CN109022437B - Target site sequence for inhibiting goat parainfluenza virus type 3 replication and application thereof - Google Patents

Target site sequence for inhibiting goat parainfluenza virus type 3 replication and application thereof Download PDF

Info

Publication number
CN109022437B
CN109022437B CN201810862854.9A CN201810862854A CN109022437B CN 109022437 B CN109022437 B CN 109022437B CN 201810862854 A CN201810862854 A CN 201810862854A CN 109022437 B CN109022437 B CN 109022437B
Authority
CN
China
Prior art keywords
parainfluenza virus
mir
bta
virus type
replication
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
CN201810862854.9A
Other languages
Chinese (zh)
Other versions
CN109022437A (en
Inventor
李基棕
钟纯燕
毛立
李文良
郝飞
孙敏
刘茂军
杨蕾蕾
张纹纹
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.)
Jiangsu Academy of Agricultural Sciences
Original Assignee
Jiangsu 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 Jiangsu Academy of Agricultural Sciences filed Critical Jiangsu Academy of Agricultural Sciences
Priority to CN201810862854.9A priority Critical patent/CN109022437B/en
Publication of CN109022437A publication Critical patent/CN109022437A/en
Application granted granted Critical
Publication of CN109022437B publication Critical patent/CN109022437B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA 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
    • C12N15/1131Non-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 viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7105Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Virology (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Biochemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Oncology (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Communicable Diseases (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Plant Pathology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Microbiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention relates to a target site sequence for inhibiting replication of goat parainfluenza virus type 3 (CPIV 3) and application thereof in preparation of anti-goat parainfluenza virus type 3 medicines. The target site sequence for inhibiting the 3-type replication of the goat parainfluenza virus is positioned in the 3' UTR of an interferon regulatory gene 2(IRF2), the nucleotide sequence of the target site sequence is shown as SEQ ID NO:19, and the target site sequence is directly targeted by a bta-miR-222 sequence, so that mRNA of IRF2 is degraded, the expression quantity of IRF2 protein is reduced, the expression level of the I-type interferon is improved, and CPIV3 proliferation is inhibited. The target site sequence provides a new drug target for developing new antiviral drugs for treating diseases caused by CPIV 3. In addition, the method can also be applied to the aspects of evaluating bta-miR-222 to regulate the transcription level of IRF2 and breeding anti-goat parainfluenza virus type 3 transgenic sheep.

Description

Target site sequence for inhibiting goat parainfluenza virus type 3 replication and application thereof
Technical Field
The invention belongs to the fields of molecular biology and antiviral science, and particularly relates to a target site sequence for inhibiting goat parainfluenza virus type 3 replication and application thereof in preparation of an anti-goat parainfluenza virus type 3 medicament.
Background
Goat parainfluenza virus type 3 (CPIV 3) is a novel member of the genus Respirovirus of the family Paramyxoviridae, a enveloped, single-stranded negative-strand RNA virus. Members of the genus also include human parainfluenza virus types 1 and 3 (HPIV1, HPIV3), Sendai Virus (SV), and bovine parainfluenza virus type 3 (BPIV 3). The pathogen is detected in goat farms in Jiangsu, Anhui and other places from 2013, a CPIV3 infected sheep can cause respiratory diseases of different degrees, and the homology of the genes with HPIV1 and HPIV3 is only 76.9-83.5 percent as found by RT-PCR amplification sequencing of N, M, F and HN genes. And then, the CPIV3 which is separated and identified in the laboratory is subjected to a goat pathogenicity test, and the result shows that the goat infected by the CPIV3 has clinical symptoms such as cough, rhinorrhea, fever, depression and the like, continuous viremia and toxin expelling are generated, severe pathological damage to the lung and the trachea can be seen through autopsy, and the CPIV3 can be horizontally transmitted through aerosol to enable adjacent healthy goats in the colony house to have respiratory symptoms, detect viremia and expel toxin, thereby bringing a new threat to the healthy development of the sheep industry. Epidemiological investigation in the laboratory shows that the detection rate of the CPIV3 pathogenic nucleic acid reaches 44.7 percent, and the detection rate of the antibody reaches 39.3 percent. Therefore, the hazard of the pathogen to the sheep industry should be paid sufficient attention. However, no report on the drug for treating the disease has been found so far.
Micro ribonucleic acid (miRNA) is a type of endogenous non-coding RNA with the length of about 19-24nt, participates in each link of regulating and controlling cell differentiation, proliferation, metabolism and apoptosis, and is closely related to reproductive development, tumor formation and virus infection process of animal organisms. Mature miRNA is identified and combined by RNA-induced silencing complex (RISC), and is complementarily combined with 5 'UTR, 3' UTR or coding region base of target mRNA, thereby degrading mRNA or inhibiting translation of mRNA into protein, and achieving the effect of regulating function after gene transcription. In recent years, mirnas have been extensively studied in antiviral therapy.
bta-miR-222 is found to be remarkably reduced (7.4 times) in a CPIV3 infected cell sample, but no report is made on the effect of bta-miR-222 in regulating virus proliferation. Based on the above, the bta-miR-222 is analyzed by an overexpression and inhibition expression test, and bta-miR-222 can obviously inhibit the replication of CPIV3 in MDBK cells. Therefore, the invention may provide a new therapeutic method or drug target for treating respiratory diseases caused by CPIV3, which is of great significance.
Disclosure of Invention
The technical problem to be solved by the invention is to provide a target site sequence for inhibiting goat parainfluenza virus type 3 replication and application thereof in preparing a CPIV3 replication-resistant medicament, wherein the sequence is positioned on mRNA of an interferon regulatory gene 2(IRF2) and can be directly targeted by a bta-miR-222 sequence, so that the mRNA of IRF2 is degraded, the expression quantity of IRF2 protein is reduced, the I-type interferon expression level is improved, and CPIV3 proliferation is inhibited.
In order to solve the technical problems, the invention provides a target site sequence for inhibiting the 3-type replication of goat parainfluenza virus, wherein the target site is positioned in the 3' UTR of IRF2, and the nucleotide sequence of the target site is shown as SEQ ID NO. 19.
Preferably, the target site is directly targeted by bta-miR-222 sequence, and the bta-miR-222 nucleotide sequence is shown as SEQ ID NO: 1.
Further, the application of the target site sequence for inhibiting the replication of the goat parainfluenza virus type 3 in preparing a medicament for inhibiting the replication of the goat parainfluenza virus type 3.
bta-miR-222 sequence in the preparation of inhibition goat parainfluenza virus type 3 replication medicine application.
An application of a target site sequence for inhibiting the replication of goat parainfluenza virus type 3 in the preparation of a medicament for treating the goat parainfluenza virus type 3.
Application of bta-miR-222 sequence in preparation of medicine for treating goat parainfluenza virus type 3.
Preferably, the recombinant luciferase reporter vector pGL3-IRF 23' UTR is applied to the aspect of evaluating the transcription level of the bta-miR-222 regulatory interferon regulatory gene 2.
The application of the target site sequence for inhibiting the replication of the goat parainfluenza virus type 3 in breeding transgenic sheep resisting the goat parainfluenza virus type 3.
Compared with the prior art, the invention has the advantages that:
1. the invention provides a target site sequence for inhibiting goat parainfluenza virus type 3 replication, and proves that bta-miR-222 can be combined with the target site sequence to inhibit the goat parainfluenza virus type 3 replication, so that similar reports are not found at home and abroad at present, a new drug target is provided for researching and developing new antiviral drugs for treating diseases caused by CPIV3, and related prompts are provided for researching and developing other antiviral drugs.
2. The bta-miR-222 can inhibit the replication of CPIV3, and because miRNA is generated by the expression of cells, compared with other artificially synthesized medicaments, the miRNA has the advantages of low toxicity to organisms and good effect.
3. The target site sequence for inhibiting the replication of the goat parainfluenza virus 3 is positioned in a 3' non-coding region of an interferon regulatory gene 2, and the interference on the site does not cause obvious damage to receptor cells, so that the safety and feasibility of the anti-goat parainfluenza virus 3 medicament and the cultivation of anti-goat parainfluenza virus 3 transgenic sheep can be improved.
4. The target site sequence for inhibiting the replication of the goat parainfluenza virus type 3 provided by the invention can inhibit the replication of the goat parainfluenza virus type 3 on a molecular level, and can be applied to inhibiting other variants of the goat parainfluenza virus type 3 due to the fact that bta-miR-222 family is widely distributed and highly conserved.
Drawings
For a better illustration of the invention, the following figures are provided, which are provided for illustration of the invention only and are not limiting of the invention:
FIG. 1 shows the inhibitory effect of Bta-miR-222 on CPIV3 proliferation.
FIG. 2 is a schematic representation of base pairing of Bta-miR-222 with its target site.
FIG. 3 shows the regulatory effect of Bta-miR-222 on IRF 2.
FIG. 4 shows Bta-miR-222 regulating the expression changes of IFN-alpha and IFN-beta.
FIG. 5 shows the effect of transfection of si-IRF2 knock-out of the bta-miR-222 target gene on CPIV3 replication.
Detailed Description
The following examples are intended to further illustrate the invention but are not intended to limit it.
Example 1 cell transfection and infection with viruses
(1) Sequence synthesis: bta-miR-222 nucleotide sequence is shown as SEQ ID NO 1; 3 sections of siRNA sequences interfering IRF2, namely Si-IRF2 nucleotide sequences, are respectively shown as SEQ ID NO. 2, SEQ ID NO. 3 and SEQ ID NO. 4. The above-mentioned nucleic acid fragments are each synthesized by Shanghai Jima BioLimited.
(2) Cell transfection: MDBK monolayers were prepared in 24-well plates and, when cells grew to 60% confluence, bta-miR-222 was transfected into MDBK cells, along with a transfection control (NC) set up. The transfection procedure was performed according to Lipofectamine 2000 instructions.
MDBK monolayers were prepared in 24-well plates and 3 fragments of si-IRF2 were transfected into MDBK cells when the cells grew to 60% confluence, along with a transfection control (NC). The transfection procedure was performed according to Lipofectamine 2000 instructions.
(3) Virus inoculation: 24h after transfection, 1000TCID was inoculated per well50CPIV3, after 1.5h, the virus solution was discarded, replaced with 500. mu.L of 2% serum DMEM medium, incubated at 37 ℃ and further inoculated with non-transfected cells as a control. Cell supernatants were harvested at 24h and 48h after inoculation, respectively, and reverse transcription real-time quantitative PCR (RT-qPCR) and TCID were used50The CPIV3 virus content was measured to evaluate the replication capacity of CPIV 3. Both the transfection bta-miR-222 and si-IRF2 were found to be effective in inhibiting CPIV3 proliferation, as shown in FIG. 1 and FIGS. 5C and 5D.
Example 2RT-qPCR technique and TCID50Method for evaluating CPIV3 for its replication capability
Reverse transcription real-time quantitative PCR: total RNA in the cell supernatant was extracted with Transzol UP reagent, and One Step PrimeScript from TaKaRa was usedTMThe RT-PCR Kit detects the content of CPIV3 nucleic acid in the supernatant. Amplification was performed using an ABI Step One fluorescent quantitative PCR instrument, and the reaction system and reaction conditions of RT-qPCR were performed with reference to the kit instructions. The sequences of the upstream primer and the downstream primer used are qCPIV3F shown in SEQ ID NO. 5, qCPIV3R shown in SEQ ID NO. 6 and qCPIV3-probe shown in SEQ ID NO. 7, respectively. The results are shown in FIGS. 1(A) and 5 (C): the transfection bta-miR-222 and si-IRF2 can effectively inhibit the proliferation of CPIV 3.
TCID50And (3) detection: the MDBK cells were seeded into a 96-well plate, the harvested virus solution was diluted 10-fold with a maintenance solution until the cells were grown to 90% confluency, and then the cells were seeded with each of the cell supernatants collected above, 100 μ L per well, 4 wells were repeated, cultured at 37 ℃ for 3d, and then the number of wells in which cytopathic effect occurred was observed for each dilution. Virus TCID calculated according to Reed-Muench method50. The results are shown in FIGS. 1(B) and 5 (D): the transfection bta-miR-222 and si-IRF2 can effectively inhibit the proliferation of CPIV 3.
Example 3Bta-miR-222 target Gene prediction and Dual-luciferase reporter System validation bta-miR-222 target Gene
The bioinformatics software TargetScan is used for predicting the target gene and the binding site of bta-miR-222, and the potential target sequence (IRF 23 'UTR: 977-984bp) of bta-miR-222 exists in the 3' UTR of IRF2 gene, and the complementary site is shown in figure 2. Primers were designed based on the 3' UTR of IRF2 mRNA. The primer sequences are IRF2-3 'UTR-F shown in SEQ ID NO. 8 and IRF 2-3' UTR-R shown in SEQ ID NO. 9 respectively. RT-PCR amplification is carried out, the amplified product is cloned in a luciferase report vector pGL3-control (Promega) through Kpn I and Bgl II enzyme cutting sites, and a recombinant luciferase report vector pGL3-IRF 23' UTR is constructed. Carrying out transfection experiments in three groups, wherein the first group is 293T cells co-transfected by pGL3-IRF 23' UTR, pRL-CMV and bta-miR-222; the second group is 293FT cells co-transfected by pGL3-IRF 23' UTR, pRL-CMV and bta-miR-222-mut (seed site of mutation bta-miR-222); wherein the bta-miR-222-mut nucleotide sequence is shown in SEQ ID NO. 10; the third control group NC was pGL3-IRF 23' UTR, pRL-CMV and NC co-transfected 293FT cells. Changes in luciferase were analyzed using a dual-luciferase reporter assay kit (Promega). A transfection control group (NC) was also established.
Luciferase changes were analyzed using the dual-luciferase reporter assay kit, and the results are shown in FIG. 3A: the luciferase activity of the bta-miR-222 transfected group is reduced by about 55% compared with that of the control group, but the luciferase activity of the bta-miR-222-mut transfected group is not significantly different from that of the control group. The RT-qPCR is used for detecting the transcription level of IRF2mRNA, and the result is shown in figure 3B, the IRF2mRNA amount of the transfected bta-miR-222 group is obviously lower than that of the control group, and is reduced by about 30%. The expression level of the IRF2 protein is analyzed by Western blot, and the result shows that the expression level of the IRF2 protein in the bta-miR-222 group is obviously lower than that in the control group, and the figure is 3C.
Example 4Bta Effect of miR-222 on IRF2 and IFN production
MDBK monolayers were prepared in 24-well plates and, when cells were 60% confluent, bta-miR-222 was transfected into MDBK cells, along with a control of transfected (NC) and a control of non-transfected cells (Mock), according to Lipofectamine 2000. Cells were harvested 24h after transfection. Total RNA from the harvested cells was extracted, and IFN mRNA levels in the cells were detected by reverse transcription and RT-qPCR using EasyScript One-Step gDNA Removal and cDNA Synthesis SuperMix and EasyScript One-Step RT-PCR SuperMix from Tokyo King. And (3) carrying out amplification by using an ABI Step One fluorescence quantitative PCR instrument, wherein the reaction system and the reaction conditions are carried out according to the kit instruction. The IFN expression changes of the transfected miRNA group and the transfected NC control group are calculated by taking GAPDH as an internal reference, and the result is expressed as-delta Ct. The used primer sequences are qIRF2-F shown in SEQ ID NO. 11, qIRF2-R shown in SEQ ID NO. 12, qIFN-alpha-F shown in SEQ ID NO. 13 and qIFN-alpha-R shown in SEQ ID NO. 14 respectively; qIFN-beta-F shown in SEQ ID NO. 15, qIFN-beta-R shown in SEQ ID NO. 16, GAPDH-F shown in SEQ ID NO. 17, and GAPDH-R shown in SEQ ID NO. 18. The result of fluorescent quantitative RT-PCR shows that the average expression levels of IFN-alpha and IFN-beta in cells are remarkably increased after Bta-miR-222 transfects the cells, and are respectively about 1.8 times and 2.1 times, as shown in figure 4.
Example 5 Effect of transfection of si-IRF2 on the bta-miR-222 target Gene
The 3 si-IRF2 described in example 1 were transfected into MDBK cells simultaneously with the establishment of a transfection control (NC), cells were harvested 24h later, and transcription levels of IRF2 gene were measured by RT-qPCR, the results are shown in fig. 5A: after si-IRF2 transfection, the expression level of IRF2 gene is obviously reduced. Western blot detection of the expression level of IRF2 protein showed that the expression level of IRF2 protein was significantly reduced after si-IRF2 transfection, as shown in FIG. 5B. Simultaneously inoculating CPIV3 virus solution after transfecting si-IRF 224 h, harvesting virus solution 24h after inoculation, RT-qPCR technology and TCID50The virus content is detected, and the result shows that the si is transfectedThe IRF2 group had significantly lower CPIV3 virus content than the control group (NC), and the results are shown in FIGS. 5C and 5D.
The foregoing shows and describes the general principles, essential features, and advantages of the invention. It will be understood by those skilled in the art that the present invention is not limited to the embodiments described above, which are described in the specification and illustrated only to illustrate the principle of the present invention, but that various changes and modifications may be made therein without departing from the spirit and scope of the present invention, which fall within the scope of the appended claims. The scope of the invention is defined by the appended claims and equivalents thereof.
Sequence listing
<110> agricultural science and academy of Jiangsu province
<120> target site sequence for inhibiting goat parainfluenza virus type 3 replication and application thereof
<160> 19
<170> SIPOSequenceListing 1.0
<210> 1
<211> 21
<212> RNA
<213> Artificial
<400> 1
agcuacaucu ggcuacuggg u 21
<210> 2
<211> 21
<212> RNA
<213> artificial
<400> 2
gcaaacagua ccucagcaau u 21
<210> 3
<211> 21
<212> RNA
<213> artificial
<400> 3
gcgguccuga cuucaacuau u 21
<210> 4
<211> 21
<212> RNA
<213> artificial
<400> 4
ggcuggaaga acagaucaau u 21
<210> 5
<211> 20
<212> DNA
<213> artificial
<400> 5
gcttggcttc tttgaaatgg 20
<210> 6
<211> 20
<212> DNA
<213> artificial
<400> 6
gcctgcagaa gttccttgtc 20
<210> 7
<211> 27
<212> DNA
<213> artificial
<400> 7
caatcggact agccaagtat ggtggga 27
<210> 8
<211> 32
<212> DNA
<213> artificial
<400> 8
ggggtacccc aagattatgc aaattctcag at 32
<210> 9
<211> 30
<212> DNA
<213> artificial
<400> 9
gaagatcttc gaatctcatg gacagagaag 30
<210> 10
<211> 21
<212> RNA
<213> artificial
<400> 10
acgauguacu ggcuacuggg u 21
<210> 11
<211> 17
<212> DNA
<213> artificial
<400> 11
gccaagttgt ggaggtg 17
<210> 12
<211> 19
<212> DNA
<213> artificial
<400> 12
cacgctatcc gtagtttca 19
<210> 13
<211> 27
<212> DNA
<213> artificial
<400> 13
gtgaggaaat acttccacag actcact 27
<210> 14
<211> 23
<212> DNA
<213> artificial
<400> 14
tgaggaagag aaggctctca tga 23
<210> 15
<211> 22
<212> DNA
<213> artificial
<400> 15
cctgtgcctg atttcatcat ga 22
<210> 16
<211> 22
<212> DNA
<213> artificial
<400> 16
gcaagctgta gctcctggaa ag 22
<210> 17
<211> 20
<212> DNA
<213> artificial
<400> 17
gattgtcagc aatgcctcct 20
<210> 18
<211> 20
<212> DNA
<213> artificial
<400> 18
ggtcataagt ccctccacga 20
<210> 19
<211> 23
<212> RNA
<213> Artificial
<400> 19
agcuuuuaug agcucaugua gca 23

Claims (5)

1. bta-miR-222 for inhibiting goat parainfluenza virus type 3 replication is combined with a nucleotide sequence which is positioned in the 3' UTR of an interferon regulatory gene 2 and is shown as SEQ ID NO. 19, and the bta-miR-222 nucleotide sequence is shown as SEQ ID NO. 1.
2. The use of bta-miR-222 for inhibiting goat parainfluenza virus type 3 replication according to claim 1 in the preparation of a medicament for inhibiting goat parainfluenza virus type 3 replication.
3. The use of bta-miR-222 for inhibiting goat parainfluenza virus type 3 replication according to claim 1 in the preparation of a medicament for treating goat parainfluenza virus type 3.
4. Use of a recombinant luciferase reporter vector pGL3-IRF 23' UTR in assessing the transcriptional level of the bta-miR-222 regulatory interferon-regulated gene 2 that inhibits goat parainfluenza virus type 3 replication of claim 1.
5. The use of bta-miR-222 for inhibiting the replication of goat parainfluenza virus type 3 in breeding transgenic sheep resistant to goat parainfluenza virus type 3 according to claim 1.
CN201810862854.9A 2018-08-01 2018-08-01 Target site sequence for inhibiting goat parainfluenza virus type 3 replication and application thereof Active CN109022437B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201810862854.9A CN109022437B (en) 2018-08-01 2018-08-01 Target site sequence for inhibiting goat parainfluenza virus type 3 replication and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201810862854.9A CN109022437B (en) 2018-08-01 2018-08-01 Target site sequence for inhibiting goat parainfluenza virus type 3 replication and application thereof

Publications (2)

Publication Number Publication Date
CN109022437A CN109022437A (en) 2018-12-18
CN109022437B true CN109022437B (en) 2021-09-14

Family

ID=64648399

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201810862854.9A Active CN109022437B (en) 2018-08-01 2018-08-01 Target site sequence for inhibiting goat parainfluenza virus type 3 replication and application thereof

Country Status (1)

Country Link
CN (1) CN109022437B (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113736799B (en) * 2021-09-17 2024-02-06 江苏省农业科学院 Construction method and application of goat parainfluenza virus 3-type infectious cDNA clone

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105154588A (en) * 2015-09-09 2015-12-16 江苏省农业科学院 Primer pair for detecting caprine parainfluenza virus type 3 (CPIV3) and peste des petits ruminants virus (PPRV)
CN105838678A (en) * 2016-04-11 2016-08-10 江苏省农业科学院 Hybridoma cell strain capable of secreting CPIV3 antibody and ELISA kit

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105154588A (en) * 2015-09-09 2015-12-16 江苏省农业科学院 Primer pair for detecting caprine parainfluenza virus type 3 (CPIV3) and peste des petits ruminants virus (PPRV)
CN105838678A (en) * 2016-04-11 2016-08-10 江苏省农业科学院 Hybridoma cell strain capable of secreting CPIV3 antibody and ELISA kit

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
山羊副流感病毒3型感染MDBK细胞的miRNA表达谱变化分析;李基棕等;《畜牧兽医学报》;20171231;第48卷(第5期);第896-906页 *

Also Published As

Publication number Publication date
CN109022437A (en) 2018-12-18

Similar Documents

Publication Publication Date Title
AU2011322114A1 (en) Compositions and methods for activating expression by a specific endogenous miRNA
CN107893078B (en) SiRNA (small interfering ribonucleic acid) of targeted synaptotagmin-11, expression vector and virus particle and pharmaceutical application thereof
WO2012149646A1 (en) Mirna inhibitors and their uses
WO2010151755A2 (en) TREATMENT OF INFLAMMATORY DISEASES USING miR-124
CN107365785B (en) Gene expression vector for regulating and controlling intracellular NF-kB activity and regulating and controlling method and application thereof
WO2016124002A1 (en) Competitive consumption carcinogenesis micrornas lncrna and oncolytic adenovirus, and application thereof
Zhang et al. Long non‐coding RNA AK085865 ablation confers susceptibility to viral myocarditis by regulating macrophage polarization
CN115725657A (en) Segmented vesicular stomatitis virus vector and preparation method and application thereof
CN109022437B (en) Target site sequence for inhibiting goat parainfluenza virus type 3 replication and application thereof
CN104694576A (en) Method for silencing IFNARI gene in DF-1 cell line
CN101880677B (en) siRNA sequence against 2009 new influenza A virus polymerase gene and nucleoprotein gene and application thereof
CN117562981A (en) SNAPIN protein and application of coding gene thereof in resisting influenza virus
CN111676222A (en) shRNA for inhibiting Mettl3 gene expression, recombinant adeno-associated virus thereof and application thereof
Daniel-Carlier et al. Viral infection resistance conferred on mice by siRNA transgenesis
CN108517335B (en) A kind of Lentiviral and its construction method of liver cell miR-199b low expression
CN113105537B (en) Host protein for promoting replication of influenza A virus and application thereof
CN102168133B (en) Kit for identifying miRNA (micro-ribonucleic acid) target genes and applications thereof
CN110964851A (en) Application of histone modification enzyme gene SETD8 in resisting DNA virus
CN100365123C (en) SiRNA sequence directing towards influenza B virus membrane protein gene and its application
AU2021105546A4 (en) MiRNA against lung injury caused by porcine reproductive and respiratory syndrome virus and its application
CN107586778B (en) shRNA sequence for inhibiting influenza A virus replication and application thereof
CN108113995A (en) The application of microRNA hsa-mir-1-3p and the like and the application for expressing the microRNA carrier
CN110093347B (en) shRNA for inhibiting mouse NPFFR2 gene expression
CN111778247A (en) miR-199-x for targeted regulation of Serpin3 gene and application thereof
CN106337048A (en) Transgenic chicken for expressing Mx-RNAis anti-influenza virus infection

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant