effective and rational disease management. The E protein domain III (EDIII protein) is proved to be a receptor recognition and binding region of dengue virus, has high immunogenicity and is considered to be an ideal serum diagnosis target.
From the data on serodiagnosis of dengue virus disclosed in the prior art, there are mainly the following problems: (1) Dengue viruses have 4 different serotypes, and reinfection with dengue viruses of different subtypes after a cross-protection period induces antibody-dependent viral infection-enhancing effects, and secondary atypical infections induce severe Dengue Hemorrhagic Fever (DHF) and Dengue Shock Syndrome (DSS) in patients. However, effective vaccines and specific medicines are still lacking in the market at present, and the research and development of the instant detection kit for dengue virus serotyping has important significance and market prospect. (2) EDIII proteins are demonstrated to be receptor recognition and binding regions of dengue virus, are highly immunogenic, and are considered ideal serum diagnostic targets. However, the recombinant protein prepared by the existing recombinant EDIII protein preparation method exists in an inactive insoluble inclusion body form, and the inclusion body is required to be dissolved and denatured for renaturation to obtain the EDIII protein with biological activity, so that the difficulty and cost for obtaining the high-activity EDIII recombinant protein are greatly increased, and the subsequent immunological diagnosis and differential diagnosis are very difficult.
Disclosure of Invention
In order to overcome the defects of the prior art, the first object of the application is to provide a soluble recombinant dengue 2 virus EDIII protein which is efficiently expressed in a soluble form, so that the fusion protein has complete biological activity and has the advantages of strong specificity and high affinity.
The second object of the application is to provide a preparation method of the EDIII protein of the soluble recombinant dengue-2 virus, which is simple and is simple to operate subsequently.
Another object of the present application is to provide an application of the soluble recombinant dengue 2 virus EDIII protein as a recombinant antigen protein for detecting dengue 2 virus antibodies.
The preparation method of the soluble recombinant dengue-2 virus EDIII protein sequentially comprises the following steps:
(1) Screening and detecting a high specificity coding gene fragment of dengue 2 virus EDIII protein virus, wherein the amino acid sequence is shown as SEQ ID NO.1, and the coding gene fragment is shown as SEQ ID NO. 2;
(2) Carrying out codon optimization on the high specificity encoding gene fragment of the dengue 2 virus EDIII protein virus, wherein the optimized sequence is shown as SEQ ID NO. 3;
(3) Designing PCR amplification primers, and amplifying coding genes through PCR;
(4) Constructing a target protein expression vector: inserting the EDIII protein coding target gene sequence into an expression vector cold shock expression vector to construct and obtain a target EDIII-pCold-TF protein expression vector;
(5) Transforming the EDIII-pCold-TF protein expression vector obtained in the step (4) into host cell escherichia coli, culturing the expression host cell to generate recombinant antigen protein, and obtaining the soluble recombinant antigen protein for detecting dengue 2 virus antibody through protein purification.
Further, the preparation method of the soluble recombinant dengue-2 EDIII protein comprises the following steps:
the upstream primer pCold-TF-F1:5' -CCACTTTCAACGAGCTGATG (SEQ ID NO. 4);
PCold-TF-F1:3’-GCGAAAGTGACTGAAAAAG(SEQ ID NO.5);
the downstream primer pCold-TF-R:5' -GGCAGGGATCTTAGATTCTG (SEQ ID NO. 6).
Furthermore, in the preparation method of the soluble recombinant dengue-2 virus EDIII protein, cloning sites are NdeI and XhoI.
Furthermore, in the preparation method of the soluble recombinant dengue 2 virus EDIII protein, the vector in the step (4) is a pCold-TF cold shock expression vector.
Furthermore, in the preparation method of the soluble recombinant dengue 2 virus EDIII protein, the carrier in the step (5) is BL21 or Rosetta series engineering bacteria as host cells.
Furthermore, according to the preparation method of the soluble recombinant dengue-2 virus EDIII protein, the target protein expression condition is that IPTG is used as an inducer to induce and express engineering bacteria, the final induction concentration is 0.01-1mM, the induction temperature is 15-20 ℃, and the induction and expression time is 18-24 hours.
Further, the preparation method of the soluble recombinant dengue-2 virus EDIII protein comprises the following steps:
(1) Transforming the constructed recombinant dengue 2 virus EDIII protein expression vector into a host system to obtain transformed host bacteria;
(2) Culturing the expression host bacteria under the expression condition, so as to express a fusion protein consisting of a target protein, a TF protein and a His tag;
(3) Under proper conditions, the fusion protein obtained in the step (2) is subjected to enzyme digestion by using FactorXa, and the target protein is released from the fusion protein into a solution;
(4) The purified recombinant protein is obtained by separation through Ni affinity chromatography, and the target protein is concentrated by ultrafiltration.
The second technical scheme provided by the application is that the soluble recombinant dengue-2 EDIII protein obtained by the preparation method provided by the first technical scheme.
The third technical scheme provided by the application is the application of the soluble recombinant dengue-2 virus EDIII protein as recombinant antigen protein for detecting dengue-2 virus antibodies.
Compared with the prior art:
the recombinant antigen protein provided by the application exists in a soluble form, and the subsequent operation of carrying out protein denaturation and renaturation is not needed, so that the defect that the inclusion body protein is required to be denatured by urea and then dialyzed and renatured and is complex in operation when the operation of preparing antibodies, vaccines and the like is needed in the follow-up operation because the EDIII protein exists in the inclusion body in the prior art is effectively solved. According to the technical scheme provided by the application, after the soluble recombinant dengue 2 virus EDIII antigen protein obtained through expression is separated through SDS-PAGE electrophoresis, western-blot experiments are carried out on the soluble recombinant dengue 2 virus EDIII antigen protein and the dengue 2 virus EDIII antibody, so that the recombinant antigen protein has extremely high specificity and high affinity, and can be used for real-time detection of dengue 2 viruses.
The soluble recombinant dengue-2 virus EDIII antigen protein provided by the application can be used for preparation of dengue-2 virus antibodies and development of vaccines.
In summary, the prepared recombinant antigen protein has better solubility and higher expression quantity, avoids the trouble of aftertreatment renaturation and unstable activity of inclusion body protein, has high and stable biological activity, and has good application prospect for large-scale production of high-activity recombinant protein.
Drawings
FIG. 1 is an electrophoretogram;
FIG. 2 shows the results of the sequencing by the selection of bacteria;
FIG. 3 shows the results of EDIII protein expression from different vectors;
FIG. 4 shows the results of EDIII protein expression induced by different inducer concentrations at 18h/24 h.
FIG. 5 is a graph of western blot identification of EDIII protein induced at 18h/24h of different inducer concentrations.
FIG. 6 is a western blot identification chart after EDIII protein purification.
Detailed Description
For a better understanding of the technical solution of the present application, the following examples are further described below, but the present application is not limited to the following examples.
In the following examples of the present application, the dengue virus protein selected was the domain III sequence of the E protein of dengue-2 virus strain (GenBank: JN 009092.1), and codons thereof were optimized and synthesized by biological engineering (Shanghai) Inc. (hereinafter referred to as "Ind.) Co. The pCold-TF vector used in the experiment was purchased from Wohan vast Ling Biotechnology Co. Mouse dengue type 2E protein antibodies were purchased from Prospecbio corporation. The high purity plasmid miniprep kit was purchased from daytime root Biochemical technology (Beijing) Co. The one-step method bacterial active protein extraction kit, the secondary antibody of the mouse anti-6 XHis monoclonal antibody and the goat anti-mouse marked by HRP enzyme, and the primary anti-diluent are purchased from the biological organism. Coli DH 5. Alpha., BL21 (DE 3) pLysS competent cells were purchased from Beijing full Biotechnology Co., ltd. SDS-PAGE gel rapid preparation kit was purchased from Biosharp.
The soluble recombinant dengue-2 virus EDIII protein provided by the application is prepared by the following method:
screening and detecting a high specificity encoding gene fragment of dengue 2 virus E protein virus, wherein in the embodiment, a dengue 2 virus strain discovered in Guangzhou in 2012 is selected. The amino acid sequence of the coding gene of the target gene fragment is shown as SEQ ID NO.1, and the coding gene fragment is shown as SEQ ID NO. 2. Codon optimization is carried out according to the information provided by National Center for Biotechnology Information, and the optimized sequence is shown in a table of SEQ ID NO. 3.
And (II) designing PCR amplification primers, and amplifying the coding genes through PCR. Primers used for sequencing:
the upstream primer pCold-TF-F1:5' -CCACTTTCAACGAGCTGATG (SEQ ID NO. 2)
pCold-TF-F1:3’-GCGAAAGTGACTGAAAAAG(SEQ ID NO.3)
The downstream primer pCold-TF-R:5' -GGCAGGGATCTTAGATTCTG (SEQ ID NO. 3)
The specific amplification method is as follows:
(1) On the bench, the reaction was added to the PCR tube and finally 2X Taq Plus Master Mix (Dye Plus) was added.
The PCR reaction system is as follows:
template 1. Mu.L
1. Mu.L of upstream primer
1. Mu.L of downstream primer
2×Taq Plus Master
(2) The bottom of the PCR tube is gently stirred by hand, and the mixture is centrifuged for a few seconds after being uniformly mixed and placed into a PCR instrument.
The reaction conditions were set as follows: 94 ℃ for 5min;94℃30s,58℃30s,72℃45s (30 cycles total); 72 ℃ for 10min;4 ℃ plus infinity.
(3) After completion of the PCR reaction, the whole PCR product was added to the well for 1% agarose gel electrophoresis to identify if the band position was correct and to isolate the product.
(4) And performing gel cutting recovery on the PCR product by using an agarose gel DNA recovery kit.
(5) The recovered product was measured for concentration and purity by Nanodrop micro-spectrophotometry and stored at-20 ℃.
Agarose gel electrophoresis basically operates as follows:
1) Washing and wiping the glue making tool with deionized water, sleeving the bottom plate into a die, and erecting a comb;
2) Agarose gel of appropriate concentration was formulated. Weighing a certain amount of agarose dry powder by using an electronic balance, adding the agarose dry powder into a clean conical flask, and adding a proper amount of freshly prepared 1 xTAE electrophoresis buffer;
3) Wrapping the bottle mouth with tinfoil, putting into a microwave oven, heating and melting, taking out, shaking and mixing uniformly, continuously heating until no particles exist, shaking slightly, and cooling to 40-50 ℃;
4) Rapidly adding nucleic acid dye Gold view into the melted agar solution, shaking and uniformly mixing, carefully pouring the solution into an electrophoresis tank, taking care that bubbles are not generated, and solidifying;
5) Solidifying for 30-40 min at room temperature, slightly shaking left and right to pull out the comb, taking out the solidified agarose gel and the bottom plate, observing whether the sample adding hole is damaged or not, placing the sample into a horizontal electrophoresis tank, moving the DNA sample from the negative electrode to the positive electrode, keeping one side of the sample adding hole close to the negative electrode, and fixing the sample adding hole in a good position;
6) Pouring freshly prepared 1 xTAE electrophoresis buffer into a horizontal electrophoresis tank; the gel is slightly higher than the gel surface by about 1mm, and bubbles in the gel sample adding holes are removed to prepare sample adding;
7) Adding a sample, namely adding 6×loading buffer into the DNA sample, blowing and uniformly mixing, and then centrifuging for a few seconds briefly, and adding the mixture into a gel sample adding hole; adding a DNA marker into one of the gel sample adding holes;
8) After the sample addition is finished, according to the red anode and the black cathode, covering the electrophoresis tank cover, switching on a power supply, and setting a constant voltage of 110V;
9) After about 40min, agarose gel electrophoresis is finished, the power supply is turned off, the agarose gel and the bottom plate are carefully taken out, the agarose gel and the bottom plate are placed in a dark box ultraviolet analyzer, the positions of the bands are observed, and the sizes of DNA markers are compared.
(III) constructing a target protein expression vector: the target protein expression vector is constructed and obtained by inserting the EDIII protein coding target gene sequence into an expression vector pCold-TF cold shock expression vector.
In this example, the synthesis of the genes of interest and the construction of the recombinant plasmids of steps (II) and (III) were completed by the organisms and confirmed by sequencing, wherein cloning sites were NdeI and XhoI. In FIG. 1, an electrophoretogram of the recombinant plasmid construction is completed, lane 1 is the original plasmid; lane 2 is restriction enzyme: ndeI/XhoI, EDIII; lane 3 is Marker.
(IV) transforming the EDIII-pCold-TF protein expression vector obtained in (3) into host cell escherichia coli, culturing the expression host cell to generate recombinant antibody protein, and obtaining the soluble recombinant antigen protein for detecting dengue 2 virus antibody through protein purification.
In the step (4), about 50ng of the recombinant plasmid was added to 100. Mu.L of E.coli DH 5. Alpha. Competent cells, after 30 minutes of ice bath, heat-shocked at 42℃for 90 seconds, transferred to ice and cooled for 2 minutes, then added to 900. Mu.L of a sterile LB liquid medium (preheated to 37 ℃) without antibiotics, mixed uniformly, placed in an incubator at 37℃and shake-cultured at 200rpm for 1 hour. After that, centrifugation was carried out at 1000rpm at 4℃for 10 minutes, the supernatant was discarded, and the suspension was resuspended in 50. Mu.L of a liquid medium, and the suspension was spread on a sodium Carbenicillin (CA) -resistant medium, and incubated at 37℃for 18 hours.
Positive clone screening: and (2) picking a plurality of single colonies into an LB culture medium with 5mLCA resistance by using a sterile gun head, vibrating at 37 ℃ and 200rpm for overnight, taking bacterial liquid for PCR identification, taking bacterial liquid with positive results, sending the bacterial liquid to a biological sequencing and determining, and comparing gene sequences by using Snap Gene software to extract plasmids from bacterial liquid with correct sequencing in a sequencing result diagram shown in FIG. 2.
The plasmid is transformed into competent cells of escherichia coli BL21 (DE 3) pLysS by the heat shock method, a plurality of single colonies are picked up to a LB culture medium with 5mLCA resistance by a sterile gun head, shaking is carried out at 37 ℃ and 200rpm for overnight, bacterial liquid is taken for PCR identification, and bacterial liquid with positive results is sent to the biological sequencing of the biology for determination. Single colonies of the correctly sequenced bacterial solutions were taken into 5 mLCA-resistant LB medium and shaken overnight at 37℃and 200 rpm. The following day, according to 1:100 ratio of the bacterial liquid was added to the LB medium with 50mLCA resistance, when the OD of the culture liquid was reached 600 When=0.4 to 0.5, the culture broth was immediately transferred to 15 ℃ and left for 30 minutes. Taking 1mL of uninduced bacterial liquid, adding IPTG to the final concentration of 1mM, respectively culturing for 18 hours and 24 hours at 15 ℃, taking induced bacterial liquid for SDS-PAGE electrophoresis analysis, configuring the concentration of separating gel to be 10%, the concentration of concentrated gel to be 4%, the constant pressure of the concentrated gel to be 70V, the constant pressure of the separating gel to be 120V, and after electrophoresis, coomassie brilliant blue staining to verify the expression condition of recombinant protein. The expression profile is shown in FIG. 3, and the results of the expression of the EDIII protein under the induction of different inducer concentrations at 18 hours and 24 hours are shown. Wherein lane 1 is Marker; lane 2 shows the expression without IPTG-induced bacteria; lane 3 shows expression after induction with 0.01mM IPTG; lane 4 shows expression after induction with 0.1mM IPTG; lane 5 is the expression after induction with 0.5mM IPTG; lane 6 is the expression after induction with 1mM IPTG.
After incubation at 15℃for 24 hours, the cells were harvested by centrifugation at 11,000rpm at 4℃for 5 minutes. The one-step method bacterial active protein extraction kit is utilized, 0.5mL of extraction reagent is added to each 100mg of wet weight thalli, and the mixture is blown and evenly mixed by a sterile gun head. Centrifugation was carried out at 13,000rpm for 5 minutes at 4℃to collect the supernatant and the precipitate, and the precipitate was suspended by vortexing with 0.25mL of the extraction reagent to obtain 20. Mu.L of the supernatant, which was analyzed by SDS-PAGE. SDS-PAGE electrophoresis, which is performed in the same manner as described above, is stained with Coomassie brilliant blue, and the expression of the recombinant protein is verified. The concentration of the separation gel was 15% in pET22 b-EDIII. As shown in FIG. 4, the results of the EDIII protein expression of the pCold-TF vector and the pET22b vector are shown. Lane 1 is Marker; lane 2 shows the expression of whole bacterial liquid; lane 3 is the soluble fraction; lane 4 is the inclusion body portion; arrows indicate the protein fusion expression portions. The experimental result shows that the constructed pCold-TF-EDIII recombinant protein is soluble protein (the theoretical molecular weight of the EDIII protein is about 10.5kDa, the molecular weight of the TF fusion protein is 52kDa, and the molecular weight of the obtained expression product is larger than that of the target protein due to fusion expression of protein tags, the target protein is detected to be about 70 kDa), and compared with the pET22b-EDIII constructed previously (the target protein is detected to be about 13 kDa), the recombinant protein has better solubility and better expression quantity. The literature reports that EDIII proteins are all expressed as inclusion bodies.
Western blot analysis was performed on pCold-TF-EDIII recombinant proteins.
The specific steps in this embodiment are: SDS-PAGE electrophoresis was performed in the same manner as described above, and after the completion of electrophoresis, the target protein was transferred to a nitrocellulose (PVDF) membrane. After blocking with blocking solution containing 5% nonfat milk powder for 2 hours at room temperature, the PVDF membrane was rinsed 3 times with TBST, and the PVDF membrane was placed in a primary anti-dilution solution to dilute mouse anti-6 XHis mab and incubated overnight at 4 ℃. The PVDF membrane was rinsed 3 times with TBST and incubated in goat anti-mouse secondary antibody diluted with HRP enzyme in blocking solution for 2 hours at room temperature. After the incubation was completed, the solution was washed 3 times with TBST, and after mixing the equal volumes of the developing solutions A and B, the solution was applied dropwise to a PVDF film for development. FIG. 5 is a Western blot identification chart showing the results of EDIII protein expression induced by different inducer concentrations at 18 hours and 24 hours. Wherein lane 1 is Marker; lane 2 shows the expression without IPTG-induced bacteria; lane 3 shows expression after induction with 0.01mM IPTG; lane 4 shows expression after induction with 0.1mM IPTG; lane 5 is the expression after induction with 0.5mM IPTG; lane 6 is the expression after induction with 1mM IPTG. FIG. 6 shows the EDIII protein obtained after the fusion protein is digested with Factor Xa and purified.
The analysis result proves that the reaction specificity of the expressed antigen can be well combined with the specific antibody, and can be used for the development of the subsequent antigen kit.

Claims (10)

1. The preparation method of the soluble recombinant dengue-2 virus EDIII protein is characterized by comprising the following steps in sequence:
(1) Screening and detecting a high specificity coding gene fragment of dengue 2 virus EDIII protein virus, wherein the amino acid sequence is shown as SEQ ID NO.1, and the coding gene fragment is shown as SEQ ID NO. 2;
(2) Carrying out codon optimization on the high specificity encoding gene fragment of the dengue 2 virus EDIII protein virus, wherein the optimized sequence is shown as SEQ ID NO. 3;
(3) Designing PCR amplification primers, and amplifying coding genes through PCR;
(4) Constructing a target protein expression vector: inserting the EDIII protein coding target gene sequence into the expression vector cold shock expression vector through a multiple cloning site on the expression vector, and constructing and obtaining a target EDIII-pCold-TF protein expression vector;
(5) Transforming the EDIII-pCold-TF protein expression vector obtained in the step (4) into host cell escherichia coli, culturing the expression host cell to generate recombinant protein, and obtaining the soluble recombinant antigen protein for detecting dengue 2 virus antibody through protein purification.
2. The method for preparing the soluble recombinant dengue-2 virus EDIII protein of claim 1 wherein the CR amplification primers are:
the upstream primer pCold-TF-F1:5' -CCACTTTCAACGAGCTGATG (SEQ ID NO. 4);
PCold-TF-F 1:3’-GCGAAAGTGACTGAAAAAG(SEQ ID NO.5);
the downstream primer pCold-TF-R:5' -GGCAGGGATCTTAGATTCTG (SEQ ID NO. 6).
3. The method for preparing the EDIII protein of the soluble recombinant dengue-2 virus according to claim 1, wherein the cloning sites are NdeI and XhoI.
4. The method for preparing the EDIII protein of the soluble recombinant dengue-2 virus according to claim 1, wherein the vector in the step (4) is a pCold-TF cold shock expression vector.
5. The method for preparing the soluble recombinant dengue-2 virus EDIII protein according to claim 1, wherein the carrier in the step (5) is a host cell which is a BL21 or Rosetta series engineering bacterium.
6. The method for preparing the EDIII protein of the soluble recombinant dengue-2 virus according to claim 1, wherein the target protein expression condition is that IPTG is used as an inducer to induce and express engineering bacteria, the final concentration of induction is 0.01-1mM, the induction temperature is 15-20 ℃, and the induction and expression time is 18-24 hours.
7. The method for preparing the soluble recombinant dengue-2 EDIII protein of claim 1, wherein the method for purifying the protein comprises the steps of:
(1) Transforming the constructed recombinant dengue 2 virus EDIII protein expression vector into a host system to obtain transformed host bacteria;
(2) Culturing the expression host bacteria under the expression condition, so as to express a fusion protein consisting of a target protein, a TF protein and a His tag;
(3) Under proper conditions, the fusion protein obtained in the step (2) is subjected to enzyme digestion by using FactorXa, and the target protein is released from the fusion protein into a solution;
(4) The purified recombinant protein is obtained by separation through Ni affinity chromatography, and the target protein is concentrated by ultrafiltration.
8. The soluble recombinant dengue-2 virus EDIII protein is characterized by being prepared by a preparation method of the soluble recombinant dengue-2 virus EDIII protein.
9. Application of a soluble recombinant dengue-2 virus EDIII protein as a test strip for preparing recombinant antigen protein for detecting dengue-2 virus antibody.
10. Use of the soluble recombinant dengue-2 virus EDIII protein of claim 1 for preparing a dengue-2 virus antibody or vaccine.
CN202311096010.5A 2023-08-28 2023-08-28 Soluble recombinant dengue 2 virus ED III protein and preparation method and application thereof Pending CN117106814A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202311096010.5A CN117106814A (en) 2023-08-28 2023-08-28 Soluble recombinant dengue 2 virus ED III protein and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202311096010.5A CN117106814A (en) 2023-08-28 2023-08-28 Soluble recombinant dengue 2 virus ED III protein and preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN117106814A true CN117106814A (en) 2023-11-24

Family

ID=88810689

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202311096010.5A Pending CN117106814A (en) 2023-08-28 2023-08-28 Soluble recombinant dengue 2 virus ED III protein and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN117106814A (en)

Similar Documents

Publication Publication Date Title
CN111499765B (en) Coronavirus fusion protein and preparation method and application thereof
CN110894217B (en) Bovine coronavirus chimeric antigen and colloidal gold immunochromatography test paper card for detecting bovine coronavirus antibody
CN111398581B (en) COVID-19 rapid diagnosis kit and preparation method thereof
CN106543272A (en) Multifunctional label fusion protein and its expression and application
CN114276445B (en) Rotavirus recombinant protein specific antibody, plasmid vector and method
CN107384933A (en) 9, pD1 PROTEIN Cs end amino acid polyclonal antibody, nucleotide sequence, preparation method and application
CN106520809A (en) Preparation method of GyV3 new ring virus VP3 fusion proteins
US20210395347A1 (en) Ns1-binding protein
CN104610443B (en) A kind of high stability restructuring Procalcitonin, Preparation method and use
CN111518174B (en) Optimized African swine fever CD2v protein and high-efficiency expression method and application thereof
CN111378017B (en) Subunit F protein of peste des petits ruminants virus and preparation method and application thereof
CN117106814A (en) Soluble recombinant dengue 2 virus ED III protein and preparation method and application thereof
CN106834324A (en) A kind of recombinant expression carrier that can promote solubility expression of protein and improve expression quantity
CN105949320A (en) Preparation and application of echovirus type 1 VP1 protein specific antigen epitope and fusion protein thereof
CN107266580B (en) Optimized target object capturing system based on bacterial cell surface display system
CN111471112B (en) Rabies virus recombinant antigen and preparation method and application thereof
CN110196325B (en) In-plug virus diagnosis kit and test paper
CN114002426A (en) Hog cholera virus E0 protein antibody ELISA detection kit
CN112301044A (en) Raw tobacco NbAPX3Gene polyclonal antibody and preparation method and application thereof
CN113307852A (en) Induced expression and purification method for 47kDa protein of orientia tsutsutsugamushi
CN112679607A (en) Preparation method of troponin I E13 single-chain antibody
CN111575315A (en) Rabbit viral hemorrhagic disease virus type II VLP vaccine
CN113122550A (en) Nucleic acid, gene and expression vector for coding bunyavirus endoviridae nucleoprotein and application thereof
JPH0242987A (en) Hybrid gene
CN113563484B (en) Fusion protein with G11-scFv-Nluc difunctional activity and application 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