CN113391067B - Indirect ELISA detection method for anti-Nipah virus G protein antibody - Google Patents

Indirect ELISA detection method for anti-Nipah virus G protein antibody Download PDF

Info

Publication number
CN113391067B
CN113391067B CN202110667511.9A CN202110667511A CN113391067B CN 113391067 B CN113391067 B CN 113391067B CN 202110667511 A CN202110667511 A CN 202110667511A CN 113391067 B CN113391067 B CN 113391067B
Authority
CN
China
Prior art keywords
protein
antibody
indirect elisa
serum
detection method
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
CN202110667511.9A
Other languages
Chinese (zh)
Other versions
CN113391067A (en
Inventor
金宁一
李昌
高子函
李乐天
许汪
郝鹏飞
鲁会军
李霄
田明尧
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.)
Military Veterinary Research Institute Academy Of Military Medical Sciences
Original Assignee
Military Veterinary Research Institute Academy Of Military Medical Sciences
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 Military Veterinary Research Institute Academy Of Military Medical Sciences filed Critical Military Veterinary Research Institute Academy Of Military Medical Sciences
Priority to CN202110667511.9A priority Critical patent/CN113391067B/en
Publication of CN113391067A publication Critical patent/CN113391067A/en
Application granted granted Critical
Publication of CN113391067B publication Critical patent/CN113391067B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • 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
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/531Production of immunochemical test materials
    • G01N33/532Production of labelled immunochemicals
    • G01N33/535Production of labelled immunochemicals with enzyme label or co-enzymes, co-factors, enzyme inhibitors or enzyme substrates
    • 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
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • 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
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Urology & Nephrology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biotechnology (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Microbiology (AREA)
  • Analytical Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Cell Biology (AREA)
  • Physics & Mathematics (AREA)
  • Virology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The invention relates to the technical field of biology, in particular to an indirect ELISA detection method for an anti-Nipag virus G protein antibody. The invention uses Nipah virus G protein to coat the ELISA plate, uses square matrix titration method to determine the optimum working condition of indirect ELISA, and analyzes the specificity, sensitivity, stability and the like of the method. The invention successfully establishes an indirect ELISA detection method for the mouse serum nipah virus G protein antibody, and can be applied to the analysis of the anti-nipah virus G protein antibody in the mouse serum and the evaluation and research of the specific antibody titer excited by the vaccine.

Description

Indirect ELISA detection method for anti-Nipah virus G protein antibody
Technical Field
The invention relates to the technical field of biology, in particular to an indirect ELISA detection method for an anti-Nipag virus G protein antibody.
Background
Nipah virus disease, which is a newly emerging virulent zoonotic infectious disease, can cause severe and high mortality (40% -70%) in humans, is a Nipah virus (NiV),is a single-stranded negative strand RNA virus belonging to Paramyxoviridae (Paramyxoviridae) Huntiepaviral (Henipavirus) [1] NiV can be transmitted by direct contact or in the form of an aerosol, resulting in infection and even death of humans or animals. The virus was first discovered after the outbreak of encephalitis in swine farmers in malaysia and singapore, and then almost every year in bangladesh or india. The virus was named because it was isolated in the Nipath region. NiV currently has mainly 2 genetic lineages: niV Malaysia strain (NiV Malaysia, niV-MY) and NiV Bengal strain (NiV Bangladesh, niV-BD). Wherein the genome of the NiV-MY strain is 18246 nucleotides, and the genome of the NiV-BD strain is 18252 nucleotides, the RNA genome of the NiV-MY strain is formed by continuously arranging 6 genes of nucleocapsid protein (N), phosphoprotein (P), matrix protein (M), fusion glycoprotein (F), attachment glycoprotein (G) and long polymerase (L), and the genes are N, P and L attached to viral RNA to form viral ribonucleoprotein (vRNP). M, F, G constitutes the envelope of the virion, where M is involved in maintaining the virus morphology and mediating viral budding, while F, G is involved in mediating viral attachment and invasion of host cells.
NiV outbreaks with mortality rates exceeding 70% -100% have occurred in Bengala and India almost annually since 2001, whereas the last outbreak was in Kerikende (Kozhikode) in India, resulting in 17 deaths in 19 people infected, with mortality rates as high as 89%, and our country bordered by India and adjacent to Bengala, which, although no case has been reported, has a risk of afferent China. Because of the high pathogenicity, potential pandemic, and the currently unapproved vaccine and mature therapeutic system of this virus, there is an urgent need to study and develop antiviral drugs and vaccines and effective detection methods to help prevent and control future epidemic situations that may be outbreak.
Disclosure of Invention
In view of this, the present invention provides an indirect ELISA detection method for mouse anti-Nipah virus (NiV) G protein antibodies. The invention establishes an indirect ELISA detection method for the mouse serum Nipag virus G protein antibody, and lays a solid foundation for the subsequent development of vaccines and the detection and evaluation of specific antibodies.
In order to achieve the above object, the present invention provides the following technical solutions:
the invention provides an indirect ELISA detection method for anti-Nipag virus G protein antibody, which comprises the steps of taking Nipag virus G protein as antigen to coat an ELISA plate, beating liquid, washing, sealing, washing, adding diluted primary antibody for incubation, beating primary antibody for washing, adding ELISA secondary antibody for incubation, beating secondary antibody for incubation, washing, developing color for incubation, and stopping reaction to obtain OD 450 A value;
the primary antibody comprises positive sample serum or negative sample serum;
the coating concentration of the antigen is 2-10 mug/mL; the dilution of the primary antibody was 1: 200-1: 800; the incubation time of the primary antibody is 30-90 min.
In some embodiments of the invention, the antigen is coated at a concentration of 10 μg/mL; the dilution of the primary antibody was 1:400, the incubation time of the primary antibody was 60min.
In some specific embodiments of the invention, the working concentration of the enzyme-labeled secondary antibody is 1:5000-1:20000; the incubation time of the enzyme-labeled secondary antibody is 30-120 min.
In some embodiments of the invention, the dilution of the second enzyme-labeled antibody is 1:20000, and the incubation time of the second enzyme-labeled antibody is 30min.
In some embodiments of the invention, the closure is specifically: 5% skim milk-TBST was used as a blocking solution, and the mixture was blocked at 37℃for 1.5 hours.
In some embodiments of the invention, the time for the color development is from 5 minutes to 20 minutes.
In some embodiments of the invention, the time for development is 15 minutes.
In some embodiments of the invention, the Cut-off value is the OD of a negative control serum 450 Mean x 2.1.
Based on the research, the invention also provides a method for determining the Nipagin virus candidate vaccine, which takes the candidate vaccine as a primary antibody and detects according to the indirect ELISA detection method.
The invention also provides a method for determining the anti-Nipag virus G protein antibody in the serum to be detected, wherein the serum to be detected is used as a primary antibody, and the detection is carried out according to the indirect ELISA detection method.
The invention uses Nipah virus G protein to coat the ELISA plate, uses square matrix titration method to determine the optimum working condition of indirect ELISA, and analyzes the specificity, sensitivity, stability and the like of the method. Results the optimal dilution of the ELISA method samples was 1:400, the optimal incubation time is 60min; HRP-labeled goat anti-mouse IgG optimal dilution is 1:20000, and optimal incubation time is 30min; the optimal coating concentration of the antigen is 10 mug/mL; the optimal color development time is 15min, and the negative control serum OD is used 450 Mean X2.1 (positive control serum greater than 4.00 calculated as 4.00) is the Cut-off value. The Coefficient of Variation (CV) of 3 replicates in the sample experiment is less than 10%, and the CV of 3 replicates in the sample experiment is less than 10%. Conclusion the indirect ELISA detection method of the mouse serum Nipag virus G protein antibody is successfully established, and the method can be applied to evaluation and research of specific antibody titers excited by vaccines and used for analysis of anti-Nipag virus G protein antibodies in mouse serum.
Drawings
In order to more clearly illustrate the embodiments of the present invention or the technical solutions in the prior art, the drawings used in the description of the embodiments or the prior art will be briefly described below.
FIG. 1 shows the results of negative serum indirect ELISA assays.
Detailed Description
The invention discloses an indirect ELISA detection method for anti-Nipagin G protein antibodies, and a person skilled in the art can properly improve the process parameters by referring to the content of the invention. It is expressly noted that all such similar substitutions and modifications will be apparent to those skilled in the art, and are deemed to be included in the present invention. While the methods and applications of this invention have been described in terms of preferred embodiments, it will be apparent to those skilled in the relevant art that variations and modifications can be made in the methods and applications described herein, and in the practice and application of the techniques of this invention, without departing from the spirit or scope of the invention.
Nipah virus as a pathogen with a broad host spectrum [7] Its host comprises common animals such as fruit bat, pig, cow, horse, goat, cat, dog, mouse, rabbit, fox, glancing bird, and mexico [8] The range of movement of the bats is wider, and NiV is likely to spread among the bats of fruit eating in a certain area, so that countries and regions with the same type of bats are exposed to the risks of virus mutation and epidemic outbreak. As a big pig raising country, once epidemic situation outbreaks occur, the method can cause great threat to the life and property safety of people in China. Although NiV is not detected in China at present, the southeast coastal area and the Yunnan area of China have ecological environment and host animals suitable for NiV epidemic, and a natural host of NiV is found, nipah virus antibodies exist in bat bodies, which suggests that China possibly faces epidemic situation outbreak epidemic threat, and therefore related researches on NiV detection means and vaccines are necessary to be carried out. Indirect ELISA developed using viral antigens for serological detection (IgM and IgG) has been used for serological antibody detection in humans and field studies in bats and other animals. However, since the virus is highly dangerous, it is difficult to detect the virus using whole virus as an antigen because the virus cannot be cultured under ordinary laboratory conditions. Eshaghi et al [10] The expression of nipah virus N protein using baculovirus expression system was established and found to be useful in place of whole virus as antigen for IgG antibody capture ELISA. Kaku et al established sandwich ELISA for antigen capture by using polyclonal antibody, constructed two vector plasmids (F protein encoding plasmid and G protein encoding plasmid) immune rabbit to obtain polyclonal antibody IgG as antigen capture antibody, respectively adding horseradish peroxidase as enzyme-labeled conjugate antibody on the two IgG, respectively comparing reactivity by ELISA, finally obtaining the polyclonal antibody of G protein with optimal reactivity, respectively testing pseudotyped VSV, niV of two different strains and HeV live virus to obtain lower detection limit, and using common VSV as negative control, and the result shows that the detection sensitivity of pseudotyped VSV and NiV is acceptable, and the detection lower limit of HeV is higher and not sensitive, the method can only be used for detecting henpa virus, and can not distinguish NiV and HeV, but the preparation of polyclonal antibody is simplerThe ELISA can be used as a standby detection method in view of the problems of genetic variation of henipa virus and reliability of various existing PCR detection.
At present, no commercial nipah virus G protein antibody detection kit is used for evaluating nipah virus candidate vaccines, so a worker tries to establish an indirect ELISA method for detecting nipah virus G protein IgG, and ELISA is very suitable for being used as an epidemiological investigation and serological screening tool due to the characteristics of sensitivity, rapidness, safety and high flux. The invention takes eukaryotic expression of Nipah virus G protein as a coating antigen, and finally successfully establishes an indirect ELISA method with high specificity, sensitivity and stability through fumbling on various conditions, thereby being applicable to epidemiological investigation and evaluation of candidate vaccines.
In the indirect ELISA detection method of the anti-Nipagin G protein antibody provided by the invention, raw materials and reagents used can be purchased from the market.
The invention is further illustrated by the following examples:
example 1 basic procedure of an indirect ELISA method
The preparation method of the Nipag G protein comprises the following steps:
nipah virus G protein sequence analysis, design and synthesis
The gene sequences of the G proteins were compared according to the 28 whole genome sequences of nipah virus on NCBI, the evolutionary tree was drawn, and the amino acid homology of the G proteins was analyzed. Analysis was performed by on-line hydrophobicity analysis means, epitope prediction means (http:// www.detaibio.com/tools /) to obtain the G protein sequence of the nipah virus representative strain (NCBI accession number: AF 212302.2), deleting cytoplasmic tail, transmembrane hydrophobic and part of the stem region, leaving only the extracellular globular head region (designated sG). The N end is added with a human tissue plasminogen activator signal peptide (tPA) sequence and Strep tag, and the two ends are respectively added with enzyme cutting sites HindIII and BamHI, after the sequence design is completed, the sequence is subjected to codon optimization and synthesis by Shanghai Biotechnology, thereby obtaining the glycerinum containing target gene plasmid (pUC 57-NiV-sG).
Construction of recombinant plasmids
The synthesized 10 mu L glycerol bacteria containing the target gene plasmid (pUC 57-NiV-sG) are inoculated in 5mL liquid LB culture medium with ampicillin resistance, shake culture is carried out for 12h at 37 ℃ to carry out small extraction preparation of plasmids, hindIII and Bam HI are connected to eukaryotic expression vector pcDNA3.1 (+) after double enzyme digestion to obtain eukaryotic expression plasmid pcDNA-sG, the recombinant plasmid is transformed into competent cells Trans1-T1 by a heat shock method, positive colonies are selected through ampicillin resistance screening, plasmids are extracted, double enzyme digestion identification is carried out, the plasmids with correct identification are sent to Shanghai bioengineering limited company for sequencing, and after sequence comparison confirmation, a large amount of plasmids are prepared and purified to determine plasmid concentration for cell transfection.
Cell transfection
The day before transfection, the Expi293F cells were cultured in suspension at 8% CO2 at 37℃under 125r/min, 60mL of a system of 293F cell cultures were assayed for viability and viable cell density, viable cells were grown to 4.2X106/mL with a viability of 97%, the cell cultures were diluted to 2.1X106/mL, the day of transfection with a cell density of 3X 106/mL, 60. Mu.g eukaryotic expression plasmid pcDNA-sG was isolated from 3mL Opti-MEM TM I Reduced Serum Medium and 160. Mu.L of Expiectamine TM 293 Reagent and 2.8mL Opti-MEM TM I Reduced Serum Medium, incubating at room temperature for 5min, mixing the two, and incubating at room temperature for 20min. The solution was then slowly transferred to a cell culture shake flask, which was gently rotated during addition.
Collecting the protein of interest
After 12h of transfection, 300. Mu.L of transfection enhancer 1 and 3mL of transfection enhancer 2 were added, culture was continued for 96h, the culture was centrifuged at 800g for 10min, the culture supernatant and cell pellet were collected, respectively, the culture supernatant was designated sG-MS, the cell pellet was resuspended in IP lysate and lysed on ice for 10min, disrupted by an ultrasonic cell disruptor for 10min, and the supernatant was collected by centrifugation and designated sG-DS.
The antigen (Nipag G protein) prepared and stored in molecular virology and immunology laboratory of military veterinary institute of medical science was prepared by ELISCoating an ELISA plate after diluting the protein A coating liquid, and coating the ELISA plate at 4 ℃ overnight (16-24 h); the liquid is beaten off, and TBST is added for washing twice for 2min each time; adding 5% skimmed milk-TBST, sealing at 37deg.C for 1.5 hr; cleaning the sealing liquid, adding TBST for washing for 3 times, and 2min each time; after diluting sample serum (primary antibody) with PBS, incubating the ELISA plate at 37 ℃ for 60min; the primary antibody was then removed by patting, washing with TBST three times for 5min each, adding TBST diluted goat anti-mouse IgG-HRP (secondary antibody), and incubating at 37deg.C for 60min; the secondary antibody is beaten, washed three times by TBST for 10min each time, 100 mu LTMB single-component color development liquid is added into each hole, and incubated for 15min at 37 ℃; the reaction was stopped by adding 50. Mu. LELISA stop solution to each well. Determination of OD Using an enzyme-labeled Instrument 450 Values. The above is the initial basic flow of the method, and then the optimal conditions are determined through fumbling of various conditions.
Example 2 determination of optimal coating concentration of antigen and optimal dilution of serum
The optimal coating concentration of antigen and the optimal dilution of serum are determined by a square titration method, nipag G protein is respectively diluted into 2 mug/mL, 4 mug/mL, 8 mug/mL and 10 mug/mL, 50 mug/well is used for coating an ELISA plate, and the ELISA plate is incubated overnight at 4 ℃. Positive sample serum and negative sample serum were diluted to 1: 200. matrix titration was performed with 1:400, 1:800 and the protein coating concentrations described above, and three parallel experiments were performed. The optimal dilution of serum and the optimal coating concentration are obtained by using the serum dilution times and the antigen coating concentration when the P/N value (positive control serum A450 mean/negative control serum A450 mean) is more than 2.1 and the maximum.
When the antigen coating concentration was 10. Mu.g/mL, the serum dilution was 1: positive control serum OD at 400 450 And the combination of the P/N values satisfies the set condition, and the P/N value is highest, see Table 1. Thus, the optimal coating concentration for antigen was determined to be 10. Mu.g/ml and the serum dilution was 1:400.
TABLE 1 results of square titration of optimal coating concentration of antigen and optimal dilution of serum (OD 450 )
Tab 1.Block titration results at various antigen concentrations for coating and serum dilutions(OD 450 )
Example 3 determination of optimal working concentration and reaction time for enzyme-labeled secondary antibody
The method comprises the steps of performing three groups of parallel experiments by respectively diluting goat anti-mouse IgG-HRP to 1:5000, 1:10000 and 1:20000 with the optimal antigen coating concentration and the optimal serum dilution determined in the previous step, incubating an ELISA plate at 37 ℃ for 30min, 60min, 90min and 120min, and displaying the reading of the ELISA plate as OVER and OD 450 The value is calculated to be 4.000, and the optimal working concentration and the optimal reaction time of the enzyme-labeled secondary antibody are taken when the P/N value is larger than 2.1 and the enzyme-labeled secondary antibody is maximum.
Goat anti-mouse IgG-HRP (enzyme-labeled secondary antibody) dilution is 1:20000, reaction time is 30min, and the result is the most suitable for the set condition, as shown in Table 2. Therefore, the optimal working concentration of the enzyme-labeled secondary antibody is 1:20000, and the optimal reaction time is 30min.
TABLE 2 results of square titration of different enzyme-labeled antibody concentrations and reaction times (OD 450 )
Tab 2.Block titration results of various HRP-labeled antibody concentrations and reaction time(OD 450 )
Example 4 determination of closed conditions
Three parallel experiments were performed using 5% skim milk-TBST and 3% BSA-TBST, respectively, at 37℃for 1h, 1.5h, and 2h, respectively, with P/N values greater than 2.1 and at maximum as optimal blocking conditions.
The two different sealing liquids were used for sealing, and 3 times were set, and as can be seen from Table 3, when 5% skim milk-TBST was used as the sealing liquid for sealing at 37℃for 1.5 hours, the P/N value was the largest, indicating that the set conditions were most satisfied. Therefore, the optimal blocking condition is 5% skim milk-TBST, and blocking is carried out at 37 ℃ for 1.5 hours.
TABLE 3 results of square matrix titration of blocking fluid and blocking time (OD 450 )
Tab 1.Block titration results at various blocking solution and blocking time(OD 450 )
Example 5 determination of the optimal response time of serum to be examined
The positive serum sample and the negative serum sample are respectively incubated for 30min, 60min and 90min at 37 ℃ under the optimal conditions determined in the previous step, and three parallel experiments are carried out on each group, wherein the optimal reaction time is the maximum P/N value which is larger than 2.1.
The best reaction time was determined by incubating the serum to be tested at 37℃for 60min, which best meets the set conditions as set forth in Table 4.
TABLE 4 results (OD 450 )
Tab 4.Values of different incubation time of serum to be tested (OD 450 )
Example 6 determination of optimal color development time
According to the determined optimal conditions, after adding the substrate, respectively reacting for 5min, 10min, 15min and 20min at 37 ℃, carrying out three parallel experiments on each group, and determining the optimal color development time when the P/N value is larger than 2.1 and the maximum value is the maximum value.
The color of the substrate deepened with the extension of the color development time, and the time node of P/N maximum at the time of development for 10min was shown in Table 5, so that the optimum development time was determined to be 15min.
TABLE 5 results (OD) 450 )
Tab 5.Values of samples after coloration for various minutes (OD 450 )
Example 7 determination of cut-off value
32 negative control sera were assayed by established indirect ELISA to calculate OD 450 The mean (x) and Standard Deviation (SD) of the method, the Cut-off value of the method is determined.
Experiments were performed on 32 negative control sera according to the established indirect ELISA method, with an OD450 average of 0.071 and a standard deviation of 0.012, according to the formulaThe theoretical threshold (Cut-off) of the indirect ELISA method was 0.095, but the main objective of the method was to evaluate the ability of the vaccine to excite specific antibodies, so that the improvement criterion was calculated as 0.149 according to the OD450 mean of the negative control serum X2.1 as the Cut-off of the method, as shown in FIG. 1.
Effect example 1 specificity verification
The established method is used for detecting positive serum and negative control serum of novel coronaviruses, chikungunya viruses, katavirus, porcine circovirus type 2, porcine circovirus type 3 and transmissible gastroenteritis viruses, and judging whether the positive serum and the negative control serum are positive so as to verify the specificity of the method.
In order to verify the specificity of the established method, the indirect ELISA method is used for detecting the positive serum of the novel coronavirus, chikungunya virus, katavirus, porcine circovirus type 2, porcine circovirus type 3 and transmissible gastroenteritis virus mice, and the results are negative, and are shown in Table 6, so that the indirect ELISA method has good specificity.
TABLE 6 method specificity test results [ (]OD 450 )
Tab 6.Result of specificity test
Effect example 2 stability verification
Three laboratory-prepared Nipag G protein mouse positive sera were respectively provided with 3 duplicate wells in the group and 3 repeated experiments between the groups, CV was calculated, and the stability of the method was judged.
To verify the repeatability of the study setup method, 3 positive serum samples prepared in the laboratory were selected, and 3 replicates in the group and three replicates between groups were tested, with an intra-group CV of 1.1% -8.5% and an inter-group CV of 6.2% -7.0%, as shown in table 7, to demonstrate that the indirect ELISA method has good reproducibility.
TABLE 7 stability test results
Tab 7.Results of stability test
The foregoing is merely a preferred embodiment of the present invention and it should be noted that modifications and adaptations to those skilled in the art may be made without departing from the principles of the present invention, which are intended to be comprehended within the scope of the present invention.

Claims (6)

1. An indirect ELISA detection method of anti-Nipah virus G protein antibody, which is characterized in that,
coating an ELISA plate with Nipah virus G protein as an antigen, cleaning liquid, washing, sealing, washing, adding diluted primary antibody, incubating, cleaning primary antibody, washing, adding ELISA secondary antibody, incubating, cleaning secondary antibody, washing, developing, incubating, and terminating reaction to obtain OD 450 A value;
the primary antibody comprises positive sample serum or negative sample serum;
the coating concentration of the antigen is 2-10 mug/mL; the dilution of the primary antibody was 1: 200-1: 800; the incubation time of the primary antibody is 30-90 min;
the working concentration of the enzyme-labeled secondary antibody is 1:5000-1:20000; the incubation time of the enzyme-labeled secondary antibody is 30-120 min;
the sealing is specifically as follows: taking 5% skim milk-TBST as a sealing liquid, and sealing for 1.5h at 37 ℃;
the color development time is 5-20 min;
the coating concentration of the antigen is 10 mug/mL; the dilution of the primary antibody was 1:400, wherein the incubation time of the primary antibody is 60min;
the preparation method of the Nipagin virus G protein comprises the following steps:
nipah virus G protein sequence analysis, design and synthesis
Comparing the gene sequences of the G proteins according to 28 whole genome sequences of the Nipagin viruses on NCBI, drawing a evolutionary tree, and analyzing the amino acid homology of the G proteins; analysis was performed by an on-line hydrophobicity analysis tool and an epitope prediction tool to obtain the nipah virus representative strain NCBI accession number: the sequence of the protein G of AF212302.2, which is named sG by design, is deleted from the tail region, the transmembrane hydrophobic region and part of the stem region of the cytoplasm, and only the extracellular spherical head region is reserved; adding a human tissue plasminogen activator signal peptide sequence and a Strep tag at the N end, respectively adding enzyme cutting sites HindIII and BamHI at the two ends, and performing codon optimization and synthesis by Shanghai Biotechnology limited company after sequence design is completed to obtain glycerol bacteria containing a target gene plasmid pUC 57-NiV-sG;
construction of recombinant plasmids
Inoculating 10 mu L of glycerol bacteria containing a target gene plasmid pUC57-NiV-sG into 5mL of liquid LB culture medium with ampicillin resistance, performing shake culture at 37 ℃ for 12h, performing small-scale extraction preparation of plasmids, connecting HindIII and Bam HI to eukaryotic expression vectors pcDNA3.1 (+) after double enzyme digestion to obtain eukaryotic expression plasmids pcDNA-sG, transforming competent cells Trans1-T1 of the recombinant plasmids by a heat shock method, screening by ampicillin resistance, picking positive colonies, extracting plasmids, performing double enzyme digestion identification, sending the plasmids with correct identification to Shanghai bioengineering limited company for sequencing, performing mass preparation and purification after sequence comparison confirmation, and determining plasmid concentration for cell transfection;
cell transfection
The day before transfection, the Expi293F cells were incubated at 8% CO 2 、37℃And suspension culturing at 125r/min, and measuring the survival rate and living cell density of 293F cell culture of 60mL system, and growing living cells to 4.2X10 6 Cell cultures were diluted to 2.1X10 at a viability of 97% per mL 6 Cell density was 3X 10 cells per mL, day of transfection 6 Mu.g eukaryotic expression plasmid pcDNA-sG was combined with 3mL Opti-MEM at a concentration of 60. Mu.g/mL TM I Reduced Serum Medium mixing, 160 mu LExPiFectamine TM 293 Reagent and 2.8mL Opti-MEM TM I Reduced Serum Medium, mixing, incubating at room temperature for 5min, mixing the two, and incubating at room temperature for 20min; slowly transferring the solution into a cell culture shake flask, and slightly rotating the shake flask during the adding process;
collecting the protein of interest
After 12h of transfection, 300. Mu.L of transfection enhancer 1 and 3mL of transfection enhancer 2 were added, culture was continued for 96h, the culture was centrifuged at 800g for 10min, the culture supernatant and cell pellet were collected separately, the cell pellet was resuspended in IP lysate and lysed on ice for 10min, and disrupted with an ultrasonic cell disruptor for 10min.
2. The indirect ELISA detection method of claim 1, wherein the dilution of the second enzyme-labeled antibody is 1:20000 and the incubation time of the second enzyme-labeled antibody is 30min.
3. The indirect ELISA detection method according to any of claims 1 to 2, characterized in that the development time is 15min.
4. The indirect ELISA detection method as claimed in any one of claims 1 to 2 wherein the Cut-off value is the OD of the negative control serum 450 Mean x 2.1.
5. A method for determining a candidate vaccine against nipah virus, wherein antibodies induced after immunization of mice with the candidate vaccine are used as primary antibodies, and are detected according to the indirect ELISA detection method according to any of claims 1 to 4.
6. A method for determining anti-nipah virus G protein antibodies in a test serum, characterized in that the test serum is used as a primary antibody, and the detection is performed according to the indirect ELISA detection method according to any of claims 1 to 4.
CN202110667511.9A 2021-06-16 2021-06-16 Indirect ELISA detection method for anti-Nipah virus G protein antibody Active CN113391067B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110667511.9A CN113391067B (en) 2021-06-16 2021-06-16 Indirect ELISA detection method for anti-Nipah virus G protein antibody

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110667511.9A CN113391067B (en) 2021-06-16 2021-06-16 Indirect ELISA detection method for anti-Nipah virus G protein antibody

Publications (2)

Publication Number Publication Date
CN113391067A CN113391067A (en) 2021-09-14
CN113391067B true CN113391067B (en) 2023-07-25

Family

ID=77621493

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110667511.9A Active CN113391067B (en) 2021-06-16 2021-06-16 Indirect ELISA detection method for anti-Nipah virus G protein antibody

Country Status (1)

Country Link
CN (1) CN113391067B (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2905332A1 (en) * 2009-11-19 2015-08-12 Solis Biodyne Compositions for increasing polypeptide stability and activity, and related methods
CN109136241A (en) * 2018-09-08 2019-01-04 新疆畜牧科学院兽医研究所(新疆畜牧科学院动物临床医学研究中心) A kind of preparation method of peste des petits ruminants V protein protokaryon and eukaryotic expression, purifying and monoclonal antibody

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102816241B (en) * 2004-02-09 2015-07-22 人类基因科学公司 Albumin fusion proteins
CN102559612B (en) * 2011-12-21 2013-11-06 中国农业科学院哈尔滨兽医研究所 Recombinant newcastle disease virus LaSota vaccine strain for expressing Nipah virus encephalitis G-protein
CN109078179A (en) * 2013-09-05 2018-12-25 硕腾服务有限责任公司 Hendra and Nipah viral G glycoprotein immunogenic composition
WO2017004022A2 (en) * 2015-06-29 2017-01-05 The Board Of Trustees Of The Leland Stanford Junior University Degron fusion constructs and methods for controlling protein production
US11326183B2 (en) * 2016-02-12 2022-05-10 Bluebird Bio, Inc. VCN enhancer compositions and methods of using the same
CN110028579B (en) * 2019-05-05 2020-12-18 中国人民解放军军事科学院军事医学研究院 Monoclonal antibody of anti-nipah virus envelope glycoprotein and application thereof
AU2020382653A1 (en) * 2019-11-12 2022-06-02 Vanderbilt University Human Hendra virus and Nipah virus antibodies and methods of use therefor
CN111676248A (en) * 2020-07-02 2020-09-18 军事科学院军事医学研究院军事兽医研究所 Construction of SARS-CoV-2 VLP for expressing chimeric of S gene of novel coronavirus and M1 gene of influenza

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2905332A1 (en) * 2009-11-19 2015-08-12 Solis Biodyne Compositions for increasing polypeptide stability and activity, and related methods
CN109136241A (en) * 2018-09-08 2019-01-04 新疆畜牧科学院兽医研究所(新疆畜牧科学院动物临床医学研究中心) A kind of preparation method of peste des petits ruminants V protein protokaryon and eukaryotic expression, purifying and monoclonal antibody

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Fc-Based Recombinant Henipavirus Vaccines Elicit Broad Neutralizing Antibody Responses in Mice;Yaohui Li 等;Viruses;第12卷(第4期);1-20 *
Hendra and Nipah viruses: different and dangerous;Bryan T. Eaton 等;Nature Reviews Microbiology;第4卷;23–35 *
Successful use of tPA for thrombolysis in COVID related ARDS: a case series;Goyal Abhishek 等;Journal of thrombosis and thrombolysis;第51卷(第2期);1-4 *

Also Published As

Publication number Publication date
CN113391067A (en) 2021-09-14

Similar Documents

Publication Publication Date Title
von Messling et al. Rapid and sensitive detection of immunoglobulin M (IgM) and IgG antibodies against canine distemper virus by a new recombinant nucleocapsid protein-based enzyme-linked immunosorbent assay
Glaria et al. Visna/Maedi virus genetic characterization and serological diagnosis of infection in sheep from a neurological outbreak
Lang et al. Development of a peptide ELISA for discrimination between serological responses to equine herpesvirus type 1 and 4
CN113956362B (en) Recombinant feline parvovirus VP2 protein antigen and application thereof in antibody diagnosis and vaccine preparation
CN110221065B (en) Poultry bursa of sliding mycoplasma indirect ELISA detection kit
Aghokeng et al. Widely varying SIV prevalence rates in naturally infected primate species from Cameroon
Work et al. Differences in antibody responses against chelonid alphaherpesvirus 5 (ChHV5) suggest differences in virus biology in ChHV5-seropositive green turtles from Hawaii and ChHV5-seropositive green turtles from Florida
Wu et al. Serologic detection of duck enteritis virus using an indirect ELISA based on recombinant UL55 protein
Naves et al. Serological diagnosis of equine infectious anemia in horses, donkeys and mules using an ELISA with a gp45 synthetic peptide as antigen
CN113943354B (en) Recombinant feline herpesvirus gB protein antigen and application thereof in antibody diagnosis and vaccine preparation
CN113684189A (en) Novel chicken circovirus type 3 strain and detection system based on same
Basagoudanavar et al. Baculovirus expression and purification of peste-des-petits-ruminants virus nucleocapsid protein and its application in diagnostic assay
CN113391067B (en) Indirect ELISA detection method for anti-Nipah virus G protein antibody
Liu et al. Seroprevalence of human metapneumovirus (hMPV) in the Canadian province of Saskatchewan analyzed by a recombinant nucleocapsid protein‐based enzyme‐linked immunosorbent assay
CN113817753A (en) Expression of SARS-CoV-2 spike protein or its variant SΔ21Construction and application of pseudotyped VSV (VSV virus)
Lei et al. Development and application of nsp5-ELISA for the detection of antibody to infectious bronchitis virus
CN103255111B (en) Green fluorescent protein marked recombinant swine fever virus, its rescue method and application
CN115073557A (en) Recombinant high-purity African swine fever virus pK205R subunit protein and preparation method and application thereof
Rabenau et al. Comparison of the neutralizing and ELISA antibody titres to measles virus in human sera and in gamma globulin preparations
CN110568189B (en) Dog adenovirus type 1 antibody ELISA detection kit and application thereof
Xu et al. Development of A recombinant nucleocapsid based indirect ELISA for the detection of antibodies to avian metapneumovirus subtypes, A, B, and C
Bold et al. Development of an Indirect ELISA for the Detection of SARS-CoV-2 Antibodies in Cats
CN114807178B (en) African swine fever virus P72 protein C-terminal multi-epitope recombinant antigen and application thereof
Kunita et al. Simultaneous detection of antibodies to mouse hepatitis virus recombinant structural proteins by a microsphere-based multiplex fluorescence immunoassay
Carrozza et al. Seroconversion against SU5 derived synthetic peptides in sheep experimentally infected with different SRLV genotypes

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