CN113563453A - Preparation and application of chicken interleukin 17B recombinant lactobacillus immune preparation - Google Patents

Preparation and application of chicken interleukin 17B recombinant lactobacillus immune preparation Download PDF

Info

Publication number
CN113563453A
CN113563453A CN202110838325.7A CN202110838325A CN113563453A CN 113563453 A CN113563453 A CN 113563453A CN 202110838325 A CN202110838325 A CN 202110838325A CN 113563453 A CN113563453 A CN 113563453A
Authority
CN
China
Prior art keywords
specific
protein
recombinant
ibv
sequence
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202110838325.7A
Other languages
Chinese (zh)
Inventor
高荣
郭绍华
彭俊杰
肖永乐
刘岩岩
杨鑫
王红宁
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.)
Sichuan University
Original Assignee
Sichuan University
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 Sichuan University filed Critical Sichuan University
Priority to CN202110838325.7A priority Critical patent/CN113563453A/en
Publication of CN113563453A publication Critical patent/CN113563453A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/74Vectors or expression systems specially adapted for prokaryotic hosts other than E. coli, e.g. Lactobacillus, Micromonospora
    • C12N15/746Vectors or expression systems specially adapted for prokaryotic hosts other than E. coli, e.g. Lactobacillus, Micromonospora for lactic acid bacteria (Streptococcus; Lactococcus; Lactobacillus; Pediococcus; Enterococcus; Leuconostoc; Propionibacterium; Bifidobacterium; Sporolactobacillus)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55522Cytokines; Lymphokines; Interferons
    • A61K2039/55527Interleukins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/20011Coronaviridae
    • C12N2770/20034Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Abstract

The invention discloses a preparation method and application of a chicken interleukin 17B recombinant lactobacillus immune preparation. The invention provides application of specific protein, which is (a1) or (a 2): (a1) preparing an immunopotentiator; (a2) as an immune enhancer; the specific protein is (b1) or (b2) or (b3) as follows: (b1) protein consisting of amino acid residues from 28 th to 199 th in a sequence 1 of a sequence table; (b2) 1 to 199 amino acid residues in a sequence 1 of a sequence table; (b3) a protein shown in a sequence 1 in a sequence table. The invention also protects the specific protein. The specific protein of the invention can promote lymphocyte proliferation, inhibit the growth of pathogenic microorganisms, promote the secretion of specific antibodies, promote the secretion of mucosal antibodies and improve the immunocompetence and survival rate of animals.

Description

Preparation and application of chicken interleukin 17B recombinant lactobacillus immune preparation
Technical Field
The invention belongs to the technical field of biology, and particularly relates to preparation and application of a chicken interleukin 17B recombinant lactobacillus immune preparation.
Background
Livestock and poultry intensive breeding plants are the main sources of livestock products in the market. With the global population growth, the demand for livestock and poultry products has also increased year by year. The annual chicken consumption worldwide accounts for about 40% of all meat products, with direct economic benefits of hundreds of billions of dollars.
Chickens are usually kept in an intensive environment conducive to infection by opportunistic pathogens. A major problem facing the poultry industry is that due to the reduced productivity caused by disease, large amounts of resources must be utilized to maintain the health of the animals. Vaccines and antibiotics (particularly antibiotics) are mainly used to control diseases. Conventional inactivated vaccines and live attenuated vaccines are not effective in inactivating certain pathogenic bacteria, including conventional IBV vaccines due to their variability, etc. Therefore, novel vaccines such as DNA vaccines, RNA vaccines, and polyepitope vaccines have become hot spots for vaccine research.
However, conventional adjuvants do not significantly enhance the humoral and cellular immunity of these novel vaccines. The most common veterinary vaccine adjuvants currently licensed on the market are aluminium gel adjuvants and oil emulsion adjuvants. Due to their safety, aluminum hydroxide (aluminum) adjuvants have been commonly used in many veterinary and human vaccines; the few cell-mediated immunity, particularly cytotoxic T cell responses, induced by the aluminous gel adjuvant, however, is a significant drawback for its use as a vaccine against intracellular parasites and certain viruses; in addition, the alumina gel adjuvant has a tendency to induce IgE-mediated immune response and may promote IgE-mediated allergic response. The use of oil emulsion adjuvants is limited by the side and adverse effects they produce; for example, the oil emulsion adjuvant CFA can cause inflammation, ulceration, and fever and allergic reactions at the injection site. The appearance quality of poultry meat is very important for poultry breeding industry, and the adverse reactions of vaccine injection points of immunized chickens caused by the aluminum gel adjuvant and the oil emulsion adjuvant can affect the appearance quality of the meat, cause discomfort of animals and are not beneficial to the welfare of the animals. The poultry vaccine industry therefore needs effective adjuvants that meet the need for an adjuvant that will not cause pain to the poultry nor cause a reduction in the quality of the poultry meat. Moreover, the abuse of antibiotics in livestock and poultry farming poses a great threat to the environment and human survival. In china, the government prohibited the addition of antibiotics to feed since 1 month 1 of 2020. Therefore, the additive or the therapeutic drug is urgently needed to be found to replace antibiotics for livestock and poultry breeding. Cytokine (CK) is a low molecular weight soluble protein that is synthesized and secreted by immune cells and some non-immune cells. After the cell factor is combined with the specific receptor, the gene expression of the cell factor network can be activated, and the immune function can be regulated. Thus, cytokines are considered to be highly potent and present only few side effects.
Disclosure of Invention
The invention aims to provide preparation and application of a chicken interleukin 17B recombinant lactobacillus immune preparation.
The invention provides application of specific protein, which is (a1) or (a 2):
(a1) preparing an immunopotentiator;
(a2) as an immune enhancer;
the specific protein is (b1) or (b2) or (b3) as follows:
(b1) protein consisting of amino acid residues from 28 th to 199 th in a sequence 1 of a sequence table;
(b2) 1 to 199 amino acid residues in a sequence 1 of a sequence table;
(b3) a protein shown in a sequence 1 in a sequence table.
The invention also provides an application of the specific recombinant bacterium, which is (a1) or (a 2):
(a1) preparing an immunopotentiator;
(a2) as an immune enhancer;
the specific recombinant strain is a recombinant strain for expressing the specific protein.
The specific recombinant bacteria are obtained by introducing specific DNA molecules or recombinant plasmids with the specific DNA molecules into a spawn-running bacteria; the specific DNA molecule is a DNA molecule for expressing the specific protein.
The starting bacterium can be lactobacillus plantarum.
The starting bacterium can be lactobacillus plantarum NC 8.
The specific recombinant bacterium can be specifically a recombinant lactobacillus plantarum NC8 obtained by introducing a recombinant plasmid pMG36e-rChIL17B into the lactobacillus plantarum NC8, and is named as a recombinant bacterium NC8-ChIL 17B. And replacing small fragments in SalI and SphI enzyme digestion recognition sequences in the pMG36e vector with double-stranded DNA molecules shown in a sequence 2 in a sequence table to obtain a recombinant plasmid pMG36e-rChIL 17B.
The fermentation product of the specific recombinant bacterium or the total thallus protein of the specific recombinant bacterium is applied as follows (a1) or (a 2):
(a1) preparing an immunopotentiator;
(a2) can be used as immunopotentiator.
The invention also provides the application of the vaccine and the immunopotentiator in the preparation of products;
the immunopotentiator is (c1), (c2), (c3) or (c4) as follows:
(c1) the specific protein;
(c2) the specific recombinant bacteria;
(c3) the fermentation product of the specific recombinant bacteria;
(c4) the total thallus protein of the specific recombinant bacteria.
In particular, the vaccine may be an IBV H120 vaccine.
The product has the function of preventing and/or treating infectious bronchitis of chicken.
The invention also protects specific proteins, which are (b1), (b2) or (b 3):
(b1) protein consisting of amino acid residues from 28 th to 199 th in a sequence 1 of a sequence table;
(b2) 1 to 199 amino acid residues in a sequence 1 of a sequence table;
(b3) a protein shown in a sequence 1 in a sequence table.
The invention also protects a specific DNA molecule, an expression cassette containing the specific DNA molecule, a recombinant vector containing the specific DNA molecule or a recombinant microorganism containing the specific DNA molecule; the specific DNA molecule is a DNA molecule for coding the specific protein.
The specific DNA molecule can be specifically a DNA molecule shown in a sequence 2 of a sequence table.
The recombinant vector can be specifically a double-stranded DNA molecule shown in a sequence 2 of a sequence table, which is obtained by replacing small fragments in SalI and SphI enzyme digestion recognition sequences in a pMG36e vector, so as to obtain a recombinant plasmid pMG36e-rChIL 17B.
The recombinant microorganism is obtained by introducing the specific DNA molecule or a recombinant plasmid having the specific DNA molecule into a starting microorganism.
The starting microorganism can be specifically lactobacillus plantarum.
The starting microorganism can be lactobacillus plantarum NC 8.
The recombinant microorganism can be specifically a recombinant lactobacillus plantarum NC8 obtained by introducing a recombinant plasmid pMG36e-rChIL17B into the lactobacillus plantarum NC8, and is named as recombinant bacterium NC8-ChIL 17B.
The invention also protects the fermentation product of the recombinant microorganism.
The invention also protects the total protein of the recombinant microorganism.
The invention also protects the product A. The product A contains the specific protein, the specific DNA molecule, the expression cassette, the recombinant vector, the recombinant microorganism, a fermentation product of the recombinant microorganism or the total protein of the recombinant microorganism. The product is an immunopotentiator.
The invention also protects the product B. The product B contains vaccine and immunopotentiator; the immune enhancer is the specific protein, the specific DNA molecule, the expression cassette, the recombinant vector, the recombinant microorganism, a fermentation product of the recombinant microorganism or the total protein of the recombinant microorganism. In particular, the vaccine may be an IBV H120 vaccine. The product has the function of preventing and/or treating infectious bronchitis of chicken.
The application of the immunopotentiator is specifically to improve the animal immunity. The animal is a non-human animal or a human.
The immune promoter is used for promoting the expression of immune related genes. The immune related gene can be any one or combination of the following genes: NF-kappa B, MyD88, SOCS-1, STAT1, IL-1 beta, IL-12p40, IL-4, IL-6, IL-10, INF-alpha, IFN-beta, TGF-beta 4, CCL-3, CCL-20, and LITAF.
The animal immunity improving effect is at least one of the following M1-M5:
m1, inhibiting the growth of pathogenic microorganisms;
m2, promoting increase of immune cells;
m3, promoting vaccine-induced immune responses;
m4, promoting cellular and/or humoral immunity;
m5, improving animal development and growth weight gain.
The pathogenic microorganism is specifically avian infectious bronchitis virus.
The animal may in particular be an avian species, such as a chicken.
The fusion protein rChIL17B molecule and the fermentation product of the recombinant bacteria expressing the rChIL17B gene provided by the invention have the following functions: promoting the proliferation of animal lymphocyte, the lymphocyte content in the animal body which is applied with rChIL17B can be increased by more than 10 percent; obviously inhibit the growth of pathogenic microorganisms, and the amount of the pathogenic microorganisms applied with rChIL17B can be reduced by more than 20 percent; promoting the secretion of specific antibodies (IgG and IgA) and non-specific sIgA, and increasing the content of specific antibodies and disease-specific antibodies in the animal body to which rChIL17B is applied by 40-60%; promote the expression of immune related genes, further improve the immune anti-infection capacity of animals, obviously improve the virus-attacking survival rate of the animals which are applied with rChIL17B by at least 60 percent, and increase the growth and weight percentage by more than 10 percent. Experiments prove that the rChIL17B can promote lymphocyte proliferation, inhibit the growth of pathogenic microorganisms, promote the secretion of specific antibodies, promote the secretion of mucosal antibodies, and improve the immunocompetence and survival rate of animals.
Drawings
FIG. 1 is a schematic structural diagram of recombinant plasmid pMG36e-rChIL 17B.
FIG. 2 is an electrophoretogram of mRNA level detection in example 1.
FIG. 3 is an electrophoretogram of the protein level detection in example 1.
FIG. 4 shows the results of the fluorescent quantitative PCR in example 2.
FIG. 5 shows the result of step one in example 3.
FIG. 6 shows the result of the measurement of the humoral immune index in example 4 (measured by IBV-specific antibody ELISA kit).
FIG. 7 shows the result of the measurement of the humoral immune index of example 4 (measured by ELISA kit corresponding to the chicken-derived cytokines IL-2, IL-4, IL-6 and IFN-. gamma.).
FIG. 8 shows the results of mucosal immunity index test in example 4.
FIG. 9 shows the results of the measurement of cellular immunity index in example 4 (change of CD4+ T cells and CD8+ T cells in peripheral blood lymphocytes).
FIG. 10 shows the results of the cellular immunity index test (expression change of immunity-related gene) in example 4.
FIG. 11 shows the results of the challenge protection test of example 4.
Detailed Description
The present invention is described in further detail below with reference to specific embodiments, which are given for the purpose of illustration only and are not intended to limit the scope of the invention. The examples provided below serve as a guide for further modifications by a person skilled in the art and do not constitute a limitation of the invention in any way.
The experimental procedures in the following examples, unless otherwise indicated, are conventional and are carried out according to the techniques or conditions described in the literature in the field or according to the instructions of the products. Materials, reagents and the like used in the following examples are commercially available unless otherwise specified. Unless otherwise stated, the quantitative tests in the following examples were performed in triplicate, and the results were averaged.
rChIL17B concentration (protein concentration) was measured using His-Tag ELISA Kit (His Tag ELISA Detection Kit, King Biotech Co., Ltd., Cat. No.: L00436). pMG36e vector: biovector Inc., cat # Biovector MG36 e. HD11 cell (chicken macrophage): shanghai ze leaf Biotech Co., Ltd. DF-1 cells (chicken fibroblasts): wuhan Punuoise Life technologies, Inc.
DMEM growth solution: fetal bovine serum containing 10% of 56 ℃ inactivated complement and 0.1% of a cyan chain double antibody were used in a DMEM basal medium. Maintenance liquid: fetal bovine serum containing 2% of 56 ℃ inactivated complement and 0.1% of a cyan chain double antibody were used in a DMEM basal medium.
Lactobacillus plantarum NC8, described in the following documents: comparing the effect of lactobacillus plantarum NC8 on chick T lymphocytes by nasal drip and oral administration; liuyuyin, Yang waves, Zhao Liang, Yang Guilian, Wang Chun Feng; china veterinary journal 2017 (volume 53), phase 1.
Infectious bronchitis virus B strain (IBV B strain) (IBV Beaudette strain) is described in the following documents: infections bronchis Virus Infection introduction Replication in Chicken macro HD11 Cells; xiaooxino Han, Yiming Tian, Ru Guan, Wenqian Gao, Xin Yang, Long Zhou, honging Wang. [ J ]. Virus, 2017,9(8): 198.
Infectious bronchitis virus M41 strain (IBV M41 strain), described in the following documents: constructing reverse genetic strains of the avian infectious bronchitis virus M41 strain; plum, xu xin, wu dong, ji gao, yangxin and wanghong ning; basic science, Chinese veterinary Abstract, 2015, volume 31, phase 5.
When the expression quantity of the gene is detected by fluorescence quantitative PCR, the reference gene adopted by animal experiments is beta-actin, and the reference gene adopted by cell experiments is GAPDH.
TABLE 1 primer sequences for real-time quantitative PCR
Figure BDA0003177988900000051
Figure BDA0003177988900000061
Example 1 preparation of recombinant bacterium
First, fusion protein rChIL17B and design of coding gene thereof
Chicken interleukin 17B, added signal peptide and His6The label is named as fusion protein rChIL 17B. The fusion protein rChIL17B is shown as a sequence 1 in a sequence table. In the sequence 1 of the sequence table, the 1 st-27 th amino acid residues form a signal peptide, the 28 th-199 th amino acid residues form interleukin 17B, and the 200 th-205 th amino acid residues form His6And (4) a label.
The gene encoding the fusion protein rChIL17B was named rChIL17B gene.
The DNA molecule shown in the sequence 2 of the sequence table codes the protein shown in the sequence 1 of the sequence table. In the sequence 2 of the sequence table, the 1 st to 81 th sites encode signal peptide, the 82 nd to 597 th sites encode interleukin 17B of chicken, the 598 th site and 615 th sites encode His6And (4) a label.
Second, construction of recombinant plasmid
And replacing small fragments in SalI and SphI enzyme digestion recognition sequences in the pMG36e vector with double-stranded DNA molecules shown in a sequence 2 in a sequence table to obtain a recombinant plasmid pMG36e-rChIL 17B. The recombinant plasmid pMG36e-rChIL17B was sequence verified. The structural schematic diagram of the recombinant plasmid pMG36e-rChIL17B is shown in figure 1.
Thirdly, preparing recombinant bacteria
The recombinant plasmid pMG36e-rChIL17B is introduced into lactobacillus plantarum NC8 to obtain recombinant lactobacillus plantarum named as recombinant bacterium NC8-ChIL 17B.
The pMG36e vector is introduced into Lactobacillus plantarum NC8 to obtain recombinant Lactobacillus plantarum named as recombinant bacterium NC 8-P.
Fourth, mRNA level detection
1. Selecting a test bacterium (a recombinant bacterium NC8-ChIL17B or a recombinant bacterium NC8-P or lactobacillus plantarum NC8) single colony, inoculating the single colony to 1mL of liquid MRS culture medium containing 10 mug/mL of erythromycin, and standing and culturing at 37 ℃ overnight; then inoculating the strain to a liquid MRS culture medium containing 10 mug/mL of erythromycin at the inoculation amount of 1%, and standing and culturing for 12-16 h; then, the mixture was centrifuged at 4000g for 10min at 4 ℃ to collect the pellet.
2. mRNA level detection
Taking the thalli obtained in the step 1, extracting RNA, and then carrying out reverse transcription to obtain cDNA; taking cDNA as a template, adopting a primer pair consisting of Pe-F and Pe-R to carry out PCR amplification, and then carrying out agarose gel electrophoresis. The electrophoretogram is shown in FIG. 2, lane M represents Marker, lane 1 represents Lactobacillus plantarum NC8, lane 2 represents recombinant bacteria NC8-P, and lane 3 represents recombinant bacteria NC8-ChIL 17B. Only lane 3 shows the specific amplified band.
Pe-F:ATGAAAAAAAAGATTATCTCAGC;
Pe-R:TTAGTGGTGGTGGTGGTGG。
Fifth, protein level detection
1. Inoculating the activated test bacterium (recombinant bacterium NC8-ChIL17B or recombinant bacterium NC8-P) bacterium liquid to a liquid MRS culture medium containing 10 mug/mL of erythromycin at a volume ratio of 1:100, and performing shaking culture at 37 ℃ and 180rpm for 24 hours; centrifuging at 10000g for 5min, and collecting thallus; then treated with lysozyme (working concentration of lysozyme is 1 mg/ml); then carrying out ultrasonic disruption, centrifuging and collecting supernatant fluid, namely the total protein solution. The total protein solution obtained by the recombinant strain NC8-ChIL17B is named as NC8-rChIL17B protein solution. The total protein solution obtained by the recombinant strain NC8-P is named as NC8-P protein solution.
2. The total protein solution obtained in step 1 was subjected to polyacrylamide gel electrophoresis, followed by Western Blot (the primary antibody used was HIS-tag mouse IgG monoclonal antibody), and the results are shown in FIG. 3. Electrophoresis of NC8-rChIL17B protein solution showed a positive protein band of about 20.17kDa, consistent with the predicted size of rChIL 17B. NC8-P protein solution did not show the positive protein band.
Example 2 biological Activity Studies of the fusion protein rChIL17B on Chicken fibroblast cell line
1. The DF-1 cells grown in a T75 cell flask are dispersed by trypsinization and then uniformly spread on 6-well cellsIn the board (about 1.0X 10 for each board)6Individual cells) were cultured with DMEM growth medium to a confluency of 70% -80%.
2. After completion of step 1, protein dilutions (to give a system protein concentration of 0.5. mu.g/mL, based on total protein provided by the protein dilutions) were added per well and incubation continued for 2h with supplemented maintenance medium. Cells were harvested at 48h exposure (timed from addition of protein diluent). An equal volume of PBS buffer was used as a PBS control group instead of the protein diluent. At least 3 duplicate wells were set for each treatment at each time point.
And diluting the test protein solution by using PBS buffer solution to obtain a protein diluent. The test protein solution was either the NC8-rChIL17B protein solution prepared in step five of example 1 or the NC8-P protein solution prepared in step five of example 1.
3. And (3) taking the cells obtained in the step (2), extracting total RNA, and then carrying out reverse transcription to obtain cDNA. And (3) carrying out fluorescence quantitative PCR by taking the cDNA as a template to detect the expression regulation and control conditions of each immune related gene. The primer sequences for detecting the respective genes are shown in Table 1. The expression level of each gene was calculated by the geometric mean method and the following formula: relative mRNA expression level 2-ΔΔCt
The results of the fluorescent quantitative PCR are shown in FIG. 4. In FIG. 4, PBS represents the PBS control group, NC8-chIL17B represents the NC8-rChIL17B treatment group, and NC8 represents the NC8-P treatment group. Statistical and one-way analysis of variance (ANOVA-Tukey) was performed on the data using GraphPad Prism 6(GraphPad software), with p <0.05, or p < 0.01. Stimulation of DF-1 cells by rChIL17B resulted in significantly higher expression levels of NF-. kappa. B, MyD88, SOCS-1, STAT1, IL-1. beta., IL-12P40, IL-4, IL-6, IL-10, INF-. alpha., IFN-. beta., TGF-. beta.4, CCL-3, CCL-20, and LITAF than in PBS control and NC8-P treated groups (P <0.05 or P < 0.01).
Example 3 Effect of the fusion protein rChIL17B on the proliferation of infectious bronchitis Virus in chickens
First, the effect of fusion protein rChIL17B on the activity of macrophage system of chicken infected by chicken infectious bronchitis virus
1. HD11 cells, which had filled T75 flasks, were dispersed by trypsinization and then inoculated into 96-well plates at approximately 1.0X 10 cells/plate6The individual cells were cultured with DMEM growth medium to 70% -80% confluency.
2. The test protein solution was diluted with PBS buffer to give each protein dilution such that the protein concentration was 800ng/mL, 400ng/mL, or 200ng/mL, respectively. The test protein solution was either the NC8-rChIL17B protein solution prepared in step five of example 1 or the NC8-P protein solution prepared in step five of example 1.
3. After completion of step 1, the cells were washed 2 times with PBS buffer, then IBV B strain (MOI 2) was added and the culture was continued for 1.5h with DMEM growth medium, then the protein dilution obtained in step 2 was added and the culture was continued for 24h (protein concentration in the system was 200ng/ml, 100ng/ml or 50ng/ml, respectively, based on the total protein provided by the protein dilution). An IBV control group was set up with an equal volume of PBS buffer instead of protein diluent.
4. After completion of step 3, 20. mu.L of CCK-8 solution was added to each well and incubation was continued for 0.5-4 hours. Blank control wells were set with no cells added and CCK8 only.
5. After completion of step 4, the absorbance was measured at a wavelength of 450nm with a microplate reader. According to OD450Values calculate the percent inhibition of cells in each well, with lower ratios indicating better cell activity. Percent (%) inhibition of cell activity (PBS control OD)450Experimental group OD450) /(PBS control OD450Blank group OD450)×100。
At least 3 multiple wells were set for each treatment.
The results are shown in FIG. 5. In FIG. 5, IBV represents the IBV control group, NC8-P + IBV represents the NC8-P test group, and NC8-rChIL17B + IBV represents the NC8-rChIL17B test group. The total protein concentration in the system was 200ng/ml (wherein rChIL17B concentration was 220pg/ml) compared to the IBV control group, resulting in a significant reduction in the inhibition of cell activity, with a very significant difference (p < 0.01). The inhibition of cell activity increased with decreasing protein concentration, indicating that the inhibition of IBV by the fusion protein rChIL17B is dose-dependent.
Second, the effect of the fusion protein rChIL17B on the proliferation titer of IBV on HD11 cells
1. The HD11 cells which had filled the T75 flask were digested and dispersed with pancreatin, and the whole wasUniformly inoculated into 6-well plates, approximately 1.0X 10 per plate6The individual cells were cultured with DMEM growth medium to 70% -80% confluency.
2. After completion of step 1, the cells were washed 2 times with PBS buffer, then IBV B strain (MOI 2) was added and the culture was continued for 1.5h with DMEM growth medium, then protein diluent was added and the culture was continued for 24h (protein concentration in the system was 200ng/ml, based on total protein provided by the protein diluent). A PBS control group was set up with an equal volume of PBS buffer instead of IBV strain B. An IBV control group was set up with an equal volume of PBS buffer instead of protein diluent.
And diluting the test protein solution by using PBS buffer solution to obtain a protein diluent. The test protein solution was either the NC8-rChIL17B protein solution prepared in step five of example 1 or the NC8-P protein solution prepared in step five of example 1.
At least 3 multiple wells were set for each treatment.
3. After the step 2 is finished, repeatedly freezing and thawing twice at minus 80 ℃, collecting cells and supernatant after freezing and thawing, namely the virus solution to be tested, and testing TCID50And (4) poison price.
The specific method for determining the toxic price comprises the following steps: virus solution was diluted in 10-fold gradient and seeded into 96-well cell plates full of approximately 70% HD11 cells, 6 replicates per dilution; culturing at 37 deg.C, recording lesion hole number every day, culturing for 72 hr, determining final lesion hole number, and measuring TCID by Reed-Muench method50(ii) a After repeating the measurement 3 times, statistics were performed by One-way ANOVA multiple comparisons.
The results are shown in Table 2. IBV stands for IBV control group, PBS stands for PBS control group, NC8-P + IBV stands for NC8-P test group, NC8-rChIL17B + IBV stands for NC8-rChIL17B test group. TCID of PBS control group50Is 0; TCID of IBV control group506.58, the difference was significant (P + IBV group) compared with NC8-P +<0.05), indicating that NC8-P protein solution can inhibit IBV proliferation on HD11 cells. NC8-rChIL17B + IBV group, total protein concentration in system 200ng/ml (wherein rChIL17B concentration is 220pg/ml), TCID50The values were lower, and the difference was very significant compared to the NC8+ IBV group (p)<0.01), indicating that the fusion protein rChIL17B can obviously inhibit the proliferation of IBV B strain on HD11 cells.
TABLE 2 variation of the proliferative Titers of IBV on HD11 cells under different treatment conditions
Lg TCID50/ml
PBS
0
IBV 6.58±0.11c
NC8-P+IBV 6.08±0.11B
NC8-ChIL17B+IBV 4.65±0.05A
The different capitalized letters show significant differences, p < 0.01; different lower case letters indicate significant difference, p < 0.05.
Example 4 immunomodulating Effect of recombinant bacteria NC8-ChIL17B on avian infectious bronchitis Virus vaccines
Firstly, preparation of NC8-ChIL17B bacterial liquid
Inoculating the activated recombinant strain NC8-ChIL17B into 50ml of liquid MRS culture medium, standing and culturing at 37 ℃ for 24h, then centrifuging at 4000g for 10min, and collecting thalli; the cells were washed 2 times with cold sterile PBS buffer and resuspended in 5X 10 cell concentration in cold sterile PBS buffer9CFU/mL, obtaining NC8-ChIL17B bacterial liquid, and storing on ice for later use.
Secondly, preparing NC8-P bacterial liquid
Inoculating the activated recombinant strain NC8-P into 50ml of liquid MRS culture medium, standing and culturing at 37 ℃ for 24h, then centrifuging at 4000g for 10min, and collecting thalli; the cells were washed 2 times with cold sterile PBS buffer and resuspended in 5X 10 cell concentration in cold sterile PBS buffer9CFU/mL to obtain NC8-P bacterial liquid, and storing on ice for later use.
Grouping and immunizing SPF chickens
SPF chicks were hatched from SPF white leghorn chick embryos purchased from Beijing Burlinger Invrigjon Biotechnology, Inc. The SPF chicks were transferred to SPF isolators for feeding and immunization. IBV vaccines are collectively known as IBV H120 vaccines (IBV H120 oral attenuated vaccines, lyophilisates): sichuan China derivative engineering group, Inc.; the using method comprises the following steps: diluting to 10% with sterile PBS buffer8EID50mL, now ready for use, stored on ice, with a single use dose of 200 μ L per chicken.
The test bacterial liquid is NC8-ChIL17B bacterial liquid prepared in the first step or NC8-P bacterial liquid prepared in the second step.
Immunization procedure: at 7 days of age, simultaneously orally taking test bacteria liquid and IBV H120 vaccine; when the strain is 14 days old, the test bacterial liquid is orally taken; at the age of 21 days, simultaneously orally taking test bacteria liquid and IBV H120 vaccine; at 28 days of age, the test bacterial liquid is orally taken; at the age of 34 days, the test bacterial liquid is orally taken.
The grouping is shown in table 3. Each group of 10 feather SPF chicks is divided into 4 groups equally by adopting a random grouping mode.
TABLE 3 grouping of SPF chickens
Numbering Packet description Immunization dose
A NC8-ChIL17B + IBV group NC8-ChIL17B bacterial liquid (1X 10)9CFU)/feather/time +200 μ L IBV H120 vaccine/feather/time
B NC8-P + IBV group NC8-P bacterial liquid (1X 10)9CFU)/feather/time +200 μ L IBV H120 vaccine/feather/time
C1 IBV control group 200 μ L PBS buffer/feather/time +200 μ L IBV vaccine/feather/time
C2 PBS control group 200 μ L PBS buffer/feather/min
Evaluation was performed according to the index in table 4.
TABLE 4 evaluation index of immunopotentiating effect
Figure BDA0003177988900000111
Fourth, evaluation results of respective indices
1. Change in weight gain
Net weight gain was calculated from time 0 body weight (body weight before immunization on day of prime) and 28 days after prime. The results are shown in Table 5. Chicks in the NC8-ChIL17B + IBV group gained more weight than chicks in the NC8-P + IBV group 28 days after priming (P < 0.0001).
Table 5 average weight gain statistics for immunized 28d chickens (n 10)
Grouping Initial body weight (g) Final body weight (g) Net weight gain (g)
PBS control group 62.0±0.761b 360.0±5.791 298.0±2.631c
IBV control group 61.0±0.955b 350.0±5.924 294.0±3.166c
NC8-P + IBV group 67.0±0.494a 375.0±4.292 309.0±2.252b
NC8-IL17B + group IBV 66.0±0.471a 380.0±7.263 314.0±2.974a
2. Humoral immunity index
Blood was collected through the infrawinged vein at time 0 (day before immunization of the prime) and 7, 14, 21 and 28 days after the prime, left to stand at 37 ℃ for 1h, and then left overnight at 4 ℃ and centrifuged to take serum, which was frozen at-20 ℃.
The detection was carried out using an IBV-specific antibody ELISA detection kit (IDEXX, cat. No. 99-09262). The results are shown in FIG. 6. The results were statistically calculated as the average S/P value + s.d for each group. The threshold value is 0.2. S/P ═ sample a (650) -NC a (650) ]/[ sample a (650) -NC a (650) ]. Tukey's multiple comparison test for statistics; p <0.05, p < 0.01. The S/P values of the IBV specific antibodies of the IBV control group, NC8-P + IBV group and NC8-ChIL17B + IBV group are all greater than 0.2 starting on day 7 after the first immunization, which indicates that the IBV vaccine induces the body to produce the antibodies. During the immunization period, the IBV-specific antibody titer of the NC8-ChIL17B + IBV group was significantly higher than that of the IBV control group and the NC8-P + IBV group, with P <0.05 at days 7 and 28 and P <0.01 at days 14 and 21. IBV-specific antibody levels were highest 14 days after first immunization.
The concentration changes of the cytokines IL-2, IL-4, IL-6 and IFN-gamma in serum samples at the time 0 (before the day of immunization of the prime) and at the days 14 and 28 after the prime are detected by ELISA kits corresponding to the chicken-derived cytokines IL-2, IL-4, IL-6 and IFN-gamma (Shanghai prefers Biotechnology Co., Ltd., IL-2 is YX-091202C, IL-4 is YX-091204C, IL-6 is YX-091206C and IFN-gamma is YX-090617C). The results are shown in FIG. 7. In fig. 7, concentrations are shown as mean + s.d for each group; p <0.05, p < 0.01. At time 0, there was no significant difference in the concentrations of IL-2, IL-4, IL-6 and IFN- γ for each group. At 14 and 28 days post prime: the IL-2 concentration of NC8-ChIL17B + IBV group was significantly higher than that of the IBV control group and NC8-P + IBV group (P < 0.05); the IL-2 concentrations in the IBV control group and NC8-P + IBV group were also significantly higher than in the PBS control group. In addition, the NC8-ChIL17B group had significantly elevated IL-4, IL-6 and IFN- γ concentrations at 14 and 28 days post priming compared to the IBV control, NC8-P + IBV and PBS control groups.
3. Index of mucosal immunity
28 days after first immunization, some experimental chickens were killed by taking blood from the heart, respectively taking fresh trachea and intestinal tract (containing Peyers' patch) each 8cm, placing on ice, repeatedly irrigating with 1mL of cold PBS buffer solution containing protease inhibitor, centrifuging at 4 ℃ and 10000g for 10min, and collecting supernatant, namely the lavage solution containing mucosal antibody. Storing at-20 deg.C for use.
And (3) detecting the content of the total sIgA antibody in the lavage fluid by adopting a chicken sIgA antibody detection kit (based on a direct sandwich ELISA principle).
The method for detecting the titer of the IBV specific mucosal antibody sIgA comprises the following steps: adding the obtained lavage fluid into an ELISA plate (Shanghai preferably Biotech Co., Ltd., product number YX-090814C) coated with IBV specific antigen (each well is 100 mu L), repeating 3 wells for each sample, incubating at 37 ℃ for 1h, washing for 5 times, drying for the last time, adding HRP-labeled goat anti-chicken IgA antibody (each well is 100 mu L) diluted at 1:2000, incubating at 37 ℃ for 1h, washing for 5 times, drying, adding substrate developing solution, incubating at room temperature for 10min, and reading absorbance OD at 630nm wavelength with an ELISA reader630
The results are shown in FIG. 8. In fig. 8, total sIgA concentrations (a, b), IBV-specific sIgA titers (c, d); mucosal antibody concentrations and titers are shown as mean + s.d for each group; statistics were multiplexed by Tukey, # p <0.05, # p < 0.01. In the trachea, the total sIgA and IBV specific sIgA of NC8-ChIL17B + IBV group are significantly higher than those of the IBV control group and NC8-P + IBV group. In the intestinal tract, the total sIgA and the IBV specific sIgA of the NC8-ChIL17B + IBV group are obviously higher than those of the control group. There was no significant difference in tracheal and intestinal total sIgA and IBV-specific sIgA concentrations between the IBV control group and NC8-P + IBV group.
4. Cellular immunity index
(1) Changes in CD4+ T cells and CD8+ T cells in peripheral blood lymphocytes
After priming, 14d and 28d, respectively, were treated with a 1: 9 the injection syringe added with anticoagulant sodium citrate is used for collecting the blood of the infrawing veins of experimental chickens, the mixture is placed on ice after being mixed evenly, and the Peripheral Blood Lymphocytes (PBLC) are separated by adopting a chicken peripheral blood lymphocyte separation kit of TBD company.
Peripheral blood lymphocytes were taken and analyzed by FACScan flow cytometry (the experimental data obtained were analyzed by flowjo7.6.1 software) to obtain the content of CD4+ T cells and CD8+ T cells in peripheral blood lymphocytes. The results are shown in FIG. 9. In fig. 9: (a) the percentage of experimental chicken peripheral blood CD4+ T cells, (b) the percentage of experimental chicken peripheral blood CD8+ T cells; (c) CD4+/CD8+ ratio; carrying out statistics on Tukey's multiple comparison; p <0.05, p < 0.01. Chicks of the NC 8-chll 17B + IBV group were significantly upregulated in both CD4+ T cells and CD8+ T cells (P <0.001) compared to the IBV control group and NC8-P + IBV group at 14 and 28 days post-priming.
(2) Change of expression of immune-related Gene in tissues of immunized Chicken (QPCR)
On day 28 after prime, 4 experimental chickens were sacrificed by heart blood sampling, the state of the organs and tissues of the chickens was observed to evaluate safety, and Thymus (Thymus) and Cecum tonsil (Cecum tonsil) were immediately frozen in liquid nitrogen.
0.1g of tissue was taken and added to 1mL of precooled PBS buffer, and after grinding and homogenization, 200. mu.L of tissue homogenate was added to Trizol solution, RNA was extracted, and then reverse transcription was performed to obtain cDNA. And (3) carrying out real-time quantitative PCR by taking the cDNA as a template so as to analyze the immunoregulation effect of the test bacteria on the IBV vaccine at the cellular immunity level.
In FIG. 10, Tukey' multiple comparisons are used for statistics; p <0.05, p < 0.01. The results show that the expression of the cytokines IL-1 beta, IL-6 and BCL-6 of the NC8-ChIL17B + IBV group is significantly higher than that of the other groups, with p <0.05 or p < 0.01. The expression of IL-22 is of great significance for the formation of mucosal barriers. In the thymus, IL-22 was significantly elevated (P <0.05) in NC8-ChIL17B + IBV, NC8-P + IBV and IBV control groups compared to PBS control group, but there was no significant difference between the three groups. In the caecum tonsils, the expression of NC8-ChIL17B + IBV group IL-22 was significantly higher than that of the 3 control groups (p < 0.05). The expression levels of TLR-3 and TLR-7 in thymus and caecum tonsils of NC8-ChIL17B + IBV group were significantly higher than those of the other 3 control groups (p <0.05 or p < 0.01). The expression of the cytokine TGF-beta 4 involved in maintaining immune balance in the NC8-ChIL17B + IBV group was also significantly higher than in the three control groups (thymic p <0.05, caecal tonsil p < 0.01). Compared with a control group, the NC8-ChIL17B + IBV group memory CD8+ T cell marker CD127 is obviously up-regulated (thymic p is less than 0.01, and cecum tonsil p is less than 0.05), and the thymic gene expression fold is higher than that of the cecum tonsil.
5. Challenge protection test
At 35 days of age, strain IBV M41 was challenged. The immunized chicken flocks are weighed and then treated with IBV M41 strain by nasal drip, eye drip and mouth dripThe medicine is taken to counteract toxic substance (one third of the volume is administered in nose, one third of the volume is administered in eye, and one third of the volume is administered orally), and the dose of counteracting toxic substance is 1mL EID per feather50Is 105.8IBV M41 strain (1). The conditions of the chicken flocks including the conditions of food intake and drinking and whether IBV infection symptoms such as cough, asthma and the like exist are observed in the morning and at night every day. After 10 days of virus challenge, weighing all chickens, killing all chickens, observing whether the IBV target organs have histopathological changes or not, and collecting tissue samples. Tissue samples included lung, liver, spleen, kidney, bursa of Fabricius, and Tracheal Lymph Nodes (TLNs).
Immediately after tissue collection, the tissue was frozen in liquid nitrogen, and 0.1g of tissue sample was added to 0.5. mu.L of cold PBS buffer and homogenized. 200. mu.L of homogenate was added to 800. mu.L of Trizol solution to extract RNA, and cDNA was obtained by reverse transcription. Taking 1 mu L of cDNA (one tenth of total RNA) as a template, simultaneously finding a gene segment with more base difference between the IBV M41 strain and the vaccine IBV H120 according to genome sequences of IBV M41(GenBank accession number AY851295.1) and IBV H120(GenBank accession number FJ888351.1), designing primers capable of identifying M41 and H120 according to the difference gene segment, and using the primers for RT-QPCR. Primer PIBV-F/PIBV-R is biosynthesized by Chengdu Onystoloni.
PIBV-F 5’-ACGGATTCAGGTGATGCTGAGG-3’;
PIBV-R 5'-TTTGTTACTTGCCGGGTCTTGTGT-3'。
The standard plasmid is obtained by inserting a DNA molecule shown in a sequence 3 of a sequence table into a pMD18-T vector. Standard curve equations were prepared using standard plasmids. The standard curve equation is that y is 3.2771x +37.947, E is 102.0%, R2=0.9971。
The IBV M41 viral load in each tissue was measured and six replicates were run for each sample.
Statistics were performed using multiple t-tests, p <0.05, p < 0.01.
The results are shown in FIG. 11. The mean IBV M41 copy number of the immunized groups IBV, NC8-P + IBV and NC8-ChIL17B + IBV was significantly reduced compared to the PBS control group (P < 0.01). The average IBV M41 copy number of the trachea, the lung, the spleen, the kidney and the bursa of Fabricius in the NC8-ChIL17B + IBV group is extremely remarkably or extremely remarkably reduced compared with that of a control group.
Under challenge with virulent IBV strains, the PBS control group of chickens developed significant cough and asthma in the morning. However, no significant symptoms were observed in the IBV group, the NC8-P + IBV group and the NC8-ChIL17B + IBV group.
The present invention has been described in detail above. It will be apparent to those skilled in the art that the invention can be practiced in a wide range of equivalent parameters, concentrations, and conditions without departing from the spirit and scope of the invention and without undue experimentation. While the invention has been described with reference to specific embodiments, it will be appreciated that the invention can be further modified. In general, this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure as come within known or customary practice within the art to which the invention pertains. The use of some of the essential features is possible within the scope of the claims attached below.
Sequence listing
<110> Sichuan university
<120> preparation and application of chicken interleukin 17B recombinant lactobacillus immune preparation
<130> GNCYX212278
<160> 3
<170> SIPOSequenceListing 1.0
<210> 1
<211> 205
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 1
Met Lys Lys Lys Ile Ile Ser Ala Ile Leu Met Ser Thr Val Ile Leu
1 5 10 15
Ser Ala Ala Ala Pro Leu Ser Gly Val Tyr Ala Met Val Leu Val Pro
20 25 30
Glu Ala Lys Asp Glu Ser Arg Ala Ala Lys Gly Arg Arg Arg Gly Pro
35 40 45
Ser Arg Leu Pro Thr Ala Ala Ser Ala Leu Ala Trp Ala Pro Asp Asp
50 55 60
Pro Tyr Thr Pro Met Glu Asp Tyr Glu Arg Ser Ile Gln Asp Met Val
65 70 75 80
His Gln Leu Arg Asn Gly Ser Glu Pro Gly Asp Thr Lys Cys Gln Val
85 90 95
Asn Leu Arg Leu Trp Arg Ser Asn Arg Arg Ser Leu Ser Pro Trp Ala
100 105 110
Tyr Ser Ile Asn His Asp Ala Thr Arg Ile Pro Ala Asp Ile Pro Glu
115 120 125
Ala Arg Cys Leu Cys Thr Gly Cys Ile Asn Pro Phe Thr Met Gln Glu
130 135 140
Asp Arg Thr Met Ala Ser Ile Pro Ile Tyr Ser Arg Leu Pro Val Arg
145 150 155 160
Arg Leu Leu Cys Gln Ala Pro Gly Lys Val Gly His Lys Ala Ser Gly
165 170 175
Arg Lys Lys Cys His Lys Lys Tyr Gln Thr Val Met Glu Thr Ile Ala
180 185 190
Val Gly Cys Thr Cys Ile Phe His His His His His His
195 200 205
<210> 2
<211> 618
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 2
atgaaaaaaa agattatctc agctatttta atgtctacag tgatactttc tgctgcagcc 60
ccgttgtcag gtgtttacgc tatggtttta gttccagaag ccaaggatga aagccgtgca 120
gccaagggtc gtcgtcgtgg cccttcacgg ttaccaacgg ccgcttcagc cttagcctgg 180
gccccagatg acccatacac tccaatggaa gactacgaac gcagcatcca agacatggtt 240
caccaattac gtaatggctc agaaccaggc gacaccaagt gccaagtcaa cttacggtta 300
tggcgttcaa accgccgtag cttatcacca tgggcctaca gcatcaacca cgatgccacg 360
cggatcccag cagacatccc agaagcccgg tgcttatgca ctggctgcat caacccattc 420
accatgcaag aagaccgcac catggccagc atcccaatct acagccggtt acctgtccgc 480
cgtttattat gccaagctcc aggcaaggtt ggtcacaagg cctcaggtcg taagaagtgt 540
cacaagaagt accaaacggt catggaaacc atcgctgttg gctgcacttg catcttccac 600
caccaccacc accactaa 618
<210> 3
<211> 403
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 3
cgatgatgac atcgaggaag aagatgcaga agaatgtgac acggattcag gtgatgctga 60
ggagtgtgac actaatttag aatgtgaaga agaagatgag gatactaaag tgttggctct 120
tatacaagac ccggcaagta acaaatatcc tctgcctctt gatgatgatt atagcgtcta 180
taatggatgt attgttcata aggacgctct cgatgttgtg aatttaccat ctggtgaaga 240
aacctttgtt gtcaataact gctttgaagg ggctgttaaa gctcttccgc agaaagttat 300
tgatgttcta ggtgactggg gtgaggctgt tgatgcgcaa gaacaattgt gtcaacaaga 360
atcaactcgg gtcatatctg agaaatcagt tgagggtttt act 403

Claims (10)

1. The specific protein is used as (a1) or (a 2):
(a1) preparing an immunopotentiator;
(a2) as an immune enhancer;
the specific protein is (b1) or (b2) or (b3) as follows:
(b1) protein consisting of amino acid residues from 28 th to 199 th in a sequence 1 of a sequence table;
(b2) 1 to 199 amino acid residues in a sequence 1 of a sequence table;
(b3) a protein shown in a sequence 1 in a sequence table.
2. The specific recombinant bacteria are applied as follows (a1) or (a 2):
(a1) preparing an immunopotentiator;
(a2) as an immune enhancer;
the specific recombinant bacterium is a recombinant bacterium for expressing the specific protein in claim 1.
3. Use according to claim 2, characterized in that:
the specific recombinant bacteria are obtained by introducing specific DNA molecules or recombinant plasmids with the specific DNA molecules into a spawn-running bacteria; the specific DNA molecule is a DNA molecule expressing the specific protein described in claim 1.
4. The use of the fermentation product of the specific recombinant bacterium of claim 2 or 3 or the total protein of the recombinant bacterium of claim 2 or 3 as follows (a1) or (a 2):
(a1) preparing an immunopotentiator;
(a2) can be used as immunopotentiator.
5. The application of vaccine and immunopotentiator in preparing product;
the immunopotentiator is (c1), (c2), (c3) or (c4) as follows:
(c1) the specific protein of claim 1;
(c2) the specific recombinant bacterium according to claim 2 or 3;
(c3) a fermentation product of the specific recombinant bacterium described in claim 2 or 3;
(c4) total protein of a recombinant bacterium according to claim 2 or 3.
6. The use of claim 5, wherein:
the vaccine is IBV H120 vaccine;
the product has the function of preventing and/or treating infectious bronchitis of chicken.
7. The specific protein is (b1) or (b2) or (b3) as follows:
(b1) protein consisting of amino acid residues from 28 th to 199 th in a sequence 1 of a sequence table;
(b2) 1 to 199 amino acid residues in a sequence 1 of a sequence table;
(b3) a protein shown in a sequence 1 in a sequence table.
8. A specific DNA molecule, an expression cassette containing the specific DNA molecule, a recombinant vector containing the specific DNA molecule, or a recombinant microorganism containing the specific DNA molecule; the specific DNA molecule is a DNA molecule encoding the specific protein according to claim 7.
9. A fermentation product or total protein of the recombinant microorganism of claim 8.
10. Product A or product B;
the product A contains the specific protein of claim 7, the specific DNA molecule of claim 8, the expression cassette of claim 8, the recombinant vector of claim 8, the recombinant microorganism of claim 8, the fermentation product of the recombinant microorganism of claim 8, or the total protein of the recombinant microorganism of claim 8;
the product B contains vaccine and immunopotentiator; the immune enhancing agent is the specific protein of claim 7, the specific DNA molecule of claim 8, the expression cassette of claim 8, the recombinant vector of claim 8, the recombinant microorganism of claim 8, the fermentation product of the recombinant microorganism of claim 8, or the total protein of the recombinant microorganism of claim 8.
CN202110838325.7A 2021-07-23 2021-07-23 Preparation and application of chicken interleukin 17B recombinant lactobacillus immune preparation Pending CN113563453A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110838325.7A CN113563453A (en) 2021-07-23 2021-07-23 Preparation and application of chicken interleukin 17B recombinant lactobacillus immune preparation

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110838325.7A CN113563453A (en) 2021-07-23 2021-07-23 Preparation and application of chicken interleukin 17B recombinant lactobacillus immune preparation

Publications (1)

Publication Number Publication Date
CN113563453A true CN113563453A (en) 2021-10-29

Family

ID=78166891

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110838325.7A Pending CN113563453A (en) 2021-07-23 2021-07-23 Preparation and application of chicken interleukin 17B recombinant lactobacillus immune preparation

Country Status (1)

Country Link
CN (1) CN113563453A (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102648002A (en) * 2009-10-10 2012-08-22 斯坦福大学托管董事会 IL-17 family cytokine compositions and uses
JP2016047051A (en) * 1999-12-23 2016-04-07 ジェネンテック, インコーポレイテッド Il-17 homologous polypeptides and therapeutic uses thereof
CN107362358A (en) * 2017-08-03 2017-11-21 中国水产科学研究院黑龙江水产研究所 Application of the IL 17A albumen in animal vaccine adjuvant is prepared
WO2019178487A2 (en) * 2018-03-15 2019-09-19 Evelo Biosciences, Inc. Compositions and methods for treating disease using klebsiella quasipneumoniae subsp. similipneumoniae

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2016047051A (en) * 1999-12-23 2016-04-07 ジェネンテック, インコーポレイテッド Il-17 homologous polypeptides and therapeutic uses thereof
CN102648002A (en) * 2009-10-10 2012-08-22 斯坦福大学托管董事会 IL-17 family cytokine compositions and uses
CN107362358A (en) * 2017-08-03 2017-11-21 中国水产科学研究院黑龙江水产研究所 Application of the IL 17A albumen in animal vaccine adjuvant is prepared
WO2019178487A2 (en) * 2018-03-15 2019-09-19 Evelo Biosciences, Inc. Compositions and methods for treating disease using klebsiella quasipneumoniae subsp. similipneumoniae

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
NCBI: "interleukin-17B [Gallus gallus]", 《GENBANK DATABASE》 *
NCBI: "PREDICTED: Gallus gallus interleukin 17B (IL17B), mRNA", 《GENBANK DATABASE》 *
QINGLI BIE 等: "IL-17B: A new area of study in the IL-17 family", 《MOLECULAR IMMUNOLOGY》 *
SHAOHUA GUO 等: "The Construction and Immunoadjuvant Activities of the Oral Interleukin-17B Expressed by Lactobacillus plantarum NC8 Strain in the Infectious Bronchitis Virus Vaccination of Chickens", 《VACCINES (BASEL)》 *
WEGMANN,U. 等: "Secreted 45 kDa protein precursor [Lactococcus cremoris subsp. cremoris MG1363]", 《GENBANK DATABASE》 *
楼曜宪 等: "IL-17作为分子佐剂增强蛋白疫苗细胞免疫应答的研究", 《中国生物工程杂志》 *

Similar Documents

Publication Publication Date Title
US8945585B2 (en) Multi-Target recombination gene and the application of its protein to prevent and cure Helicobacter pylori
CN101522210A (en) Compositions and methods of enhancing immune responses
CN101918440A (en) New avian cytokines and its genetic sequence of encoding
CN101289666B (en) Method for preparing recombination porcine alpha-type interferon
CN114015660B (en) Construction of ten-gene deleted attenuated African swine fever virus strain and application thereof as vaccine
CN111840530A (en) Preparation method of Eimeria tenella recombinant polypeptide vaccine VNQS and application method thereof in chicken coccidiosis resistance
EP3200820B1 (en) Broad-spectrum vaccine against avian reovirus
CN109735504B (en) Canine distemper virus attenuated vaccine strain and application thereof
CN113563453A (en) Preparation and application of chicken interleukin 17B recombinant lactobacillus immune preparation
CN111840529B (en) Preparation of recombinant polypeptide vaccine VKVQ of Eimeria tenella and application method of recombinant polypeptide vaccine VKVQ in resisting chicken coccidiosis
CN111471107B (en) Anti-coccidiosis multivalent recombinant protein yolk antibody and preparation method and application thereof
Marcano et al. Evaluation of chickens infected with a recombinant virulent NDV clone expressing chicken IL4
KR102239927B1 (en) Vaccine Composition Containing Inactivated Bovine Rotavirus
CN112370524A (en) Antiviral composition and preparation method and application thereof
CN113135986A (en) Recombinant polypeptide and vaccine for preventing and treating eimeria necatrix
CN117402260A (en) Preparation and application of chicken interleukin 2, 17B and 26 recombinant lactobacillus immune preparation
CN110092840A (en) A kind of infectious laryngotracheitis of chicken, egg drop syndrome bigeminy polyepitope vaccines
KR20200061508A (en) An attenuated avian metapneumovirus and a vaccine composition including the same
CN105349498B (en) A kind of method that optimization prepares inactivated vaccine and/or attenuated live vaccine
Yu et al. Tissue distribution and developmental changes of interferon regulatory factors in chickens and effects of infectious bursal disease virus infection
US20230190909A1 (en) Infectious bronchitis (ib) virus variants and related compositions, uses and method
CN116606366B (en) Grouper interleukin 15 and preparation method and application thereof
US20220257710A1 (en) Anti-infection effects of hnrnpa2b1 and use thereof
CN108129570B (en) Preparation and application of fusion bovine antibacterial peptide and interleukin 2 co-expression recombinant yeast preparation
KR101104911B1 (en) Avirulent infectious bursal disease virus and use thereof as a vaccine

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
WD01 Invention patent application deemed withdrawn after publication

Application publication date: 20211029

WD01 Invention patent application deemed withdrawn after publication