SARS-COV-2 virus B.1.617.2 mutant strain DNA vaccine and application

文档序号:462064 发布日期:2021-12-31 浏览:58次 中文

阅读说明:本技术 Sars-cov-2病毒b.1.617.2突变株dna疫苗及应用 (SARS-COV-2 virus B.1.617.2 mutant strain DNA vaccine and application ) 是由 赵干 侯佳望 丁媛 俞庆龄 于 2021-12-03 设计创作,主要内容包括:本发明涉及生物技术领域,具体而言,提供了一种编码SARS-COV-2病毒B.1.617.2突变株抗原的DNA分子、DNA疫苗及应用。本发明提供的SEQ ID NO:1核酸序列在真核表达系统中能够高效转录和表达,而且具有免疫原性,表现在体液免疫和细胞免疫应答中,以此作为活性成分的核酸疫苗同样具有良好的免疫原性。(The invention relates to the field of biotechnology, and particularly provides a DNA molecule for coding a SARS-COV-2 virus B.1.617.2 mutant strain antigen, a DNA vaccine and application thereof. The invention provides SEQ ID NO: 1 in eukaryotic expression system, and has immunogenicity as shown in humoral immunity and cell immunity response, and the nucleic acid vaccine with the nucleic acid sequence as active component has excellent immunogenicity.)

1. A DNA molecule having the sequence set forth in SEQ ID NO: 1.

2. Biomaterial, characterized in that it comprises at least one of (a) - (c):

(a) a recombinant expression vector comprising the DNA molecule of claim 1;

(b) a cell comprising the DNA molecule of claim 1 or the recombinant expression vector of (a);

(c) a polypeptide encoded by the DNA molecule of claim 1.

3. The biomaterial according to claim 2, wherein the recombinant expression vector is a eukaryotic expression vector having a vector backbone of pVAX 1.

4. Use of the DNA molecule of claim 1 or the biomaterial of claim 2 or 3 in (a) or (B) as follows:

(A) preparing vaccine for preventing and/or treating SARS-COV-2 virus infection;

(B) preparing medicine for preventing and/or treating SARS-COV-2 virus caused relevant diseases.

5. The use of claim 4, wherein the SARS-COV-2 virus comprises a mutant B.1.617.2 strain, a wild-type strain, a mutant B.1.1.7 strain, a mutant B.1.351 strain, a mutant P.1 strain, a mutant B.1.2 strain, a mutant B.1 strain, a mutant B.1.525 strain, a mutant B.1.526 strain, a mutant C.37 strain, or a mutant B.1.617.1 strain.

6. A DNA vaccine comprising the DNA molecule of claim 1 or the recombinant expression vector of claim 2 or 3.

7. The DNA vaccine of claim 6, further comprising a pharmaceutically acceptable adjuvant, carrier, diluent or excipient;

and/or at least one drug having a therapeutic effect on SARS-COV-2 virus.

8. The DNA vaccine of claim 7, wherein the adjuvant comprises an aluminum adjuvant and/or a TLRs ligand and/or a metal ion and/or a cytokine and/or a chemokine adjuvant.

9. The method for producing a DNA vaccine according to any one of claims 6 to 8, wherein a recombinant vector comprising the DNA molecule according to claim 1 is introduced into a host cell and cultured, and the recombinant vector in the host cell is extracted to obtain the DNA vaccine.

10. The use of the DNA vaccine of any one of claims 6-8, comprising the following (i) - (ii):

(i) preparing a product for resisting SARS-COV-2 virus infection;

(ii) preparing products for preventing immunopathological damage caused by SARS-COV-2 virus;

the SARS-COV-2 virus comprises a B.1.617.2 mutant strain, a wild strain, a B.1.1.7 mutant strain, a B.1.351 mutant strain, a P.1 mutant strain, a B.1.2 mutant strain, a B.1 mutant strain, a B.1.525 mutant strain, a B.1.526 mutant strain, a C.37 mutant strain or a B.1.617.1 mutant strain.

Technical Field

The invention relates to the field of biotechnology, in particular to a DNA molecule for coding SARS-COV-2 virus B.1.617.2 mutant strain antigen, a DNA vaccine and application thereof.

Background

SARS-CoV-2 is a single-stranded positive-strand RNA virus with an envelope structure, which is highly susceptible to mutation. The B.1.617 mutant strain that has been identified so far has three subtypes: b.1.617.1, B.1.617.2 and B.1.617.3. Genetic testing and more detailed sequencing data on viral samples show that the b.1.617.2 variant carries mutations designated 452R and 478K, and therefore, a more effective vaccine against the mutant is needed.

Disclosure of Invention

It is an object of the present invention to provide a DNA molecule encoding the antigen of the mutant strain B.1.617.2 of SARS-COV-2 virus, which alleviates at least one of the technical problems of the prior art.

Another object of the present invention is to provide a biomaterial comprising the DNA molecule.

The invention also aims to provide application of the biological material.

The fourth purpose of the present invention is to provide a SARS-COV-2 virus B.1.617.2 mutant DNA vaccine comprising the above DNA molecule.

The fifth object of the present invention is to provide a method for producing the DNA vaccine.

The sixth object of the present invention is to provide the use of the above DNA vaccine.

In order to achieve the above purpose of the present invention, the following technical solutions are adopted:

the invention provides a DNA molecule for coding SARS-COV-2 virus B.1.617.2 mutant strain antigen, the DNA molecule has the nucleotide sequence shown in SEQ ID NO: 1, or a nucleotide sequence corresponding to SEQ ID NO: 1 has a nucleotide sequence of at least 90% identity.

The present invention also provides a biomaterial comprising:

(a) recombinant expression vectors comprising the DNA molecules described above;

(b) a cell comprising the DNA molecule of (a) or the recombinant expression vector of (a);

(c) a polypeptide encoded by the DNA molecule described above.

Further, the recombinant expression vector comprises a eukaryotic expression vector, and the eukaryotic expression vector comprises pVAX 1.

Preferably, the cells include HEK293, CHO, COS-7, and the like.

The invention also provides the application of the DNA molecule or the biological material in the following (A) or (B):

(A) preparing vaccine for preventing and/or treating SARS-COV-2 virus infection;

(B) preparing medicine for preventing and/or treating SARS-COV-2 virus caused relevant diseases.

Furthermore, the SARS-COV-2 virus comprises a mutant B.1.617.2 strain, a wild strain, a mutant B.1.1.7 strain, a mutant B.1.351 strain, a mutant P.1 strain, a mutant B.1.2 strain, a mutant B.1 strain, a mutant B.1.525 strain, a mutant B.1.526 strain, a mutant C.37 strain or a mutant B.1.617.1 strain.

The invention also provides a DNA vaccine of the mutant strain of SARS-COV-2 virus B.1.617.2, which comprises the DNA molecule.

Further, the DNA molecule is present in a recombinant expression vector comprising pVAX 1.

Further, the DNA vaccine also comprises pharmaceutically acceptable adjuvant, carrier, diluent or excipient;

and/or at least one drug having a therapeutic effect on SARS-COV-2 virus.

Preferably, the adjuvant comprises an aluminium adjuvant and/or a TLRs ligand and/or a metal ion such as Mn2+、Zn2+And/or cytokine and/or chemokine adjuvants, and the like.

The invention also provides a preparation method of the DNA vaccine, which comprises the steps of introducing the recombinant vector containing the DNA molecule into host cells, culturing, and extracting the recombinant vector in the host cells to obtain the DNA vaccine.

In addition, the invention also provides the application of the DNA vaccine, which comprises the following (i) to (iii):

(i) regulating the immune function of the organism;

(ii) resisting SARS-COV-2 virus infection;

(iii) prevention of immunopathological damage;

the SARS-COV-2 virus comprises a B.1.617.2 mutant strain, a wild strain, a B.1.1.7 mutant strain, a B.1.351 mutant strain, a P.1 mutant strain, a B.1.2 mutant strain, a B.1 mutant strain, a B.1.525 mutant strain, a B.1.526 mutant strain, a C.37 mutant strain or a B.1.617.1 mutant strain.

Compared with the prior art, the invention has the following beneficial effects:

the invention optimizes the coding DNA sequence of the mutant Spike protein of SARS-COV-2 virus B.1.617.2 by using different optimization algorithms to obtain the protein with the sequence shown in SEQ ID NO: 1 or a nucleotide sequence corresponding to SEQ ID NO: 1 has a nucleotide sequence of at least 90% identity. The DNA molecule can efficiently transcribe and express the SARS-COV-2 virus B.1.617.2 mutant Spike antigen, has immunogenicity, and can induce specific humoral immunity and cell immunity response.

Based on the beneficial effect of the DNA molecule of the SARS-COV-2 virus B.1.617.2 mutant antigen, the invention also provides a DNA vaccine comprising the DNA molecule. The DNA vaccine can be effectively transcribed and expressed in mammalian cells, has good immunogenicity, and can remarkably stimulate experimental animals to generate antigen-specific antibodies on 14 days after primary immunization and 7 days after boosting immunization for humoral immune response; in the case of cellular immune responses, the DNA vaccine is capable of inducing not only high levels of antigen-specific IFN- γ and IL-4 responses, but also the generation of antigen-specific CD8IFN γ T cell subsets.

Based on the DNA vaccine, the DNA vaccine provided by the invention can adjust the immune function of organisms and effectively prevent

SARS-COV-2 virus and its mutant strain, especially B.1.617.2 mutant strain infection, and can also be used for dry treatment of diseases caused by SARS-COV-2 virus and its mutant strain, especially B.1.617.2 mutant strain.

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, and it is obvious that the drawings in the following description are some embodiments of the present invention, and other drawings can be obtained by those skilled in the art without creative efforts.

FIG. 1 is a graph showing the result of codon optimization index scoring of the DNA sequence encoding the Spike protein of the B.1.617.2 mutant strain;

FIG. 2 is a graph showing the results of GC content scoring after optimization of the DNA sequence encoding the Spike protein of the B.1.617.2 mutant strain;

FIG. 3 is a graph showing the result of scoring the number of negative regulatory elements after optimizing the DNA sequence encoding the Spike protein of the B.1.617.2 mutant strain;

FIG. 4 is a graph of the results of qPCR fold expression after optimization of the DNA sequence encoding the Spike protein of the B.1.617.2 mutant strain;

FIG. 5 shows the qPCR expression results of the candidate DNA vaccines of the Xinguan wild strain and the B.1.617.2 mutant strain provided by the embodiment of the invention;

FIG. 6 shows the Western Blot detection result of antigen protein of the new crown wild strain and the candidate DNA vaccine of B.1.617.2 mutant strain provided by the embodiment of the invention;

FIG. 7 shows the results of antigen-specific antibodies 14 days after the primary immunization of the candidate DNA vaccines of the new crown wild strain and the B.1.617.2 mutant strain provided by the embodiment of the present invention;

FIG. 8 shows the results of antigen-specific antibodies at day 7 after the boosting immunization of the candidate DNA vaccines of the Xinguan wild strain and the B.1.617.2 mutant strain provided by the embodiment of the present invention;

FIG. 9 shows the results of antigen-specific IFN-. gamma.ELISOPT at day 7 after the boosting immunization of the candidate DNA vaccines of the new crown wild strain and the B.1.617.2 mutant strain provided in the example of the present invention;

FIG. 10 shows the results of antigen-specific IL-4 ELISOPT at day 7 after the boosting immunization of the candidate DNA vaccine of the new crown wild strain and the B.1.617.2 mutant strain provided by the embodiment of the present invention;

FIG. 11 shows the result of antigen-specific CD8IFN gamma T cell subsets at 7 days after the booster immunization of the candidate DNA vaccines of the new crown wild strain and the B.1.617.2 mutant strain provided by the embodiment of the invention.

Detailed Description

Unless defined otherwise herein, scientific and technical terms used in connection with the present invention shall have the meanings that are commonly understood by one of ordinary skill in the art. The meaning and scope of a term should be clear, however, in the event of any potential ambiguity, the definition provided herein takes precedence over any dictionary or extrinsic definition. In this application, the use of "or" means "and/or" unless stated otherwise. Furthermore, the use of the term "including" and other forms is not limiting.

Generally, the nomenclature used, and the techniques thereof, in connection with the cell and tissue culture, molecular biology, immunology, microbiology, genetics and protein and nucleic acid chemistry and hybridization described herein are those well known and commonly employed in the art. Unless otherwise indicated, the methods and techniques of the present invention are generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification. Enzymatic reactions and purification techniques are performed according to the manufacturer's instructions, as commonly practiced in the art, or as described herein. The nomenclature used in connection with the analytical chemistry, synthetic organic chemistry, and medical and pharmaceutical chemistry described herein, and the laboratory procedures and techniques thereof, are those well known and commonly employed in the art.

The technical solutions of the present invention will be described clearly and completely with reference to the following embodiments, and it should be understood that the described embodiments are some, but not all, embodiments of the present invention. All other embodiments, which can be derived by a person skilled in the art from the embodiments given herein without making any creative effort, shall fall within the protection scope of the present invention.

According to one aspect of the invention, the invention optimizes the coding DNA sequence of the SARS-COV-2 virus B.1.617.2 mutant strain Spike protein by using different optimization algorithms to obtain the protein with the sequence shown in SEQ ID NO: 1 or a nucleotide sequence corresponding to SEQ ID NO: 1 has a nucleotide sequence of at least 90% identity. The DNA molecule can be transcribed efficiently, is more beneficial to efficiently expressing the SARS-COV-2 virus B.1.617.2 mutant Spike antigen in a eukaryotic expression system, has good immunogenicity, and can induce specific humoral immunity and cellular immune response.

It is understood that, in the present invention, "identity" refers to similarity between nucleotide sequences, including nucleotide sequences that are identical to SEQ ID NO: 1 (e.g., can be, but is not limited to, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more) identical.

Optionally, the optimization also comprises replacing the SARS-COV-2 virus B.1.617.2 mutant gene signal peptide with a high-efficiency expression signal peptide, so as to improve the expression efficiency of the DNA sequence in the host.

The invention also provides biological materials related to the DNA molecules:

(a) recombinant expression vectors comprising the DNA molecules provided by the invention. Wherein the vector can be a eukaryotic expression vector, and the protein encoded by the DNA molecule is generated through a cell transcription and translation mechanism. Alternatively, the vector may have expression signals such as a strong promoter, a strong stop codon, regulation of the distance between the promoter and the cloned gene, and insertion of transcription termination sequences and PTIS. Preferably, the eukaryotic expression vector includes pVAX1, but is not limited to any other expression vector capable of expressing DNA and enabling a cell to translate sequences into antigens recognized by the immune system.

(b) The cell is obtained by introducing the DNA molecule provided by the invention or the recombinant expression vector of (a) into a host cell. The host cell may be a eukaryotic cell, typically an insect cell, a yeast cell, an avian cell, or a mammalian cell, among other suitable host cells. The cells include HEK293, CHO, COS-7 and the like.

(c) Polypeptides encoded by the DNA molecules provided by the invention. Based on the polypeptide, an antibody, such as a monoclonal antibody or a polyclonal antibody, capable of specifically binding thereto may also be provided.

It can be understood that the biological material provided by the invention can be directly applied to the production of different requirements and scenes as a biological module.

According to another aspect of the present invention, the present invention also provides the use of the above DNA molecule or biomaterial in the preparation of a vaccine for preventing and/or treating SARS-COV-2 virus infection, and/or in the preparation of a medicament for preventing and/or treating related diseases caused by SARS-COV-2 virus, such as lung injury, brain injury, liver and kidney injury, and heart injury.

Preferably, the SARS-COV-2 virus comprises a mutant B.1.617.2 strain, a wild strain, a mutant B.1.1.7 strain, a mutant B.1.351 strain, a mutant P.1 strain, a mutant B.1.2 strain, a mutant B.1 strain, a mutant B.1.525 strain, a mutant B.1.526 strain, a mutant C.37 strain or a mutant B.1.617.1 strain.

Based on the beneficial effect of the DNA molecule of the SARS-COV-2 virus B.1.617.2 mutant antigen, the invention also provides a DNA vaccine comprising the DNA molecule.

The DNA vaccine can effectively transcribe and express the mutant Spike antigen of SARS-COV-2 virus B.1.617.2 in mammalian cells, more efficient immune response is excited, and for humoral immune response, the DNA vaccine can obviously excite experimental animals to generate antigen specific antibodies on the 14 th day after primary immunization and the 7 th day after enhanced immunization; in the case of cellular immune responses, the DNA vaccine is capable of inducing not only high levels of antigen-specific IFN- γ and IL-4 responses, but also the generation of antigen-specific CD8IFN γ T cell subsets.

In some embodiments, the DNA vaccine further comprises a pharmaceutically acceptable adjuvant, carrier, diluent, or excipient to increase the ability of its active ingredient DNA molecule to generate an immune response in a subject. Wherein the pharmaceutically acceptable adjuvant may be selected from aluminium adjuvants and/or TLRs ligands and/or metal ions such as Mn2+、Zn2+And/or cytokine and/or chemokine adjuvants, and the like.

In other embodiments, the DNA vaccine further comprises at least one drug that is therapeutic against SARS-COV-2 virus to enhance the therapeutic effect of the vaccine against the associated disease caused by SARS-COV-2 virus.

The action mechanism of the DNA vaccine provided by the invention is as follows: the SARS-COV-2 virus B.1.617.2 mutant strain surface antigen Spike antigen coding DNA is firstly optimized by different optimization algorithms, and then the wild type gene signal peptide is replaced by high-efficiency expression signal peptide and inserted into eukaryotic expression vector, and the vector is introduced into host cell, so that the virus Spike antigen is efficiently expressed in the host cell, and the antiviral humoral immune response and the cellular immune response are systematically activated by the antigen presentation process. Antibodies generated by the activated humoral immune response can prevent viral entry, and the activated cellular immune response can further clear virus-infected cells and modulate adverse reactions due to potential side effects of ADE.

Based on the action mechanism, the invention also provides the application of the DNA vaccine, which comprises the following steps:

(i) regulating the immune function of the organism;

(ii) resisting SARS-COV-2 virus infection;

(iii) preventing immunopathological damage.

The invention is further illustrated by the following examples. The materials in the examples are prepared according to known methods or are directly commercially available, unless otherwise specified.

Example 1: optimized screening of nucleic acids encoding S proteins

In order to increase the protein expression of the target protein in the host cell, the nucleic acid sequence of the target gene needs to be optimized, and the general principle of nucleic acid sequence optimization is as follows: (1) optimizing the degenerate codon according to the preference of the host cell to the nucleic acid codon, so that the optimized sequence contains more nucleic acid codons which are beneficial to the recognition of the host cell; (2) further optimizing the GC content in the nucleic acid sequence on the basis of codon preference optimization, so that the sequence with the optimized GC content can express more target proteins; (3) optimizing the nucleic acid sequence to make it able to transcribe more stable mRNA, facilitating translation of the target protein; (4) the host-biased codon frequency was changed to increase the CAI index (codon adaptation index). The application optimizes the coding nucleotide sequence of the surface protein Spike of the wild SARS-COV-2 virus B.1.617.2 mutant strain, and adjusts the GC content in the nucleotide sequence; meanwhile, the codon frequency of host preference is changed, and the CAI (codon adaptation index) index is improved; the method has the advantages of reducing free energy for forming an RNA secondary structure, reducing the proportion of Negative CIS elements, reducing the proportion of repeated sequences in the sequence, optimizing signal peptide, and combining the algorithm which is formed by the inventor through years of experience in the field and is specific to the company, so that the expression level of the signal peptide can be further improved, the optimized nucleotide sequence can be obtained, and the optimized nucleotide sequence can be prepared into a nucleic acid vaccine.

The optimization process comprises the following steps: selecting a wild B.1.617.2 mutant strain Spike (S protein) sequence (EPI _ ISL _3161912, GISAID) before optimization as an antigen sequence, and obtaining the sequence shown in SEQ ID NO: 1, and obtaining the nucleotide sequence shown in SEQ ID NO: 3. For the optimized wild B.1.617.2 sequence and the optimized SEQ ID NO: 1 and the conventional commercial optimization of SEQ ID NO: 3 is scored; in the aspect of optimizing and increasing expression of DNA sequences, key indexes of optimizing effect and DNA optimization are as follows: codon optimization indices are positively correlated, GC content is positively correlated, and the number of negative regulatory elements is negatively correlated. As shown in FIGS. 1-3, the results show that after the B.1.617.2 mutant strain Spike sequence is optimized, the optimization strategy adopted by the invention is found to be obviously improved in key indexes compared with the conventional commercial optimization strategy, and the optimization strategy adopted by the invention can be predicted to increase the expression efficiency of the optimized gene.

The B.1.617.2 mutant strain is optimized into a wild sequence and an optimized SEQ ID NO: 1 and SEQ ID NO: 3 into pVAX1 vector (ThermoFisher, cat # V26020) to obtain 3 kinds of plasmid DNA, pB.1.617.2-wild, pB.1.617.2 and pB.1.617.2-optimized. 3 plasmids are respectively transfected into a HEK293T cell strain for 48h, RNA is extracted, and the transcription level of plasmid DNA obtained by different optimization modes is identified by adopting a qPCR method. The results are shown in FIG. 4, the optimized B.1.617.2 mutant DNA sequence of the invention can be increased by more than 300 times at the RNA transcription level compared with the wild sequence before optimization, and can be increased by more than 6 times at the RNA transcription level compared with the molecule conventionally optimized in the commercial database, further demonstrating that the optimized nucleic acid molecule of the invention is superior to the molecule conventionally obtained in the commercial database. The improvement of the transcription level of the DNA vaccine can improve the protein expression quantity, thereby improving the immune effect of the DNA vaccine, the sequence designed and obtained by the invention is obviously improved in the transcription level, the protein expression quantity is also obviously improved, and the obvious better immune effect is obtained.

Example 2: construction process of DNA vaccine

1. Preparation method of new coronavirus candidate DNA vaccine

1.1. Construction of plasmids

As described in example 1, the mutant b.1.617.2 (EPI _ ISL _3161912, GISAID) sequence was optimized to obtain the sequence shown in SEQ ID NO: 1, the nucleotide sequence shown as SEQ ID NO: 1 is inserted between the BanH I and Xho I sites of the vector pVAX1 to obtain the new coronavirus B.1.617.2 mutant plasmid (pB.1.617.2).

Optimization based on the new crown wild type sequence (MN 908947.3, NCBI) gave SEQ ID NO: 2, the nucleotide sequence shown as SEQ ID NO: 2 is inserted between the BanH I and Xho I sites of the vector pVAX1 to obtain a new coronavirus wild strain plasmid (pWT). pWT the wild strain vaccine is a product aiming at the wild strain at the early stage of the company, is about to enter the phase III clinic at present, and has very excellent immune effect.

DNA vaccine sequence transformation

From a freezer at-80 ℃ 100. mu.l of DH10B competent cell suspension was removed and thawed on ice. Add plasmid DNA solution (volume not more than 10 u l) gently shake, ice placed for 30 min. The mixture was heated in a water bath at 42 ℃ for 70 seconds and rapidly cooled on ice for 5 min. 0.9ml of LB liquid medium (containing no antibiotics) was added to the tube, mixed well and cultured with shaking at 37 ℃ for 45min to restore the bacteria to normal growth state. Shaking the bacterial liquid uniformly, coating 100 μ L of the bacterial liquid on a screening plate containing appropriate antibiotics, placing the bacterial liquid with the front side upward, inverting the culture dish after the bacterial liquid is completely absorbed by the culture medium, and culturing for 12-16h at 37 ℃. The single-clone cells with uniform shapes were selected, and the colonies were picked up by using a sterile pipette tip and then cultured overnight at 37 ℃ in 5mL of LB selection medium containing 50mg/mL of kanamycin.

DNA vaccine plasmid extraction

The above-mentioned bacterial suspension was added to 200-400mL LB selection medium containing kanamycin (50 mg/mL of mother liquor, 1:1000 used) at 1:1000, and cultured at 37 ℃ at 200rpm for 12-16 h. Plasmid extraction was performed with an EndoFreen Plasmid Maxi kit (QIAGEN, Germany): centrifuging the cultured bacterial liquid for 12-16h at 8000rpm and 4 ℃ for 10min, removing the supernatant, collecting the bacterial body, adding 10ml of Buffer P1 heavy suspension, adding 10ml of Buffer P2, slightly reversing for 4-6 times, mixing, incubating at room temperature for 5min, and fully lysing. 10ml of Buffer P3 was added to the mixture, after termination of lysis by gentle inversion for 4-6 times, all were transferred to a QIAfilter Cartridge, incubated at room temperature for 10min, and the supernatant was filtered by adding a plug. The filtrate was transferred to a clean endotoxin-free 50ml centrifuge tube, 2.5ml Buffer ER was added, the mixture was mixed by gentle inversion 10 times and incubated on ice for 30 min. The QIAGEN-tip 500 was removed and added to a 10ml Buffer QBT equilibrated column, and the above liquid was transferred to the column, and the plasmid was adsorbed by gravity flow, washed 2 times with 30ml Buffer QC, and eluted with 15ml Buffer QN. Each tube was precipitated with 10.5ml isopropanol and centrifuged at 4000g for 30min at 4 ℃. The supernatant was discarded, washed with 70% ethanol 1 time, centrifuged at 4000g for 10min at 4 ℃. Abandoning the supernatant, air-drying the precipitate, and adding 500 μ l of endotoxin-free water into each sample to resuspend the plasmid, thereby obtaining the DNA vaccine plasmid.

Example 3: mammalian cell transcriptional identification of new coronavirus candidate DNA vaccines

To verify whether the plasmid constructed in example 2 can be transcribed efficiently in mammalian cells, it was identified by methods of in vitro transfection of DNA, extraction of RNA, and qPCR.

1. DNA vaccine in vitro transfection

The frozen HEK293T cell line was removed from the liquid nitrogen and DMSO was removed by centrifugation at 1000rpm for 5 minutes after a 37 ℃ water bath. Washing with serum-free DMEM medium at 37 deg.C and 5% CO in 5ml DMEM medium containing 10% calf serum2Culturing for 2-3 generations. The cells were digested with pancreatin (containing 0.25% EDTA) at 37 ℃ for 1min and stopped with complete medium, and then incubated at 2-4X 106The density of cells/well was plated on a 60mm dish and 5ml growth medium (without 1% double antibody) was added at 37 ℃ with 5% CO2Culturing in an incubator for 24 h.

Mu.g of pWT and 4. mu.g of pB.1.617.2 were added to 500. mu.l of serum-free OPTI-MEM medium, and gently mixed, and 24. mu.l of cationic liposome was added to 500. mu.l of serum-free OPTI-MEM medium, and gently mixed, and left at room temperature for 5min, and the two plasmids were mixed with liposome 1:1, and left at room temperature for 20min, to obtain a plasmid DNA/liposome complex.

The plasmid DNA/liposome complex was added to a 60mm culture dish at 1 ml/dish and incubated for 24 hours at 37 ℃ with 5% CO2Incubators were incubated for 48 hours for subsequent experiments.

2. Post-transfection RNA extraction

The cells transfected for 48 hours above were collected by digestion, resuspended in 1ml complete medium, 100. mu.l was aspirated for RNA extraction, and the remaining resuspension was subjected to subsequent WB sample preparation.

The aspirated 100. mu.l of cell suspension was centrifuged at 4000rpm for 5 minutes, the supernatant was discarded, and 350. mu.l of TRK Lysis Solution (containing 20%. beta. -mercaptoethanol) was added to each sample for Lysis. Each sample was then quenched with an additional 350. mu.l of 70% ethanol (made up with DEPC water) and mixed by blowing with a gun.

The mixture was transferred to a HiBind RNA Column, centrifuged at 10000g for 1min, and the filtrate was discarded. Mu.l of Wash Buffer I was added to each column, 10000g was centrifuged for 1min, and the filtrate was discarded. 500. mu.l of Wash Buffer II was added to each column and washed 2 times, each time, 10000g was used for centrifugation for 1min, and the filtrate was discarded. The centrifuge speed was adjusted to the highest speed (17000 g) and centrifuged for 2min to volatilize ethanol from the column. The column was transferred to a clean 1.5ml centrifuge tube without DNA and RNase, left at room temperature for 3-5min, after ethanol was completely evaporated, 50. mu.l of RNase-Free Water was added to each sample, incubated at room temperature for 5min, and centrifuged at 17000g for 1 min. The filtrate was aspirated and added to the column again, incubated at room temperature for 5min, centrifuged at 17000g for 1min to collect RNA, and stored at-80 ℃.

3. RNA reverse transcription, qPCR reaction

The RNA concentration was quantified using a microplate reader (readings were performed using OD 260/280), a solution was prepared based on the number of desired PCR samples n (n = sample number +1 tube negative control +1 tube positive control), and 10. mu.l of a reaction system (2. mu.l of 5 Xg DNA digaster Buffer, 1. mu.l of gDNA digaster, 100ng of RNA) was prepared for each sample, and RNase free ddH was used2The volume was adjusted to 10. mu.l) by O, the mixture was gently blown down by a gun and incubated at 42 ℃ for 2 min. To each sample was added 10. mu.l of 2 XHifair II Supermix plus, gently blown with a gunMixing, and incubating at 25 deg.C for 5min, 42 deg.C for 30min, and 85 deg.C for 5 min. The collected cDNA was stored at-20 ℃ for further use.

And (3) carrying out reaction on the cDNA product obtained by reverse transcription according to a qPCR kit. The reaction system is as follows: 10 mul of Hieff qPCR SYBR Green Master Mix (No Rox), 0.4 mul of each target forward primer and reverse primer, 1 mul of cDNA template and 20 mul of sterile ultrapure water complement total volume. And (3) PCR reaction conditions: 95 ℃, 5min, 95 ℃, 10 s, 56 ℃, 30s, 72 ℃, 30s for 40 cycles. Comparing the expression level of the target gene with that of an internal reference and then adopting 2-△△CAnd (4) calculating by using the method.

And (4) conclusion: as shown in FIG. 5, the candidate DNA vaccines of the new crown wild strain pWT and the pB.1.617.2 mutant strain were able to promote the transcription of antigen RNA at a high level compared with the empty vector (pVAX 1) 48 hours after in vitro transfection.

Example 4: identification of mammalian cell antigen protein expression of new coronavirus candidate DNA vaccine

To further verify whether the plasmid constructed in example 2 can be efficiently expressed in mammalian cells, it was identified by extracting antigen proteins and Western Blot method.

1. Protein extraction

The new crown plasmid pWT and pB.1.617.2 were transfected into HEK293T cell line, respectively, after 48 hours of transfection, the transfected culture solution was removed, washed once with precooled PBS, PBS was discarded, 150. mu.l of lysis buffer (EDTA and protease inhibitor were added at a ratio of 1:100 before use) was added, mixed well and blown 10 times. Centrifuge at 4 degrees at 12,000rpm for 5 minutes. The supernatant was aspirated into a 1.5mL centrifuge tube, 50. mu.L of the supernatant was removed for each sample, 12.5. mu.L of 5 XP buffer was added, and the mixture was boiled in boiling water for 10min and then immediately centrifuged.

2. Sample loading and SDS-PAGE electrophoresis

Adding 62.5 μ l of boiled and centrifuged supernatant sample into SDS-PAGE gel well, switching on power supply, adjusting to constant voltage of 200V, and performing electrophoresis for 45 min. After the electrophoresis, SDS-PAGE was taken out to prepare a membrane. Soaking the PVDF membrane in methanol for 30s for activation, and placing the PVDF membrane in a 1 × rotating membrane equilibrium solution for 1 min.

3. Rotary film

With the positive electrode as the bottom surface, the following steps are carried out: the eBlot L1 membrane-transfer gasket, the PVDF membrane, the gel and the eBlot L1 membrane-transfer gasket were sequentially stacked, and the interlayer air bubbles were removed by a tube every time the stack was stacked. And (3) sealing: the PVDF membrane was removed and placed in a glass box containing 1 XTBST +5% skimmed milk powder and incubated for 1h at room temperature at 90rpm in a shaker. Washing: the PVDF membrane was washed 3 times in 1 XTSST for 10 minutes each time with shaking at 90rpm on a shaker. Primary antibody incubation: the PVDF membrane was reacted with a primary antibody (Rabbit anti S protein polyclonal antibody,1: 4000) and incubated at 90rpm in a shaker at room temperature for 1 hour. Washing: the PVDF membrane was washed 5 times in 1 XTSST for 10 minutes each time, shaking at 90rpm in a shaker. And (3) secondary antibody incubation: the PVDF membrane was placed in a secondary antibody solution (BD Pharmingen HRP Anti human IgG, 1:5000 dilution) for reaction and incubated at room temperature for 1h at 90rpm on a shaker. Washing: the PVDF membrane was washed 5 times in 1 XTSST for 10 minutes each time with shaking at 90rpm on a shaker. Color development: taking 3ml of chemiluminescence solution A and 3ml of chemiluminescence solution B, mixing the chemiluminescence solution A and the chemiluminescence solution B in a proportion of 1: mixing the materials according to the proportion of 1, adding the mixture into a PVDF membrane, incubating for 1-2min, and photographing.

And (4) conclusion: as shown in FIG. 6, the candidate DNA vaccines of the new crown wild strain pWT and pB.1.617.2 mutant strain were able to express high level of antigen protein in cells compared to the empty vector (pVAX 1) 48 hours after transfection in vitro.

Example 5: immunogenicity verification of new crown candidate DNA vaccine

To assess the immunogenicity of the vaccine prepared in example 2, and the impact of the immunization strategy on humoral and cellular immune responses, 6-week-old C57BL/6 female mice, free of specific pathogens, were purchased from Calvens bagger and maintained in the animal facility at the Amelanchivenn Advaccine laboratory (Suzhou). For vaccination with DNA vaccines: the DNA vaccine described in example 1 was injected into the anterior femoral muscle sequentially according to different grouped injection doses, followed by Electrical Pulses (EP). The Electrical Pulse (EP) device consists of two sets of pulses with a constant current of 0.2 Amp. The second pulse group is delayed by 3 seconds. In each group there are two 52 ms pulses with a delay of 198 ms between the pulses. The first prime was counted as day 0 and the second immunization (boost) was performed on day 14. Grouping experiments: (1) control group vector plasmid pVAX1-25 μ g; (2) the experimental group wild strain pWT-25 μ g; (3) the experimental group mutant pB.1.617.2-25 mug; on day 14, 21, a blood sample was collected from the mouse, and the serum was assayed for the specific antibody titer by ELISA. Immunized mice were sacrificed on day 7 post booster immunization to analyze cellular immune responses.

1. Evaluation of DNA vaccine elicited antigen-specific humoral immune responses

1.1 ELISA detection of antibody concentration

Antibody binding to SARS-CoV-2 RBD protein was assessed using an ELISA-based method. Nunc 96 well ELISA plates were coated overnight at 4 ℃ with 1 μ g/mL SARS-Cov-2 RBD protein (Acro Biosystems, DE, USA). The plates were washed 3 times and then blocked with 5% Bovine Serum Albumin (BSA) in PBS (0.05% Tween 20, PBST buffer) for 1 hour at 37 ℃. Three serial dilutions of mouse serum were added to each well and incubated for 1 hour at 37 ℃. The plates were washed five times again and then 1: goat anti-mouse IgG-HRP (GenScript, NJ, CN) at 8000 dilutions was incubated for 1 hour, followed by detection of bound antibody. After the final wash, the plates were developed by using TMB substrate and 50 μ l/well 2M H2SO4The reaction was terminated. Reading at 450 nm and 620 nm, determining the endpoint of the serum antibody titer as the reciprocal of the highest dilution, wherein the highest dilution of the sample is 2.1 times higher than the absorbance of a negative control (determination standard: experimental group: control (negative) OD450-620 value ≧ 2.1, and the corresponding highest dilution at the OD value is determined as the serum antibody titer).

And (4) conclusion: as shown in fig. 7 and 8, the test animals were significantly stimulated to produce antigen-specific antibodies by the new crown wild strain pWT, the pb.1.617.2 mutant strain candidate DNA vaccines both at day 14 after the initial immunization and at day 7 after the boost immunization. In the above ELISA test, the SARS-Cov-2 RBD protein of the neocoronary wild-type is used as the in vitro envelope antigen, and the above conditions are all favorable for the nucleic acid vaccine pWT of the neocoronary wild-type, however, the DNA vaccine of pb.1.617.2 provided by the present invention also achieves considerable technical effects, and as mentioned above, pWT is a prior product with excellent immune effect against the wild-type strain of the neocoronary wild-type, thereby further illustrating the excellent immunogenicity and broad spectrum of the vaccine of the present invention.

2. Further evaluation of DNA vaccine elicited antigen-specific cellular responses

We investigated whether DNA vaccines could promote cellular immunity by ELISpot analysis. Splenocytes were isolated 7 days after the booster immunization and subjected to IFN-. gamma.and IL-4 positive cell ELISpot experiments.

2.1 IFN-. gamma.IL-4 ELISpot experiments

On day 7 after boosting, in a sterile environment, mice were euthanized, spleens were removed and ground into single cell suspensions; centrifuging to obtain cells, lysing the red blood cell lysate after resuspension, and stopping lysis by PBS containing FBS; filtering, and counting the prepared single cell suspension; single cells were suspended in RPMI1640 medium supplemented with 10% FBS, 1% penicillin/streptomycin. IL-4 ELISpot, IFN-. gamma.ELISpot assays were performed by using the mouse IL-4, IFN-. gamma.two-color FlouroPot kit (MabTech, USA). Spleen cell suspension of each mouse isolated by the above method was inoculated at a density of 250,000 to each well coated with an anti-IL-4 antibody, an anti-IFN-. gamma.antibody, and CO at 37 deg.C2The incubators were stimulated with SARS-CoV-2 RBD peptide library for 20 hours at a peptide library concentration of 10. mu.g/mL (final concentration) per well (in RPMI + 10% FBS). The operation was performed according to the product instructions. Culture medium and PMA/Iono served as negative and positive controls, respectively. Positive spots were quantified by iSpot Reader (AID, Stra beta berg, Germany). Spot Forming Units (SFU) per million cells were calculated by subtracting negative control wells.

And (4) conclusion: IFN-gamma and IL-4 ELISPOT results are shown in figure 9 and figure 10, and high-level antigen-specific IFN-gamma and IL-4 responses can be effectively induced on the 7 th day after the new crown wild strain pWT and the pB.1.617.2 mutant strain candidate DNA vaccines are boosted. In the ELIspot test, the neocoronal wild-type SARS-Cov-2 RBD protein is used as the in vitro stimulating peptide, and the conditions are all favorable for the neocoronal wild-type nucleic acid vaccine pWT, but the pb.1.617.2 mutant DNA vaccine provided by the present invention also achieves significant technical effects, as mentioned above, pWT is a prior product with excellent immune effect against the neocoronal wild-type strain, thereby further illustrating the good immunogenicity and broad spectrum of the neocoronal pb.1.617.2 mutant DNA vaccine of the present invention.

3. Further evaluation of the effects of the vaccine-elicited antigen-specific cellular immune response, in particular the effects of CD 8T cell function, splenocytes were isolated 7 days after booster immunization and subjected to flow cytometry assays.

Isolation of splenocytes: 7 days after the booster immunization, the procedure was carried out in a sterile environment, the mice were euthanized, the spleens were taken out, and ground into single cell suspensions; centrifuging to obtain cells, lysing the red blood cell lysate after resuspension, and stopping lysis by PBS containing FBS; filtering, and counting the prepared single cell suspension; single cells were suspended in RPMI1640 medium supplemented with 10% FBS, 1% penicillin/streptomycin.

Flow cytometry detection experiment: spleen cell suspension from each mouse obtained by the above method, 37 ℃, 5% CO2Next, the cells were stimulated with SARS-CoV-2 RBD peptide library or PMA/Iono, while blocking with 1. mu.g/ml Breededlin A (BD, CA, USA) for 6 hours. Extracellular and intracellular cytokine staining of splenocytes, stimulated splenocytes were stained with FVD-eFluor780, then washed, and stained with anti-mouse CD4, CD8a antibody, respectively, in the dark at room temperature for 30 minutes. Cells were permeabilized with the fixation/permeation buffer and stained intracellularly with anti-mouse IFN-. gamma.and anti-mouse TNF-. alpha.for 45 minutes at 4 ℃. Cells were washed twice and resuspended in 200 μ L PBS before being harvested using a flow cytometer (ThermoFisher, MA, usa) and analyzed using FlowJo software (BD, CA, usa).

And (4) conclusion: the results are shown in fig. 11, and the candidate DNA vaccine of the new crown wild strain pWT and the pb.1.617.2 mutant strain can significantly induce antigen specificity and generation of CD8IFN γ T cell subset 7 days after the boost immunization. In the FACS test, the Xinguan wild SARS-Cov-2 RBD protein is used as in vitro stimulating peptide, and the conditions are all favorable for the Xinguan wild nucleic acid vaccine pWT, but the pB.1.617.2 mutant DNA vaccine provided by the invention also obtains considerable technical effects, as mentioned above, pWT is a prior product with excellent immune effect, and better immunogenicity and broad spectrum of the pB.1.617.2 mutant DNA vaccine are demonstrated.

In conclusion, it is clear from the results of examples 1-5 that the pB.1.617.2 mutant DNA vaccine of the present invention is capable of efficient transcription and expression not only in mammalian cells; the pB.1.617.2 mutant strain candidate DNA vaccine can remarkably stimulate experimental animals to generate antigen specific antibodies at 14 days after primary immunization and 7 days after boosting immunization for humoral immune response; for cellular immune response, the pB.1.617.2 mutant candidate DNA vaccine can not only induce high-level antigen-specific IFN-gamma and IL-4 response, but also induce generation of antigen-specific CD8IFN gamma T cell subset.

It is worth noting that pWT wild strain vaccine is a product aiming at SARS-Cov-2 wild strain at the early stage of the company, and has entered the clinical stage III at present, and has very excellent immune effect. In the above tests, for example, ELISA, ELIspot and FACS detection, the Xinguan wild type SARS-Cov-2 RBD protein is used as in vitro envelope antigen or stimulating peptide, and the above conditions are all favorable for the Xinguan wild type nucleic acid vaccine pWT, however, the pB.1.617.2 mutant DNA vaccine provided by the invention also has significant technical effect, and better immunogenicity and broad spectrum of the pB.1.617.2 mutant DNA vaccine are demonstrated.

Finally, it should be noted that: the above embodiments are only used to illustrate the technical solution of the present invention, and not to limit the same; while the invention has been described in detail and with reference to the foregoing embodiments, it will be understood by those skilled in the art that: the technical solutions described in the foregoing embodiments may still be modified, or some or all of the technical features may be equivalently replaced; and the modifications or the substitutions do not make the essence of the corresponding technical solutions depart from the scope of the technical solutions of the embodiments of the present invention.

Sequence listing

<110> Amelanchine biopharmaceutical Limited

<120> SARS-COV-2 virus B.1.617.2 mutant strain DNA vaccine and application

<130> 20211119

<160> 3

<170> SIPOSequenceListing 1.0

<210> 1

<211> 3834

<212> DNA

<213> Artificial Sequence (Artificial Sequence)

<400> 1

atggattgga cttggattct ctttctcgtt gctgcagcca cacgcgttca tagctcgcag 60

tgcgtgaacc tgagaacacg gacccagctg cctccagctt acacaaatag cttcaccaga 120

ggcgtgtact acccggacaa ggtgttccgg tcctctgtgc tgcacagcac ccaggacctc 180

ttcctgccct ttttcagcaa cgtgacctgg ttccacgcta tccacgtgtc tggcacaaac 240

ggaaccaaaa gattcgacaa ccccgtgctg cctatcaatg atggagtcta cttcgcctct 300

atcgaaaaga gcaacatcat ccgcggctgg atcttcggca ccaccctgga cagtaagacc 360

cagagcctgc tcatcgtgaa caacgccacg aacgtggtga tcaaggtgtg tgaattccaa 420

ttttgcaacg acccctttct cgacgtgtac taccacaaga acaataaatc ttggatggag 480

agcggcgtgt acagctctgc taacaactgc actttcgagt acgtgtccca gccattcctg 540

atggacctgg aaggcaagca gggcaatttc aagaacctga gagaattcgt gtttaagaac 600

atcgacggct acttcaaaat ctattctaag cacaccccaa tcaacctggt ccgggacctg 660

ccacaaggct tcagcgccct ggaacctctg gtggacctgc ctatcggaat caacatcacc 720

cggttccaga ccctgctggc cctgtaccgg agctacctga cacctggcga cagcagctct 780

ggctggaccg ccggcgctgc cgcatattac gtcggctact tgcaacctag gaccttcctg 840

ctgaaataca acgagaacgg caccatcaca gatgccgttg attgcgccct ggaccccctg 900

agcgaaacca agtgtaccct gaaatccttc accgtggaaa agggcatcta ccagaccagc 960

aactttagag tacagcctac agaatctatc gttcggtttc caaacattac caacctgtgt 1020

cctttcggcg aggtgtttaa cgccacacgg ttcgccagcg tgtatgcctg gaatagaaag 1080

cggatcagca actgtgtggc cgactactcc gtgctgtaca atagcgccag cttctctaca 1140

tttaagtgct acggcgtgtc ccctacaaag ctgaacgacc tgtgcttcac aaacgtgtat 1200

gccgatagct tcgtgatccg gggcgatgag gtccggcaga tcgctcctgg ccagacaggc 1260

aagattgccg actacaacta caagctgccc gatgacttca ccggatgtgt gatagcctgg 1320

aacagcaaca acctggatag caaggtgggc ggcaactaca actaccggta ccgactgttt 1380

agaaagagca acctgaaacc ttttgagcgg gacatcagca cagagatcta ccaagccggc 1440

tctaagcctt gtaacggcgt ggagggcttc aactgttact tccctctgca gtcttacgga 1500

ttccagccta caaacggcgt gggataccag ccctatagag tggtggtgct gtcattcgag 1560

ctgctacatg cccctgccac cgtgtgcggc cctaagaagt ctaccaacct cgtgaagaac 1620

aagtgcgtga attttaactt caatggactg acaggcacag gcgtgctgac agagagcaac 1680

aaaaagttcc tgcccttcca gcagtttggc agagatatcg ctgacaccac agacgccgtg 1740

cgcgatcctc agaccctgga gatcctggac atcacccctt gctcctttgg aggagtgtcc 1800

gtgatcacac ctggaacgaa caccagcaac caggttgccg tgctgtacca gggcgtgaac 1860

tgcacagaag ttcctgtggc catccatgcc gatcagctga cgcccacgtg gcgggtgtac 1920

tctaccggca gcaatgtgtt ccagaccaga gccggctgcc ttattggcgc tgagcacgtg 1980

aataatagct atgaatgcga tatcccaatc ggagccggca tttgcgccag ctaccagacc 2040

cagacaaata gtcggagaag agccagatct gtggcctccc agagcatcat cgcatatacc 2100

atgagcctag gagccgaaaa cagcgtcgcc tattccaaca atagcatcgc catcccgaca 2160

aacttcacca tcagcgtgac caccgaaatc ctgcccgtga gcatgaccaa gacaagcgtg 2220

gactgtacaa tgtacatctg tggagactcc accgagtgca gcaacctgct gctgcagtac 2280

ggcagcttct gcacccagct gaacagagcc ctgacaggga tcgccgtgga acaggataag 2340

aacacccaag aggtgttcgc ccaagtgaag cagatctata agactccacc tattaaggac 2400

tttggcggct tcaacttcag ccaaatcctg cccgatccta gcaagccaag caagcggtcc 2460

ttcatcgagg acctgctgtt caacaaggtg accctggccg acgccggctt catcaagcag 2520

tatggcgact gtctgggcga tatcgccgct agagacctga tctgcgccca gaagttcaat 2580

ggcctgaccg tgctcccacc tctgctcacc gacgagatga tcgcccagta cacctctgcc 2640

ctgctggccg gcaccatcac cagcgggtgg acattcgggg ctggagctgc tctgcaaatc 2700

cccttcgcca tgcagatggc ctacagattc aacggcatcg gcgttaccca gaatgtgctg 2760

tatgaaaacc agaaactgat agctaaccag ttcaacagcg ccataggcaa aatccaggat 2820

agtctgagct ctacagccag cgccctggga aaactgcaga acgtggtgaa tcagaacgcc 2880

caggccctga atacactggt gaaacaactg agcagcaatt tcggcgccat cagcagcgtg 2940

ctgaatgata tcctgtctag actggacccc cccgaggccg aggtgcagat cgatagactg 3000

atcaccggca gactgcagtc cctgcagaca tacgtgactc aacagctgat cagagccgct 3060

gagatcagag cttctgctaa tttggctgcc acaaagatga gcgagtgcgt gctgggccag 3120

agcaaaagag tggacttctg cggcaagggc taccacctga tgagcttccc ccagagcgcc 3180

cctcacggcg tcgtgttcct gcacgtgact tacgtgcctg cccaggagaa gaacttcacc 3240

accgcccctg ccatctgcca cgacggcaag gcccacttcc cccgggaggg cgtgttcgtg 3300

agcaatggca cccactggtt cgtgacccaa agaaactttt acgagcccca gattatcacc 3360

accgacaaca ccttcgtgtc aggcaactgc gacgtggtga tcggcatcgt gaacaacact 3420

gtgtacgacc ctctgcagcc tgagctggac agcttcaagg aggaactgga caagtacttc 3480

aaaaaccaca catctcctga cgtggacctg ggcgatatca gcggcattaa cgcctctgtg 3540

gtgaacatcc agaaggaaat cgacagactg aacgaggtgg ccaagaacct gaatgagagc 3600

ctgatcgacc tgcaggagct gggcaagtac gagcagtaca tcaagtggcc ttggtacatc 3660

tggctgggct ttatcgccgg cctgatcgcc atcgtgatgg tcaccatcat gctgtgctgc 3720

atgaccagct gttgcagctg cctgaaaggc tgttgcagct gcggaagttg ctgcaagttt 3780

gacgaggacg actctgagcc tgtgctgaag ggcgtcaagc tgcactacac atga 3834

<210> 2

<211> 3852

<212> DNA

<213> Artificial Sequence (Artificial Sequence)

<400> 2

atgtggtggc gcctgtggtg gctgctgctg ctgctgctgc tgctgtggcc catggtgtgg 60

gcctctcagt gcgtgaacct gaccaccaga acccagctgc ctcctgctta caccaactcg 120

ttcacacggg gagtgtacta ccccgacaag gtgttcagga gctcagtgct gcatagcacc 180

caagacctgt tcctgccatt cttcagcaac gtcacgtggt tccacgccat ccacgtgtct 240

ggaaccaacg gcaccaagag attcgacaac cccgtgctgc ctttcaacga tggagtgtac 300

ttcgctagca ccgagaagag caacatcatc cggggctgga tcttcggcac cacactggac 360

tccaagacac agagtctgct gatcgtgaac aacgccacca acgtcgtgat caaggtgtgt 420

gagttccagt tctgcaacga tcctttcctc ggcgtttact accacaagaa caacaagagc 480

tggatggaat cagaatttag ggtatattct tctgccaata actgtacgtt tgaatacgtg 540

tctcagcctt tcctaatgga cctggaaggc aaacagggca actttaagaa cctgagagaa 600

ttcgtgttta agaacatcga cggctatttc aagatctaca gtaagcacac ccctatcaac 660

ctggtgcggg acctgcccca ggggttttcc gcccttgaac ctctggtgga cctgcccatt 720

ggcatcaata tcacaagatt ccagaccctg ctggccctgc acagaagcta cctgacccct 780

ggcgacagca gcagcggatg gaccgccggc gccgccgcct actacgtggg atacctgcag 840

cctagaacct tcctactgaa atacaacgaa aacggtacca tcaccgacgc cgtggattgc 900

gctctggacc ctctgagcga aaccaagtgc accctgaaaa gctttaccgt ggagaagggc 960

atttatcaga caagcaactt tcgggtgcag cctaccgaga gcatcgtgag attccctaac 1020

atcaccaacc tgtgtccttt cggcgaggtg ttcaatgcca cacggttcgc cagcgtgtac 1080

gcctggaacc ggaagcggat cagcaactgc gtggccgact acagcgtgct gtataatagc 1140

gccagcttca gcacattcaa gtgctacggc gtgagcccca ccaagctgaa tgatctgtgc 1200

tttaccaacg tgtatgccga tagctttgtg atccgggggg acgaggtaag acagattgcc 1260

ccaggacaga caggcaaaat cgcagattac aactacaaac tgcctgacga cttcaccggc 1320

tgcgttatcg cctggaactc caacaacctg gacagcaagg tgggaggaaa ctacaactac 1380

ctgtaccgac tgttcagaaa gagcaacctg aagccattcg agagagatat ttcgacagag 1440

atctaccagg ccggaagcac accttgcaac ggcgtggaag gcttcaactg ctacttcccc 1500

ctgcagagct acggctttca gcccacaaac ggcgtcggct accagcctta cagagtggtg 1560

gtgctgagct tcgagctgct gcatgcccct gccaccgtgt gcgggcctaa gaagtccaca 1620

aatctggtaa agaataagtg tgtgaacttc aatttcaatg gcctgaccgg aacgggtgtg 1680

ctgaccgaat ctaataagaa gttcctgcct ttccagcagt tcggccgtga tatcgccgac 1740

accaccgacg ctgtccgcga tcctcaaacc ctggaaatcc tggacattac accttgcagc 1800

ttcggcggcg tgtccgtgat cacaccaggc acaaacacca gcaaccaggt ggctgtgctg 1860

taccaggacg tgaactgtac agaggtgcct gtggccatcc acgccgacca gctgacacct 1920

acatggagag tgtattcaac aggcagcaac gtcttccaga ccagagcagg atgcctgatc 1980

ggcgctgagc atgtgaacaa ctcctacgag tgcgacatcc ctatcggcgc cggcatctgc 2040

gctagttacc agactcaaac caactctcct cggcgggcta gaagcgtcgc ctcccagagc 2100

atcatcgctt ataccatgtc tctgggcgcc gagaacagcg tggcctacag caacaactcc 2160

atcgccattc ctaccaactt cacgatctca gttaccaccg agatcctgcc tgtgagcatg 2220

acaaagacca gcgtcgactg caccatgtac atctgcggcg attccacaga atgctccaac 2280

ctgctgctcc agtacggctc tttctgtacc cagctgaaca gagccctgac aggcatcgcc 2340

gtggaacagg ataagaacac tcaggaggtg ttcgcccagg tgaagcagat ctacaagacc 2400

cctccaatca aggactttgg cggctttaat ttcagccaaa tcctcccaga tcctagcaag 2460

cccagcaaga gaagcttcat cgaggacctg ctgttcaaca aggtcaccct ggctgacgcc 2520

ggcttcatca agcagtatgg cgactgcctg ggcgatatcg ccgcgaggga tctaatttgt 2580

gctcagaagt tcaacggcct gaccgtgctg ccccccctgc tgacagacga aatgatcgct 2640

cagtacacat ctgccctgct ggccggcacc atcacgagcg gctggacctt cggagccggc 2700

gccgccctgc agatcccctt cgctatgcag atggcctata gattcaacgg catcggcgtg 2760

acccagaacg tgctgtacga gaaccaaaaa ctgattgcca atcaatttaa ttccgcgatc 2820

ggaaagatcc aggactctct gagctctact gccagcgccc tgggcaagct gcaagacgtg 2880

gtgaaccaga atgctcaagc cctgaacacc ctggtgaagc agctgagcag caatttcgga 2940

gcaatcagct ctgtcctcaa cgacattctg tctagactag acaaggtgga agccgaagtg 3000

cagatcgatc ggcttatcac cggaagactg cagagcctgc agacatatgt tacacagcag 3060

ctgatcagag ccgccgagat cagagccagc gccaacctgg cagccacaaa aatgtccgag 3120

tgcgtcctcg gccaatctaa gcgggttgat ttctgtggca aaggctacca cctgatgagc 3180

ttcccccaaa gcgctcctca cggcgtggtg tttctgcacg tcacctacgt gcccgcccaa 3240

gagaagaact tcaccaccgc ccccgctatc tgccacgacg gcaaggccca cttccctcgg 3300

gaaggcgtgt tcgtgagtaa cggtacacac tggtttgtga cccaaagaaa cttctacgag 3360

cctcagatca tcaccaccga taacaccttt gtgagcggca actgcgatgt ggtgatcggc 3420

atcgtgaaca acacagtata cgaccccctg cagcccgagc tggacagctt taaagaggag 3480

ctcgataagt acttcaagaa ccacacatct ccagacgtgg acctgggcga catcagcggc 3540

atcaacgcca gtgttgtgaa catccagaaa gaaatcgata gactgaacga agtggccaag 3600

aatctgaacg agagcctgat cgacctgcag gagctgggca aatacgagca gtacatcaag 3660

tggccttggt acatctggct gggctttatc gccggcctga tcgccattgt gatggtgaca 3720

atcatgctgt gctgtatgac ctcttgctgc tcctgcctga aaggctgttg tagttgcggc 3780

agctgctgta aattcgatga ggatgactcc gagccggtcc tcaaaggcgt caagctgcac 3840

tacacctgat aa 3852

<210> 3

<211> 3831

<212> DNA

<213> Artificial Sequence (Artificial Sequence)

<400> 3

atggattgga cctggattct gtttctggtg gcggcggcga cccgcgtgca tagcagccag 60

tgcgtgaacc tgcgcacccg cacccagctg ccgccggcgt ataccaacag ctttacccgc 120

ggcgtgtatt atccggataa agtgtttcgc agcagcgtgc tgcatagcac ccaggatctg 180

tttctgccgt tttttagcaa cgtgacctgg tttcatgcga ttcatgtgag cggcaccaac 240

ggcaccaaac gctttgataa cccggtgctg ccgattaacg atggcgtgta ttttgcgagc 300

attgaaaaaa gcaacattat tcgcggctgg atttttggca ccaccctgga tagcaaaacc 360

cagagcctgc tgattgtgaa caacgcgacc aacgtggtga ttaaagtgtg cgaatttcag 420

ttttgcaacg atccgtttct ggatgtgtat tatcataaaa acaacaaaag ctggatggaa 480

agcggcgtgt atagcagcgc gaacaactgc acctttgaat atgtgagcca gccgtttctg 540

atggatctgg aaggcaaaca gggcaacttt aaaaacctgc gcgaatttgt gtttaaaaac 600

attgatggct attttaaaat ttatagcaaa cataccccga ttaacctggt gcgcgatctg 660

ccgcagggct ttagcgcgct ggaaccgctg gtggatctgc cgattggcat taacattacc 720

cgctttcaga ccctgctggc gctgtatcgc agctatctga ccccgggcga tagcagcagc 780

ggctggaccg cgggcgcggc ggcgtattat gtgggctatc tgcagccgcg cacctttctg 840

ctgaaatata acgaaaacgg caccattacc gatgcggtgg attgcgcgct ggatccgctg 900

agcgaaacca aatgcaccct gaaaagcttt accgtggaaa aaggcattta tcagaccagc 960

aactttcgcg tgcagccgac cgaaagcatt gtgcgctttc cgaacattac caacctgtgc 1020

ccgtttggcg aagtgtttaa cgcgacccgc tttgcgagcg tgtatgcgtg gaaccgcaaa 1080

cgcattagca actgcgtggc ggattatagc gtgctgtata acagcgcgag ctttagcacc 1140

tttaaatgct atggcgtgag cccgaccaaa ctgaacgatc tgtgctttac caacgtgtat 1200

gcggatagct ttgtgattcg cggcgatgaa gtgcgccaga ttgcgccggg ccagaccggc 1260

aaaattgcgg attataacta taaactgccg gatgatttta ccggctgcgt gattgcgtgg 1320

aacagcaaca acctggatag caaagtgggc ggcaactata actatcgcta tcgcctgttt 1380

cgcaaaagca acctgaaacc gtttgaacgc gatattagca ccgaaattta tcaggcgggc 1440

agcaaaccgt gcaacggcgt ggaaggcttt aactgctatt ttccgctgca gagctatggc 1500

tttcagccga ccaacggcgt gggctatcag ccgtatcgcg tggtggtgct gagctttgaa 1560

ctgctgcatg cgccggcgac cgtgtgcggc ccgaaaaaaa gcaccaacct ggtgaaaaac 1620

aaatgcgtga actttaactt taacggcctg accggcaccg gcgtgctgac cgaaagcaac 1680

aaaaaatttc tgccgtttca gcagtttggc cgcgatattg cggataccac cgatgcggtg 1740

cgcgatccgc agaccctgga aattctggat attaccccgt gcagctttgg cggcgtgagc 1800

gtgattaccc cgggcaccaa caccagcaac caggtggcgg tgctgtatca gggcgtgaac 1860

tgcaccgaag tgccggtggc gattcatgcg gatcagctga ccccgacctg gcgcgtgtat 1920

agcaccggca gcaacgtgtt tcagacccgc gcgggctgcc tgattggcgc ggaacatgtg 1980

aacaacagct atgaatgcga tattccgatt ggcgcgggca tttgcgcgag ctatcagacc 2040

cagaccaaca gccgccgccg cgcgcgcagc gtggcgagcc agagcattat tgcgtatacc 2100

atgagcctgg gcgcggaaaa cagcgtggcg tatagcaaca acagcattgc gattccgacc 2160

aactttacca ttagcgtgac caccgaaatt ctgccggtga gcatgaccaa aaccagcgtg 2220

gattgcacca tgtatatttg cggcgatagc accgaatgca gcaacctgct gctgcagtat 2280

ggcagctttt gcacccagct gaaccgcgcg ctgaccggca ttgcggtgga acaggataaa 2340

aacacccagg aagtgtttgc gcaggtgaaa cagatttata aaaccccgcc gattaaagat 2400

tttggcggct ttaactttag ccagattctg ccggatccga gcaaaccgag caaacgcagc 2460

tttattgaag atctgctgtt taacaaagtg accctggcgg atgcgggctt tattaaacag 2520

tatggcgatt gcctgggcga tattgcggcg cgcgatctga tttgcgcgca gaaatttaac 2580

ggcctgaccg tgctgccgcc gctgctgacc gatgaaatga ttgcgcagta taccagcgcg 2640

ctgctggcgg gcaccattac cagcggctgg acctttggcg cgggcgcggc gctgcagatt 2700

ccgtttgcga tgcagatggc gtatcgcttt aacggcattg gcgtgaccca gaacgtgctg 2760

tatgaaaacc agaaactgat tgcgaaccag tttaacagcg cgattggcaa aattcaggat 2820

agcctgagca gcaccgcgag cgcgctgggc aaactgcaga acgtggtgaa ccagaacgcg 2880

caggcgctga acaccctggt gaaacagctg agcagcaact ttggcgcgat tagcagcgtg 2940

ctgaacgata ttctgagccg cctggatccg ccggaagcgg aagtgcagat tgatcgcctg 3000

attaccggcc gcctgcagag cctgcagacc tatgtgaccc agcagctgat tcgcgcggcg 3060

gaaattcgcg cgagcgcgaa cctggcggcg accaaaatga gcgaatgcgt gctgggccag 3120

agcaaacgcg tggatttttg cggcaaaggc tatcatctga tgagctttcc gcagagcgcg 3180

ccgcatggcg tggtgtttct gcatgtgacc tatgtgccgg cgcaggaaaa aaactttacc 3240

accgcgccgg cgatttgcca tgatggcaaa gcgcattttc cgcgcgaagg cgtgtttgtg 3300

agcaacggca cccattggtt tgtgacccag cgcaactttt atgaaccgca gattattacc 3360

accgataaca cctttgtgag cggcaactgc gatgtggtga ttggcattgt gaacaacacc 3420

gtgtatgatc cgctgcagcc ggaactggat agctttaaag aagaactgga taaatatttt 3480

aaaaaccata ccagcccgga tgtggatctg ggcgatatta gcggcattaa cgcgagcgtg 3540

gtgaacattc agaaagaaat tgatcgcctg aacgaagtgg cgaaaaacct gaacgaaagc 3600

ctgattgatc tgcaggaact gggcaaatat gaacagtata ttaaatggcc gtggtatatt 3660

tggctgggct ttattgcggg cctgattgcg attgtgatgg tgaccattat gctgtgctgc 3720

atgaccagct gctgcagctg cctgaaaggc tgctgcagct gcggcagctg ctgcaaattt 3780

gatgaagatg atagcgaacc ggtgctgaaa ggcgtgaaac tgcattatac c 3831

24页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:编码SARS-COV-2病毒C.37突变株抗原的DNA分子、DNA疫苗及应用

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!