CN113105537B - Host protein for promoting replication of influenza A virus and application thereof - Google Patents

Host protein for promoting replication of influenza A virus and application thereof Download PDF

Info

Publication number
CN113105537B
CN113105537B CN202110418462.5A CN202110418462A CN113105537B CN 113105537 B CN113105537 B CN 113105537B CN 202110418462 A CN202110418462 A CN 202110418462A CN 113105537 B CN113105537 B CN 113105537B
Authority
CN
China
Prior art keywords
virus
influenza
ser
arg
leu
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202110418462.5A
Other languages
Chinese (zh)
Other versions
CN113105537A (en
Inventor
刘月月
黄中利
殷斌
吴家强
李桂明
林树乾
杨世发
赵增成
祝钰玮
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.)
Shandong Yiyuan Pharmaceutical Co ltd
Poultry Research Institute Shandong Academy of Agricultural Sciences
Original Assignee
Shandong Yiyuan Pharmaceutical Co ltd
Poultry Research Institute Shandong Academy of Agricultural Sciences
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shandong Yiyuan Pharmaceutical Co ltd, Poultry Research Institute Shandong Academy of Agricultural Sciences filed Critical Shandong Yiyuan Pharmaceutical Co ltd
Priority to CN202110418462.5A priority Critical patent/CN113105537B/en
Publication of CN113105537A publication Critical patent/CN113105537A/en
Application granted granted Critical
Publication of CN113105537B publication Critical patent/CN113105537B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

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
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • 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
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • 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
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1138Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against receptors or cell surface proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0684Cells of the urinary tract or kidneys
    • C12N5/0686Kidney cells
    • 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
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • 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
    • C12N2510/00Genetically modified cells
    • 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
    • C12N2510/00Genetically modified cells
    • C12N2510/02Cells for production
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16151Methods of production or purification of viral material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/106Plasmid DNA for vertebrates
    • C12N2800/107Plasmid DNA for vertebrates for mammalian
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Abstract

The invention provides a host protein for promoting the replication of influenza A virus, and the amino acid sequence of the host protein is shown as SEQ ID NO. 1. The host protein can be used for proliferating the influenza A virus strain or preparing a medicament for preventing and treating the influenza A virus strain as a target spot, and has important significance in the aspect of developing new medicaments.

Description

Host protein for promoting replication of influenza A virus and application thereof
Technical Field
The invention belongs to the field of molecular biology and immunology, and particularly relates to an application of a host protein immunoglobulin-like receptor ILDR1 in promoting the expression of an influenza virus NP protein and the proliferation of an influenza virus.
Background
Influenza A virus once causes a plurality of outbreaks of influenza in human history, becomes one of main viruses seriously threatening human health, and has important significance for public health safety in prevention and treatment. Because influenza viruses belong to RNA viruses with single negative strand segments, novel viruses can be continuously generated through high mutation rate and gene rearrangement, so that the current vaccine immunization and the drug development taking influenza virus proteins as drug targets are relatively delayed. The emergence of drug-resistant strains of influenza is further accelerated by the extensive use of drugs directed against viral targets. Therefore, the development of new mechanism of action anti-influenza drugs is particularly urgent.
The life cycle of influenza viruses includes adsorption, penetration, peeling of the virus, synthesis of mRNA, synthesis of protein, replication of vRNA, assembly and release of viral particles. The various stages of the replication cycle require the assistance of host proteins. Host factors involved in viral replication are relatively invariant compared to viral proteins, and some of the host factors necessary for viral replication are not critical to the host cell.
The influenza a virus ribonucleoprotein (vRNP) complex is the structural basis for transcription and replication of viral RNA and plays an important role in the process of viral infection of cells. In the early stage of virus infection, the vRNP compound is released into cytoplasm of host cells after endocytosis and membrane removal, and enters into nucleus under the action of nuclear localization signal and transport protein, thus promoting virus transcription and replication. vRNP includes RNA-dependent RNA polymerase complex (RdRp) and nucleoprotein (NP protein), where NP protein is a specific functional protein of influenza virus that can serve as a target for specifically blocking influenza virus replication. It is now found that its entry into the nucleus is regulated by host proteins, and a series of host proteins regulating its entry into the nucleus have been reported. Such as alpha-actin-4, CRM1, UAP56, Hsp40 and MOV10, play an important role in promoting or inhibiting the replication process of viruses. The discovery of new interacting proteins will help provide new targets for the control of influenza viruses.
Disclosure of Invention
Aiming at the problems that the vaccine protection effect is reduced and ineffective easily caused by influenza virus variation, the invention provides a novel protein which can act on influenza virus proliferation, is derived from a host, can interact with influenza A virus, and can promote the proliferation of the influenza A virus.
In order to achieve the purpose, the invention adopts the following technical scheme.
A host protein for promoting the replication of influenza A virus has an amino acid sequence shown in SEQ ID NO. 1.
A nucleotide sequence of the host protein. Preferably, the nucleotide sequence of the host protein is shown in SEQ ID NO. 2.
A recombinant vector, an engineering bacterium and a cell line containing the host protein nucleic acid sequence. The recombinant vector is selected from plasmids. The engineering bacteria are selected from escherichia coli. The cell line is selected from COS7 cell line, HEK293T cell line, A549 cell line and MDCK cell line.
An application of the host protein in the propagation of the influenza A virus strain.
An interfering RNA of the above host protein. The interfering RNA is double-stranded siRNA, and the sequence of the interfering RNA is shown as SEQ ID NO. 3 or SEQ ID NO. 4.
An application of the siRNA in preparing a medicament for preventing and treating influenza A.
The invention has the following advantages:
the host protein provided by the invention can regulate and control the replication of the influenza A virus, can be used for improving the virus amplification amount in a scientific research process, can be used for researching and developing influenza A medicines by taking the host protein as a target, and has important significance in the aspect of new medicine research and development. The interfering RNA provided by the invention can obviously reduce the expression level of host protein for promoting the replication of influenza A virus, thereby obviously inhibiting the replication of influenza A virus.
Drawings
FIG. 1 shows the expression difference of ILDR1 at different times after mice are infected with H1N1 virus;
FIG. 2 shows the H1N1 virus after infection of HEK293T cellsIldr1Change in expression level of (3);
FIG. 3 shows the localization of ILDR1-GFP in COS7 cells by immunofluorescence assay;
FIG. 4 is a Western blot of protein expression by ILDR1-GFP in COS7 cells;
FIG. 5 shows the expression level of H1N1 virus after overexpression of ILDR 1;
FIG. 6 shows the expression level of H1N1 virus after knockdown of ILDR 1;
FIG. 7 shows the expression level of H9N2 virus after overexpression of ILDR 1;
FIG. 8 shows the expression level of H9N2 virus after knockdown of ILDR 1.
Detailed Description
The present invention will be further described with reference to the following examples and drawings, but the present invention is not limited to the following examples.
Example 1 obtaining of ILDR1 Gene
H1N1 strain (isolated from a pig farm with a certain disease in Qingdao Shandong) is infected into 42-day-old C57BL/6J mice, and lungs infected with experimental groups and control groups at different time points are collected for high-throughput sequencing. The bioinformatics analysis finds that: the expression level of ILDR1 was significantly increased compared to the control group. Total protein was extracted from the lungs immediately after 0, 1, 3, 5, 7 and 14 days of infection, respectively, and Western blot analysis was performed, whereby it was found that the level of ILDR1 was significantly increased by 15-fold or more on the first day after infection, the expression level began to decrease on 7 days as the viral load decreased, and the normal level was substantially restored on 14 days (FIG. 1).
HEK293T cells were infected with H1N1 SIV at different MOI (MOI =0, 0.01, 0.1, 1, 10), harvested 24H after infection and detected by qRT-PCRIldr1Change in expression (FIG. 2A). Similarly, HEK293T cells were infected with MOI = 1H 1N1 SIV, harvested 0H, 6H, 12H, 24H, 48H, 72H post infection, and detected using qRT-PCRIldr1Change in expression (FIG. 2B). The results show that ILDR1 mRNA expression following SIV infection is significantly increased in a dose and time dependent manner.
Referring to a mouse genome reference sequence on an NCBI website, designing a cloning primer of a full-length CDS region of a mouse ILDR1 gene according to a transcriptome splicing sequence, introducing Ecor1 enzyme cutting sites and Sal1 enzyme cutting sites (underlined) into an upstream primer and a downstream primer respectively, and leading the sequences to be as follows:
Figure DEST_PATH_IMAGE001
using mouse lung tissue cDNA as a template to carry out PCR amplification, wherein an amplification system is as follows:
Figure 369244DEST_PATH_IMAGE002
the amplification reaction was as follows:
Figure DEST_PATH_IMAGE003
the product of about 1600bp is obtained by amplification, the product is connected to pEGPFN2 plasmid after being recovered by glue, the sequencing is carried out on the positive plasmid (ILDR 1-GFP plasmid), the sequence of the target gene is shown as SEQ ID NO. 2, the total code is 538 amino acids, and the sequence of the amino acids is shown as SEQ ID NO. 1.
Example 2 expression of ILDR1 protein
The COS7 cell line was transfected with the ILDR1-GFP plasmid obtained in example 1 and pEGPFN2 empty vector plasmid not linked to the target gene using Lip2000 liposome, and the group treated with only the transfection reagent and without any plasmid was also used as a negative control group. The transfection efficiency was determined by observing the green fluorescence under the microscope 24h after transfection. As a result, the transfection efficiency of ILDR1-GFP reached 50% or more, as shown in FIG. 3. The above cells transfected with ILDR1-GFP and pEGPFN2 vectors were cultured in CO 2 Culturing in an incubator for 24h, extracting total protein of cells, and performing Western blot detection by using Anti-GFP tagged antibodies, wherein the result is shown in figure 4, and the ILDR1-GFP can be effectively and highly expressed in COS7 cells.
Example 3 Effect of regulation of ILDR1 protein amount on the proliferation of influenza A Virus
1. Over-expressionIldr1Promoting influenza virus replication
HEK with good growth state293T cells at 2X 10 5 Inoculating into 6-well plate at density of one/mL, transfecting ILDR1-GFP and pEGPFN2 vector the next day, infecting virus with MOI =0.1 24h later, collecting cell supernatant 24h later after inoculation, and performing TCID 50 And viral RNA detection. The results are shown in FIG. 5: overexpression compared to negative control groupIldr1Rear endThe expression level of influenza virus is increased.
2. Knock-downIldr1Inhibiting influenza virus replication
According to the general principle of si-RNA design, siRNA targets were designed and the following sequences were synthesized:
Figure DEST_PATH_IMAGE005
the si-ILDR1 and NC described above were transfected into HEK293T cells in CO 2 After culturing for 48h in an incubator, extracting total RNA, and detecting the knockout efficiency by using Q-PCR. The results show that: both pairs of si-RNAs can be knocked downIldr1Above 50%.
HEK293T cells in good growth state were cultured at 2X 10 5 one/mL density was inoculated in 6-well plates, si-ILDR1 and NC were transfected the next day, virus with MOI =0.1 was infected 24h later, cell supernatants were collected 48h later for virus RNA detection. The results are shown in FIG. 6: knockdown compared to negative control group (NC group)Ildr1Thereafter, replication of influenza virus can be inhibited.
Example 4 Effect of modulation of ILDR1 protein amount on the proliferation of H9N2 influenza Virus
Referring to the method in example 3, HEK293T cells with good growth status were transfected with ILDR1-GFP and pegfn 2 vectors, infected with H9N2 virus with MOI =0.1 24H later, and cell supernatants were collected 24H later for virus RNA detection. The results are shown in FIG. 7: overexpression compared to negative control groupIldr1Rear endThe expression level of the H9N2 influenza virus is obviously increased by more than 6 times. si-ILDR1 and NC were transfected 24h before infection with virus with MOI =0.1, cell supernatants were collected 48h after inoculation, and viral RNA was detected. The results are shown in FIG. 8: knockdown compared to negative control group (NC group)Ildr1After that, the replication of the H9N2 influenza virus can be obviously inhibited.
Sequence listing
<110> poultry institute of academy of agricultural sciences of Shandong province
<120> host protein for promoting replication of influenza A virus and application thereof
<130> 20210409
<160> 7
<170> PatentIn version 3.5
<210> 1
<211> 537
<212> PRT
<213> Mus musculus
<400> 1
Met Gly Cys Gly Leu Leu Ala Ala Gly Leu Leu Leu Phe Thr Trp Leu
1 5 10 15
Pro Ala Gly Cys Leu Ser Leu Leu Val Thr Val Gln His Thr Glu Arg
20 25 30
Tyr Val Thr Leu Phe Ala Ser Val Thr Leu Lys Cys Asp Tyr Thr Thr
35 40 45
Ser Ala Gln Leu Gln Asp Val Val Val Thr Trp Arg Phe Lys Ser Phe
50 55 60
Cys Lys Asp Pro Ile Phe Asp Tyr Phe Ser Ala Ser Tyr Gln Ala Ala
65 70 75 80
Leu Ser Leu Gly Gln Asp Pro Ser Asn Asp Cys Ser Asp Asn Gln Arg
85 90 95
Glu Val Arg Ile Val Ala Gln Arg Arg Gly Gln Ser Glu Pro Val Leu
100 105 110
Gly Val Asp Tyr Arg Gln Arg Lys Ile Thr Ile Gln Asn Arg Ala Asp
115 120 125
Leu Val Ile Asn Glu Val Met Trp Trp Asp His Gly Val Tyr Tyr Cys
130 135 140
Thr Ile Glu Ala Pro Gly Asp Thr Ser Gly Asp Pro Asp Lys Glu Val
145 150 155 160
Lys Leu Ile Val Leu His Trp Leu Thr Val Ile Phe Ile Ile Leu Gly
165 170 175
Ala Leu Leu Leu Leu Leu Leu Ile Gly Val Cys Trp Cys Gln Cys Cys
180 185 190
Pro Gln Tyr Cys Cys Cys Tyr Ile Arg Cys Pro Cys Cys Pro Thr Arg
195 200 205
Cys Cys Cys Pro Glu Glu Ala Leu Ala Arg His Arg Tyr Met Lys Gln
210 215 220
Val Gln Ala Leu Gly Pro Gln Met Met Glu Lys Pro Leu Tyr Trp Gly
225 230 235 240
Ala Asp Arg Ser Ser Gln Val Ser Ser Tyr Ala Met Asn Pro Leu Leu
245 250 255
Gln Arg Asp Leu Ser Leu Gln Ser Ser Leu Pro Gln Met Pro Met Thr
260 265 270
Gln Met Ala Ala His Pro Pro Val Ala Asn Gly Val Leu Glu Tyr Leu
275 280 285
Glu Lys Glu Leu Arg Asn Leu Asn Pro Ala Gln Pro Leu Pro Ala Asp
290 295 300
Leu Arg Ala Lys Ser Gly His Pro Cys Ser Met Leu Ser Ser Leu Gly
305 310 315 320
Ser Ala Glu Val Val Glu Arg Arg Val Ile His Leu Pro Pro Leu Ile
325 330 335
Arg Asp Pro Pro Ser Ser Arg Thr Ser Asn Pro Ser His Gln Gln Arg
340 345 350
Leu Asn Ala Val Ser Ser Arg His Cys Asp Leu Ser Glu Arg Pro Arg
355 360 365
Gln Arg His His Ser Asp Phe Leu Arg Glu Leu Gln Asp Gln Gly Met
370 375 380
Arg Pro Trp Ala Pro Gly Arg Gly Glu Leu Asp Pro His Trp Ser Gly
385 390 395 400
Arg His His Arg Ser Arg Pro Ser Glu Ser Ser Met Pro Trp Ser Asp
405 410 415
Trp Asp Ser Leu Ser Glu Cys Pro Ser Ser Ser Glu Ala Pro Trp Pro
420 425 430
Pro Arg Arg Pro Glu Pro Arg Glu Gly Ala Gln Arg Arg Glu Arg Arg
435 440 445
Arg His Arg Ser Tyr Ser Pro Pro Leu Pro Ser Gly Pro Ser Ser Trp
450 455 460
Ser Ser Glu Glu Glu Lys Glu Ser Leu Pro Arg Asn Trp Gly Ala Gln
465 470 475 480
Arg Arg His His His Arg Arg Arg Arg Ser Gln Ser Pro Asn Trp Pro
485 490 495
Glu Glu Lys Pro Pro Ser Tyr Arg Ser Leu Asp Val Thr Pro Gly Lys
500 505 510
Asn Asn Arg Lys Lys Gly Asn Val Glu Arg Arg Leu Glu Arg Glu Ser
515 520 525
Ser His Ser Gly Arg Ser Val Val Ile
530 535
<210> 2
<211> 1614
<212> DNA
<213> Mus musculus
<400> 2
atgggctgcg gattgctcgc tgctggcctg ctcctcttca cctggctccc agcagggtgt 60
ctgtccttgc tagtcacagt ccagcacaca gaacgctatg ttactctgtt tgcctccgtt 120
accctcaagt gtgactacac cacctctgcc cagctccagg acgtggttgt gacatggcgc 180
ttcaagtcct tctgcaagga tcccatcttt gactacttct ctgcctcata ccaggcagct 240
ttgtccctgg gccaggaccc ctccaatgac tgtagtgaca atcagaggga agttcgcatc 300
gtggcgcagc ggcgtgggca gagtgagccc gtgctggggg tggattaccg gcaacgcaag 360
atcaccatcc agaaccgagc agatcttgtg attaatgaag tgatgtggtg ggatcatgga 420
gtatactatt gtaccatcga ggctccagga gacacgtcag gagacccaga taaggaggtg 480
aagctcattg tcctgcattg gctgacagtg attttcatca ttcttggagc cctcctactc 540
ctgctgctga ttggtgtatg ctggtgccag tgttgtccgc agtattgctg ctgctatatc 600
cgctgcccct gctgtcctac ccgctgttgc tgccctgagg aagccctggc ccgccaccgc 660
tacatgaagc aggttcaggc cctaggtcct cagatgatgg aaaaacccct gtactggggg 720
gcggacagga gctcccaagt ttcatcttat gcaatgaacc cgctgctgca gcgagatctg 780
tccttacagt ccagccttcc acagatgcca atgacccaga tggctgctca ccctccggtg 840
gctaatggtg tcctggaata tttggagaaa gaattgcgga acctcaaccc agcccaacct 900
ctgcctgcgg atctcagagc caaatctggc cacccttgca gcatgctctc ctccctgggc 960
tccgcagagg ttgtggaacg cagagtcatc cacctgcccc cactaatcag agacccaccg 1020
tcctccagga ccagcaaccc ctcacaccag cagcggctca atgctgtttc ttccagacac 1080
tgcgatctga gtgagcgccc gaggcagcgc catcactccg atttcctccg agagctccag 1140
gaccagggga tgagaccctg ggccccgggg agaggggagc tggaccccca ttggagtggg 1200
agacaccacc gctctaggcc cagcgagtca tccatgcctt ggtcagactg ggacagcctg 1260
agcgaatgtc cctcatccag tgaggctcct tggcccccca gacgaccaga gcccagggaa 1320
ggcgcccaga gacgtgagag acgcaggcat cgcagctact cgcctcctct accctcgggc 1380
cccagctctt ggagctctga agaggagaaa gagtcgctgc ccaggaactg gggtgcccag 1440
cgacgtcacc atcaccgccg ccgccgctca cagtctccaa actggcctga ggagaagccg 1500
cccagctacc gctcactgga tgtgactcca ggcaagaaca acaggaaaaa agggaatgtg 1560
gagaggcgct tggagagaga gagctcccat agtggacgga gtgtggtcat ttag 1614
<210> 3
<211> 42
<212> DNA
<213> Artificial Sequence
<220>
<223> si-ILDR1-1
<400> 3
cugcaaggac ccuaucuuut taaagauagg guccuugcag tt 42
<210> 4
<211> 42
<212> DNA
<213> Artificial Sequence
<220>
<223> si-ILDR1-2
<400> 4
gaguuggacc caucguggat tuccacgaug gguccaacuc tt 42
<210> 5
<211> 42
<212> DNA
<213> Artificial Sequence
<220>
<223> NC
<400> 5
uucuccgaac gugucacgut tacgugacac guucggagaa tt 42
<210> 6
<211> 27
<212> DNA
<213> Artificial Sequence
<220>
<223> 1F
<400> 6
aatgaattca tgggctgcgg attgctc 27
<210> 7
<211> 28
<212> DNA
<213> Artificial Sequence
<220>
<223> 1R
<400> 7
attccgtcga ctaatgacca ctccgtcc 28

Claims (4)

1. The application of a host protein in propagating influenza A virus strains is characterized in that the amino acid sequence of the host protein is shown as SEQ ID NO. 1.
2. The use according to claim 1, wherein the nucleotide sequence of the host protein is as shown in SEQ ID NO 2.
3. The application of a host protein in preparing a medicament for treating influenza A is characterized in that interfering RNA of the host protein is prepared into the medicament, and the amino acid sequence of the host protein is shown as SEQ ID NO. 1.
4. The use of claim 3, wherein the interfering RNA is a double-stranded siRNA having the sequence shown in SEQ ID NO. 3 or SEQ ID NO. 4.
CN202110418462.5A 2021-04-19 2021-04-19 Host protein for promoting replication of influenza A virus and application thereof Active CN113105537B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110418462.5A CN113105537B (en) 2021-04-19 2021-04-19 Host protein for promoting replication of influenza A virus and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110418462.5A CN113105537B (en) 2021-04-19 2021-04-19 Host protein for promoting replication of influenza A virus and application thereof

Publications (2)

Publication Number Publication Date
CN113105537A CN113105537A (en) 2021-07-13
CN113105537B true CN113105537B (en) 2022-08-23

Family

ID=76718371

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110418462.5A Active CN113105537B (en) 2021-04-19 2021-04-19 Host protein for promoting replication of influenza A virus and application thereof

Country Status (1)

Country Link
CN (1) CN113105537B (en)

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2104737B1 (en) * 2006-12-08 2013-04-10 Asuragen, INC. Functions and targets of let-7 micro rnas
EP2159280A4 (en) * 2007-05-15 2010-08-11 Univ Tsukuba Replication/transcription system for influenza virus genome using yeast cell
CN107441480A (en) * 2010-06-30 2017-12-08 卡姆普根有限公司 Polypeptide and its purposes as the medicine for treating multiple sclerosis, rheumatoid arthritis and other autoimmune disorders

Also Published As

Publication number Publication date
CN113105537A (en) 2021-07-13

Similar Documents

Publication Publication Date Title
US20230287401A1 (en) Rna guided compositions for preventing and treating hepatitis b virus infections
Hale et al. The multifunctional NS1 protein of influenza A viruses
US20130245096A1 (en) COMPOSITIONS AND METHODS FOR ACTIVATING EXPRESSION BY A SPECIFIC ENDOGENOUS miRNA
CN109415728A (en) The excision of retroviral nucleic acid sequence
EA037359B1 (en) Methods and compositions for rna-guided treatment of hiv infection
KR20050084607A (en) Influenza therapeutic
US11597948B2 (en) Use of ANP32 protein in maintaining the polymerase activity of influenza virus in hosts
CN110066769B (en) Avian cells for improved virus production
CA3170630A1 (en) On demand expression of exogenous factors in lymphocytes to treat hiv
Zhang et al. miR-146a promotes Borna disease virus 1 replication through IRAK1/TRAF6/NF-κB signaling pathway
CN113105537B (en) Host protein for promoting replication of influenza A virus and application thereof
AU2013298632B2 (en) Production of infectious influenza viruses
US20230310555A1 (en) Compositions for genome editing and methods of use thereof
CN107096015A (en) The antivirus action of novel antiviral molecule Kdm6a a kind of and its application
CN109022437B (en) Target site sequence for inhibiting goat parainfluenza virus type 3 replication and application thereof
CN112266912A (en) gRNA of target miR-29b, AAV8-CRISPR/Cas9 system and application thereof
WO2018168586A1 (en) Borna viral vector and utilization thereof
Ramirez-Carvajal et al. Down-regulation of viral replication by lentiviral-mediated expression of short-hairpin RNAs against vesicular stomatitis virus ribonuclear complex genes
Wang et al. Effectiveness of lentivirus-mediated RNA interference targeting mouse tumor necrosis factor α in vitro and in vivo
CN108517335A (en) A kind of Lentiviral and its construction method of liver cell miR-199b low expressions
WO2021212892A1 (en) Broad-spectrum antiviral drug and use thereof
CN107586778B (en) shRNA sequence for inhibiting influenza A virus replication and application thereof
CN117562981A (en) SNAPIN protein and application of coding gene thereof in resisting influenza virus
US20190336617A1 (en) CRISPRs IN SERIES TREATMENT
Ahmad et al. Recent patents involving virus nucleotide sequences; host defense, RNA silencing and expression vector strategies

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
TA01 Transfer of patent application right
TA01 Transfer of patent application right

Effective date of registration: 20220729

Address after: 250023 No. 202, Gongye North Road, Jinan, Shandong Province

Applicant after: Poultry Research Institute of Shandong Academy of Agricultural Sciences

Applicant after: Shandong Yiyuan Pharmaceutical Co.,Ltd.

Address before: 250023 No. 1 Jiao Tong Road, Tianqiao District, Shandong, Ji'nan

Applicant before: Poultry Research Institute of Shandong Academy of Agricultural Sciences

GR01 Patent grant
GR01 Patent grant