CN110669735A - System for inducing formation of HBV cccDNA and construction method - Google Patents

System for inducing formation of HBV cccDNA and construction method Download PDF

Info

Publication number
CN110669735A
CN110669735A CN201910524757.3A CN201910524757A CN110669735A CN 110669735 A CN110669735 A CN 110669735A CN 201910524757 A CN201910524757 A CN 201910524757A CN 110669735 A CN110669735 A CN 110669735A
Authority
CN
China
Prior art keywords
hbv
plasmid
glu
gene
phic31
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
CN201910524757.3A
Other languages
Chinese (zh)
Inventor
李锋
冯成千
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.)
Guangzhou City No8 People's Hospital
Original Assignee
Guangzhou City No8 People's Hospital
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 Guangzhou City No8 People's Hospital filed Critical Guangzhou City No8 People's Hospital
Priority to CN201910524757.3A priority Critical patent/CN110669735A/en
Publication of CN110669735A publication Critical patent/CN110669735A/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
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from 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
    • C12N15/65Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression using markers
    • 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
    • C12N15/86Viral vectors
    • 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/66Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving luciferase
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5067Liver 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
    • C12N2730/00Reverse transcribing DNA viruses
    • C12N2730/00011Details
    • C12N2730/10011Hepadnaviridae
    • C12N2730/10111Orthohepadnavirus, e.g. hepatitis B virus
    • C12N2730/10122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15041Use of virus, viral particle or viral elements as a vector
    • C12N2740/15043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/10Screening for compounds of potential therapeutic value involving cells

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Immunology (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicinal Chemistry (AREA)
  • Hematology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Plant Pathology (AREA)
  • Virology (AREA)
  • Cell Biology (AREA)
  • Urology & Nephrology (AREA)
  • Analytical Chemistry (AREA)
  • Toxicology (AREA)
  • Food Science & Technology (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The invention discloses a system and a construction method for inducing and forming HBV cccDNA, wherein a stable cell line is constructed by integrating Phic31 recombinase genes, and the 5 'end and the 3' end of linear HBV are provided with complementary luciferase gene fragments and specific sites recognized by Phic31 recombinase, and are cyclized to form cccDNA under the action of Phic31 recombinase, and simultaneously, the complementary luciferase is expressed along with the cccDNA; luciferase provides a shortcut for rapid detection of active HBV cccDNA, and is suitable for high-throughput screening.

Description

System for inducing formation of HBV cccDNA and construction method
Technical Field
The invention relates to a system for inducing and forming HBV cccDNA and a construction method, in particular to HBV cccDNA containing luciferase can be generated in liver cancer cells expressing PhiC31 recombinase by a linear genome, and the luciferase provides a shortcut for quickly detecting active HBV cccDNA and is suitable for high-throughput screening.
Background
Hepatitis B Virus (HBV) is a DNA virus belonging to the Hepadnaviridae (Hepadnaviridae) family. The HBV genome is about 3.2kb in total length and encodes 7 functional proteins: HBV core antigen (HBcAg), e antigen (HBeAg), surface antigen (L, M, S HBsAg), polymerase (pol) and X protein (HBx). There are now about 2.4 million people worldwide infected with HBV, of which there are perhaps 2500 million chronic infectors (Global Hepatitis Report,2017), which greatly increase the risk of cirrhosis and liver cancer, with over 70 million people dying from HBV-related diseases each year, which causes Hepatitis b to be one of the most serious public health problems worldwide.
After HBV enters hepatocytes through hepatocyte surface receptors, its viral nucleic acid (RC-DNA) is released into the cytoplasm, and immediately thereafter, the RC-DNA enters the nucleus to form covalently closed circular DNA (cccdna) or is directly integrated into the host genome. Among them, HBV cccDNA usually exists in the form of minichromosome, and can be used as a template to translate viral mRNA, including four 3.5kb mRNA (encoding core antigen, e antigen and polymerase), 2.4kb and 2.1kb S mRNA (encoding L, M, S three surface antigens), 0.7kb HBx mRNA. Wherein, 3.5kb pregenomic RNA (pgRNA) generates RC-DNA through reverse transcription, and finally forms complete virus particles to be released outside cells to complete the whole life cycle (Chen et al, 2015; Seegand Mason, 2015).
Clinical treatment of HBV at present stage usually involves two regimens of nucleoside analogues(s) analogues (NAs) and Interferon (IFN). Among them, nucleoside analogs can effectively inhibit the generation of new viruses by inhibiting reverse transcription links, and can not achieve the clinical effect of curing HBV by eliminating cccDNA in mechanism, and the risk of drug resistance is also existed in long-term administration, thereby reducing the curative effect (Zoulim and Locanini, 2009, 2012). Interferons inhibit viral replication and clear viruses mainly by activating the immune system, but the cure rate is very low (< 10%), far from meeting clinical needs; furthermore, interferon therapy is costly and has many non-adaptive limitations. Therefore, there is an urgent need to develop new therapeutic strategies with the aim of eliminating HBV cccDNA.
Currently, the cell models for HBV virus research are divided into two broad categories, infected and non-infected cell lines, which are mainly achieved by introducing non-viral vectors into the target cell line. In 1987, scientists in the United states used human hepatoma cell HepG2 to introduce pdolTHBV-1 plasmid containing double-length HBV genome and constructed the currently most commonly used non-infectious cell line HepG2.2.15(Sells et al, 1987), in which HBV gene was continuously expressed and HBV virus production was uncontrolled. In 2006 and 2007, the hepatitis virus institute, de lesel biotechnology and virology institute in the U.S. constructed two cell lines, HepAD38(Zhou et al, 2006) and HepDES19(Guo et al, 2007), which could induce the production of HBV, in which transcription of pgRNA of HBV is under the control of Tetracycline (Tetracycline); in the presence of tetracycline, pgRNA transcription is stopped, cells stop producing RC-DNA and HBV viral particles, and HBV cccDNA with relatively increased abundance can be obtained; the generation of these two cell lines greatly facilitated the understanding of HBV cccDNA. However, the efficiency of cccDNA formation by this method is generally low, and the real reaction of cccDNA in vivo cannot be simulated well.
In infected cell lines, cccDNA production can be efficiently induced by introducing HBV into a host cell line via a series of viral vectors or wild-type viral strains to induce cccDNA formation. In 2003, German scientists constructed HBV primary human liver cell lines using adenovirus vectors (Schulze-Bergkamen et al, 2003). Recently, the national institute of health selects human pluripotent stem cell differentiated hepatocyte-like cells to construct HBV infected cell lines (Xia et al, 2017), which enhance the research depth of cccDNA to some extent. However, since this cell line can efficiently produce cccDNA, which results in the formation of a large amount of useless cccDNA, while in hepatocytes of patients with hepatitis b, the copy number of cccDNA is low (average <1 copy/cell), and the excessive amount of cccDNA makes the immune response generated by host cells to be more different than that of hepatocytes of patients, and thus the physiological state of cccDNA cannot be truly reflected. In addition, the introduction of viral vectors can cause partial HBV genome integration into the genome of host cells, which can affect the judgment of experimental data in the later detection stage due to the inability to distinguish between genomic HBV and cccDNA, thereby leading to different conclusions.
In summary, direct research models of HBV cccDNA are rare, mainly due to: cccDNA and RC-DNA are highly similar on DNA sequence, and the copy number of HBV cccDNA in infected cells is far lower than that of RC-DNA, PCR detection cannot effectively distinguish HBV cccDNA, the Southern Blot detection mode has long time and low sensitivity; 2. the cell model and the animal model supporting HBV infection are lacked, the HBV cell infection model has lower efficiency and rigorous infection conditions, and the method is not suitable for high-throughput screening; the liver humanized mouse model is the only animal model supporting HBV infection, but has high cost and is only suitable for preclinical evaluation of drugs. Therefore, a simple and high-throughput technical platform for directly researching HBV cccDNA is urgently needed in the research and development field of HBV curative drugs.
Reference to the literature
1.Global Hepatitis Report 2017.World Health Organization,GenevaLicence: CC BY-NC-SA 3.0IGO
2.Chen,J.,et al.,2015.New insights into hepatitis B virus biology andimplications for novel antiviral strategies.Natl.Sci.Rev.2,296–313.
3.Seeger,C.,Mason,W.S.,2015.Molecular biology of hepatitis B virusinfection. Virology 479–480,672–686.
4.Zoulim F,Locarnini S.Hepatitis B virus resistance to nucleos(t)ideanalogues. Gastroenterology.2009;137:1593–1608.e1591–e1592.
5.Zoulim F,Locarnini S.Management of treatment failure in chronichepatitis B.Journal of hepatology.2012;56(Suppl 1):S112–S122.
6.Sells,M.A.,Chen,M.L.,Acs,G.,1987.Production of hepatitis B virusparticles in HepG2cells transfected with cloned hepatitis B virus DNA.Proc.Natl.Acad.Sci.U.S.A.84,1005–1009.
7.Zhou,T.,et al.,2006.Hepatitis B virus e antigen production isdependent upon covalently closed circular(ccc)DNA in HepAD38cell cultures andmay serve as a cccDNA surrogate in antiviral screening assays.Antivir.Res.72,116–124.
8.Guo,H.,et al.,2007.Characterization of the intracellulardeproteinized relaxed circular DNA of hepatitis B virus:an intermediate ofcovalently closed circular DNA formation.J.Virol.81,12472–12484.
9.Schulze-Bergkamen,H.,et al.,2003.Primary human hepatocytes–avaluable tool for investigation of apoptosis and hepatitis B virusinfection.J.Hepatol.38, 736–744
10.Xia,Y.,et al.,2017.Human stem cell-derived hepatocytes as a modelfor hepatitis B virus infection,spreading and virus-hostinteractions.J.Hepatol.66, 494–503.
Disclosure of Invention
The invention aims to overcome the defects of the prior art and provides a system and a construction method for inducing the formation of HBV cccDNA; the detection of HBV cccDNA is facilitated by introducing a luciferase reporter gene in HBV cccDNA.
In order to achieve the purpose, the invention adopts the technical scheme that: a system for inducing the formation of HBV cccDNA, comprising a liver cancer cell capable of expressing Phic31 recombinase, wherein the liver cancer cell comprises a linear genome formed by transfection, the linear genome comprises an HBV gene, the 5 'end and the 3' end of the HBV gene are respectively connected with a luciferase gene fragment and a Phic31 recombinase specific recognition site, the luciferase gene fragments connected with the 5 'end and the 3' end of the HBV gene constitute a complete luciferase gene, and the 5 'end and the 3' end of the HBV gene are respectively located at the 5 'end and the 3' end of the linear genome;
when the Phoc 31 recombinase expressed by the liver cancer cells induces the linear genome to form cccDNA, the cccDNA can be transcribed to form a complete open reading frame of a luciferase gene, and the luciferase is translated and expressed by the liver cancer cells.
As the improvement of the technical proposal, the liver cancer cell is HepG2 cell, and the nucleotide sequence of the HBV gene is shown as SEQ ID NO: 1; HBV gene is C genotype of China epidemic strain.
As an improvement of the technical scheme, the nucleotide sequence of the Phic31 recombinase is shown as SEQ ID NO. 2.
As an improvement of the technical proposal, the sequence of the luciferase gene fragment connected with the 5 'end of the HBV gene is shown as SEQ ID NO. 3, and the sequence of the luciferase gene fragment connected with the 3' end of the HBV gene is shown as SEQ ID NO. 4; the specific recognition sites of the Phic31 recombinase are attP and attB.
As a further improvement of the technical proposal, the Phic31 recombinase induces specific sites attP, attR sites formed by attB recombination and luciferase gene fragments Glu-N and Glu-C are positioned between the positions 85 and 86 after the initiation codon of the core antigen, and Glu-N and Glu-C are positioned at the outer sides; the sequence of the luciferase gene fragment Glu-N is shown as SEQ ID NO. 3, and the sequence of the luciferase gene fragment Glu-C is shown as SEQ ID NO. 4; the linear genome is transfected by ligation on an adenovirus plasmid.
In addition, the invention also provides HBV cccDNA formed by the system.
In addition, the present invention also provides a method for constructing a system for inducing the formation of HBV cccDNA, comprising the steps of: constructing liver cancer cells expressing Phic31 recombinase; constructing recombinant adenovirus plasmid connected with linear genome, packaging, purifying and transfecting liver cancer cell.
As an improvement of the technical scheme, the liver cancer cell expressing the Phic31 recombinase is constructed by the following method:
s11) replacing the Phyc 31 recombinase gene with the fluorescent protein gene EGFP by taking the plasmid pRlenti-Puro-EGFP as a vector to obtain a lentiviral vector plasmid pRenti-Puro-Phyc 31; the plasmid pRlenti-Puro-EGFP is a lentiviral vector with a screening gene Puro and a fluorescent protein gene EGFP, and the Puro-EGFP is connected by a self-shearing polypeptide 2A; the nucleotide sequence of the Phic31 recombinase is shown as SEQ ID NO. 2;
s12) lentivirus vector plasmid pRenti-Puro-Phic31, then the lentivirus pRenti-Puro-Phic31 is collected, the lentivirus pRenti-Puro-Phic31 is transfected into HepG2 cells, and HePG2-Phic31 cells which stably express Phic31 recombinase are screened.
As an improvement of the technical scheme, the recombinant adenovirus plasmid is constructed by the following method:
s21) connecting the luciferase fragment Glu-N and the luciferase fragment Glu-C to the 5 'end and the 3' end of the linear HBV gene respectively by adopting an overlap extension PCR method, thus constructing a Glu-N-HBV-Glu-C fusion fragment; the sequence of the luciferase fragment Glu-N is shown as SEQ ID NO. 3, and the sequence of the luciferase fragment Glu-C is shown as SEQ ID NO. 4;
s22) cutting an exogenous gene P2B9 of the PMC-P2B9 plasmid to obtain the PMC plasmid; the PMC-P2B9 plasmid is a T cloning vector with multiple cloning sites, and attP and attB recognition sites are respectively positioned at two ends of the multiple cloning sites; cloning the Glu-N-HBV-Glu-C fusion fragment onto a PMC plasmid, and carrying out transfection and purification to obtain a recombinant plasmid PMC-HBV-C-Gluc;
s23) taking the PGA1-empty plasmid as a skeleton vector, taking the PMC-HBV-C-Gluc as a target fragment, and cloning the HBV-C-Gluc fusion fragment to the PGA1-empty plasmid through enzyme digestion and connection to obtain a shuttle plasmid PGA 1-HBV-C-Gluc; the PGA1-empty plasmid does not carry exogenous genes;
s24) carrying out enzyme digestion on shuttle plasmid PGA1-HBV-C-Gluc and adenovirus plasmid AD5, recovering enzyme digestion products for transfection, and collecting recombinant adenovirus plasmid AD5-HBV-C-Gluc after transfection.
In addition, the invention also provides the system and/or the application of the HBV cccDNA in constructing an HBV drug screening system.
The invention has the beneficial effects that: the invention provides a system for inducing and forming HBV cccDNA and a construction method, wherein a stable cell line is constructed by integrating Phic31 recombinase genes, and the 5 'end and the 3' end of linear HBV are provided with complementary luciferase gene fragments and specific sites recognized by Phic31 recombinase, and are cyclized to form cccDNA under the action of Phic31 recombinase, and the complementary luciferase is expressed along with the cccDNA; the design of the invention has the following advantages: the liver cancer cells are preferably HepG2 cells, the HepG2 cells have simple biological background, the liver cancer cells are good vectors for researching hepatitis B virus and cccDNA, the quantity of introduced linear HBV can adjust the formation quantity of the cccDNA, the influence of a large quantity of useless cccDNA on an experiment is avoided, no virus is generated in the process, HBV can not be integrated into the genome of a host cell, the obtained experimental data can more intuitively reflect the state of the cccDNA, and expressed luciferase is secreted out of cells and can be detected by directly sucking a small amount of culture medium supernatant.
Drawings
FIG. 1 shows the transcript levels of the Phic31 recombinase gene in HePG2-Phic31 cells;
FIG. 2 shows the expression level of Phic31 recombinase in HePG2-Phic31 cells;
FIG. 3 shows the expression level of luciferase in HePG2-Phic31 cells;
fig. 4 is a schematic structural diagram of HBV cccDNA; wherein, the Phic31 recombinase induces specific sites attP, attR sites formed by attB recombination and two sections of luciferase gene fragments Glu-N and Glu-C are positioned between the positions 85 and 86 after the initiation codon of the Core gene, and Glu-N and Glu-C are positioned at the outer sides; when the Phic31 recombinase induces linear HBV to form cccDNA, Glu-N and Glu-C can be synthesized into Luciferase reporter gene Luciferase with complete ORF again, and the Luciferase reporter gene Luciferase is transcribed and translated to be expressed.
Detailed Description
To better illustrate the objects, aspects and advantages of the present invention, the present invention will be further described with reference to the following detailed description and accompanying drawings.
Obtaining genomic DNA from a patient sample
Hepatitis B virus RNA of a patient sample is extracted by using a nucleic acid extraction or purification kit (Daan gene at Zhongshan university), and the operation is as follows:
1) adding 200ul of sample into a centrifuge tube, and adding 50ul of protease K;
2) adding 200ul of cracking solution, performing vortex oscillation for 15sec, centrifuging for 10sec, and performing centrifugation at 72 ℃ for 10 min;
3) adding 250ul of absolute ethyl alcohol, and carrying out vortex oscillation for 15 sec;
4) sucking the mixed solution into a centrifugal column, centrifuging for 1min at room temperature at 12,000g, and transferring the centrifugal column to a new collecting pipe;
5) adding 500ul inhibitor removing solution into centrifugal column, centrifuging at room temperature for 1min at 12,000g, transferring the centrifugal column to new collection tube;
6) adding 500ul deionized liquid into a centrifugal column, centrifuging at room temperature for 1min at 12,000g, and transferring the centrifugal column to a new collection tube;
7) repeating the step 6;
8) centrifuging the spin column collection tube at 14,000g for 3min at room temperature to remove residual ethanol;
9) taking out the centrifugal column, placing in a new 1.5ml centrifugal tube, and placing for 2min at 72 ℃;
10) 50ul of eluent preheated at 72 ℃ is carefully added just above the membrane of the centrifugal column, the tube cover is tightly covered, the mixture is kept stand for 1min at room temperature, and 14,000g is centrifuged for 1 min.
Secondly, obtaining the C-type HBV fragment
The C-type HBV gene is amplified from the extracted and purified cDNA by utilizing the PCR technology, and the nucleotide sequence is shown as SEQ ID NO. 1.
By using
Figure BDA0002097844710000081
The Taq 2x Mix kit (NEB, M0287S) amplifies HBV type C, and the reaction solution is prepared according to the following ratio:
and reacted according to the following procedure: 1) denaturation 94 ℃ for 30 seconds, 2) annealing 60 ℃ for 30 seconds, 3) extension 68 ℃ for 3 minutes, 4) cycle number 35 times.
The C-type HBV gene was extracted from the PCR product using agarose gel recovery kit (Tiangen Biochemical technology). Wherein, the PCR product is separated according to the molecular weight by agarose gel electrophoresis, a single target DNA band is cut off from the agarose gel, and the cut single target DNA band is put into a clean centrifugal tube and weighed. To the gel block was added an equal volume of solution PN (100. mu.l of PN solution if the gel weighed 0.1g and the volume was considered to be 100. mu.l), and placed in a water bath at 60 ℃ with the centrifuge tube gently turned upside down to ensure adequate dissolution of the gel block. Adding the solution obtained in the previous step into an adsorption column CA2 (column equilibrium step: adding 500. mu.l of equilibrium solution BL into adsorption column CA2, centrifuging at 12,000rpm for 1min, pouring off the waste liquid in the collection tube, placing the adsorption column into the collection tube), standing at room temperature for 2min, centrifuging at 12,000rpm (13,400 Xg) for 30-60sec, pouring off the waste liquid in the collection tube, and placing adsorption column CA2 into the collection tube. Add 600. mu.l of rinsing solution PW (check whether absolute ethanol has been added before use) to adsorption column CA2, centrifuge at 12,000rpm (. about.13,400 Xg) for 30-60sec, dump the waste liquid from the collection tube, and place adsorption column CA2 into the collection tube. Centrifuge at 12,000rpm (13,400 Xg) for 2min to remove the rinse as much as possible. The adsorption column CA2 was left at room temperature for several minutes and thoroughly dried to prevent the residual rinse from affecting the next experiment. Placing the adsorption column CA2 in a clean centrifuge tube, suspending and dropwise adding appropriate amount of elution buffer EB into the middle position of the adsorption membrane, and standing at room temperature for 2 min. The DNA solution was collected by centrifugation at 12,000rpm (. about.13,400 Xg) for 2 min.
Thirdly, obtaining the truncated two-part fragment of the Luciferase
The plasmid containing the Luciferase is used as a template, the PCR technology is utilized to amplify the Luciferase into two parts, marked as Luciferase gene segments Glu-N and Glu-C, and the nucleotide sequences are shown as SEQ ID NO. 3 and SEQ ID NO. 4.
PCR products are recovered by an agarose gel recovery kit (Tiangen Biotechnology), and the specific recovery steps are detailed in step 2.
Fourthly, obtaining the fused HBV-Glu fragment
By utilizing the overlap extension PCR technology, Luciferase gene fragments Glu-N and Glu-C are respectively connected to the 5 'end and the 3' end of the HBV gene to construct Glu-N-HBV-Glu-C fusion fragments, and when HBV is cyclized, Glu-N and Glu-C form a complete Luciferase gene, thereby being used as a report system to prompt the formation of cccDNA. The PCR reaction conditions were as follows:
1)PCR Round1:
Figure BDA0002097844710000091
pre-denaturation at 94 deg.C for 5 min; denaturation, 30s at 94 ℃; annealing at 60 ℃ for 30 s; extension, 3min at 65 ℃ 20 s; 20 cycles; extending, 5min at 65 ℃; keep at 4 ℃ keep.
2)PCR Round2:
Figure BDA0002097844710000092
Pre-denaturation at 94 deg.C for 5 min; denaturation, 30s at 94 ℃; annealing at 60 ℃ for 30 s; extension, 3min at 65 ℃ 20 s; 35 cycles; extending, 5min at 65 ℃; keep at 4 ℃ keep.
Fifthly, obtaining the recombinant plasmid PMC-HBV-C-Gluc
The plasmid PMC-P2B9 is a T cloning vector (TaKaRa) with Multiple Cloning Sites (MCS), two elements attP and attB specifically recognized by a Phic31 recombinase are respectively positioned at two ends of the multiple cloning sites, the vector is used as a basic skeleton for transformation, and a self-carried exogenous gene P2B9 is excised, so that the PMC plasmid is obtained; the Glu-N-HBV-Glu-C fusion fragment was cloned on PMC plasmid. PMC-P2B9 is taken as a framework vector, Glu-N-HBV-Glu-C is taken as a fragment, the fragments are respectively cut by enzyme and then connected, and finally, a connection product is taken to transform competent cells (Tiangen biochemical technology), and the transformation process is as follows:
add 100. mu.l of competent cell suspension to a 1.5ml centrifuge tube and place on ice. Add 10ul ligation product, mix gently with a pipette, and let stand on ice for 30 min. And thermally shocking the mixture in a water bath at 42 ℃ for 90sec, and then rapidly putting the mixture on ice for 3-5 min. Adding 1ml LB liquid culture medium (containing 10g tryptone per liter yeast extract 5g, NaCl 5g, pH 7.0, no antibiotics, autoclaving), mixing, shaking culturing at 37 deg.C (180rpm) for 1 hr to restore the normal growth state of bacteria, and expressing plasmid-encoded antibiotic resistance gene. 100 mul of the bacterial liquid is taken and put on LB solid culture medium containing antibiotic Amp (15 g/L of agar for bacterial culture is added into LB liquid culture medium, the bacterial liquid culture medium is autoclaved, slightly cooled and then is led into a bacterial culture plate for continuous cooling), and the bacterial liquid is evenly coated. After the bacterial liquid is absorbed by the culture medium, carrying out inverted culture at 37 ℃ for 12-16 hours.
Sixthly, obtaining shuttle plasmid PGA1-HBV-C-Gluc and adenovirus plasmid AD5-HBV-C-Gluc
PGA1-empty is the shuttle plasmid necessary for constructing the adenovirus vector, does not carry any exogenous gene, takes the plasmid as the skeleton vector, PMC-HBV-C-Gluc is the target segment, utilizes the restriction enzyme of TaKaRa company to carry out enzyme digestion, and the reaction system is as follows:
reagent Volume of
Plasmids 10ug
10X M Buffer 2ul
KpnI 1ul
NotI 1ul
RNase-Free ddH2O Make up to 20ul
PGA1 and HBV-C-Gluc were connected by using a Solutioni ligation kit from TaKaRa, the reaction time was 8 hours, and the system was as follows:
Figure BDA0002097844710000101
Figure BDA0002097844710000111
after the reaction is completed, TOP10 competent cells (Tiangen Biochemical company) are transformed, and the specific steps are shown in step five; colony PCR and enzyme digestion identify the correct clone.
Restriction enzymes SgrAI and SgrAI of TaKaRa, BstZ17I are used for enzyme digestion of PGA1-HBV-C-Gluc and adenovirus plasmid AD5-E1E3 respectively, the reaction time is 12 hours, after enzyme digestion products are recovered, BJ5183 competent cell transformation is carried out, and the two are recombined. The competence transformation step is shown in step five.
The transformed clone is subjected to agarose electrophoresis to greatly identify a recombinant plasmid (AD5 is 32K bp), and the successfully recombined clone is inevitably the large plasmid. And (3) identifying successful clone, then carrying out XL-Blue competent transformation, and limiting the growth of small plasmid by XL-Blue competent cells to achieve the effect of purifying large plasmid.
Clones after XL-Blue transformation were identified by using the restriction enzyme HindIII of TaKaRa, and agarose electrophoresis revealed the insertion of the target band.
Seventhly, packaging and purifying adenovirus AD5-HBV-C-Gluc
293T cells were plated in 15cm dishes and transfected at a cell density of 70% by replacing fresh medium (Gibco DMEM + 10% FBS) before transfection. The transfection reagent was Lipo3000, AD5-HBV-C-Gluc and Lipo3000 were diluted with Opti-MEM, mixed well and then left to stand at room temperature for 15 minutes, and then added gently to the cell dish medium without changing the medium. And after 48 hours, observing the pathological state of the cells, wherein cytoplasm is bright, cell nucleus is reduced, collecting the cells, repeating the steps in a liquid nitrogen-37 ℃ water bath for three times, and centrifuging to collect supernatant, namely virus supernatant. The virus supernatant was purified by cesium chloride gradient centrifugation and stored in a freezer at-80 ℃.
Eighthly, obtaining recombinase Phic31CDS fragment
The plasmid PT-Phic31 is a T cloning vector (Promega) with Phic31 recombinase gene, Phic31CDS is amplified by PCR technology by taking the plasmid as a template, the nucleotide sequence is shown as SEQ ID NO:2, and a reaction system and a kit are shown in a step two.
Ninthly, obtaining a lentiviral vector plasmid pRenti-Puro-Phic31
The plasmid pRlenti-Puro-EGFP is a lentiviral vector with a screening gene Puro and a fluorescent protein gene EGFP, and is constructed and modified by the laboratory, wherein the Puro-EGFP is formed by connecting self-cleavage polypeptide 2A (self-cleavage 2A peptide,2A), and the plasmid pRenti-Puro-Phic31 can be obtained by replacing a Phic31 recombinase gene with the fluorescent protein gene EGFP on the basis of the vector. And (3) performing SpeI and EcoRI (TaKaRa) double enzyme digestion on the Phic31PCR product and pRlenti-Puro-EGFP, fully reacting, and performing gel electrophoresis to recover enzyme digestion fragments, wherein the specific operation is shown in the step two. Ligation was performed using a Solutioni kit (TaKaRa), and TOP10 competent cell transformation was performed after 6 hours of reaction, as detailed in step five. And selecting the colony with good growth for PCR and enzyme digestion identification.
Ten, packaging and collecting of lentivirus pRenti-Puro-Phic31
293T cells were plated in 10cm dishes and transfected at a cell density of 70% by replacing fresh medium (Gibco DMEM + 10% FBS) before transfection. The transfection reagent was Lipo3000, and pRenti-Puro-Phic31 and Lipo3000 were diluted with Opti-MEM, mixed well and then allowed to stand at room temperature for 15 minutes, and then added gently to the cell dish medium without changing the medium. And collecting cell supernatant after 48 hours, namely virus supernatant.
Eleven, establishing a HePG2-Phic31 stable cell line
HepG2 cells were plated in 6cm dishes and infected at 90% cell density, with fresh medium (Gibco DMEM + 10% FBS) being replaced before infection. 2ml of the supernatant of pRenti-Puro-Phic31 virus was added to each 6cm dish, the medium was replaced after 12 hours with fresh medium and puromycin (Puro, 5mg/ml) was added at 48 hours with continued application of screening pressure until the control cells were completely apoptotic.
Identification of twelve, HePG2-Phic31 Stable cell lines
Real-Time PCR detection of Phic31RNA expression
HePG2-Phic31 and control HePG2 cells were collected and total RNA was extracted using TRIZOL lysis method. 1ml of TRIZOL solution was added to each sample, and after sufficient lysis, 0.2ml of chloroform was added thereto, followed by vigorous shaking for 30 seconds and standing at room temperature for 5 minutes. The supernatant liquid was aspirated, transferred to a new centrifuge tube, added to an equal volume of isopropanol, placed on ice for 10 minutes, and centrifuged at 12000g for 10 minutes. The supernatant was discarded, 1ml of 75% ethanol was added, the mixture was inverted upside down until RNA pellet was suspended, and then RNA was dissolved in 50ul of RNase-FreedH 2O, and if necessary, dissolved in water at 55 ℃ to 60 ℃ for 10 minutes. Using ReverTra
Figure BDA0002097844710000121
The kit (TOYOBO, FSK-101) carries out reverse transcription to synthesize cDNA, and the reaction system is as follows:
reagent Volume of
5x RT buffer 4ul
dNTP mixture(10mM) 2ul
RNase inhibitor(10U/μl) 1ul
Oligo(dT)20 2ul
RNA 2ug(~ul)
RNase-Free ddH2O Make up to 20ul
The reaction procedure was as follows: the incorporated at 42 ℃ for 20min, the Heat at 99 ℃ for 5min, the stored activated solution at 4 ℃ or-20 ℃ using SsoadvancedTMUniversal
Figure BDA0002097844710000131
The GreenSupermix kit (BIO-RAD, 172-5274) detects the expression level of Phic31RNA of HepG2-Phic31 cell line.
As shown in FIG. 1, the invention succeeded in HePG2-Phic31 cells.
Western-blot detection of expression level of Phic31 protein
HePG2-Phic31 and control HePG2 cells were collected and total protein was extracted using RIPA lysis method. Adding 1ml of RIPA lysate to each sample to lyse cells to release protein, centrifuging at 14000g for 30 minutes, transferring the supernatant to a new centrifuge tube, adding 5 SDS-PAGE protein loading buffer, and heating in a boiling water bath for 3-5 minutes to fully denature the protein. SDS-PAGE gel electrophoresis was used to separate the proteins sufficiently, after which membrane transfer was performed, proteins were transferred from the gel to nitrocellulose (NC membrane) and then blocked to exclude non-specific sites on the membrane from binding to primary antibodies, typically by incubation overnight with prepared milk, PBST rinsed three times, washing away residual milk followed by incubation of the primary antibody for 1 hour 30 minutes, and finally by incubation with horseradish peroxidase (HRP) -labelled secondary antibody for 2 hours.
Detection of the Phic31 protein was performed using the Immobilon Western kit (Millipore, WBKLS 0500).
As shown in FIG. 2, the present invention succeeded in HePG2-Phic31 cells.
Thirteen, induction identification of cccDNA formation
HepG2-Phic31 and HepG2 cells are planted in a six-hole plate culture dish, an experimental group of HepG2-Phic31 + AD5-HBV-C-Gluc is set, a control group of HepG2-Phic31 and HepG2+ AD5-HBV-C-Gluc and adenovirus AD5-HBV-C-Gluc are infected with fresh culture medium (Gibco DMEM + 10% FBS) after 2 hours, and the culture medium is collected after 48 hours to detect Luciferase.
Detecting the Luciferase by using a Renilla Luciferase Assay System kit (Promega, E2810), mixing a sample to be detected with the same volume of a detection reagent, fully reacting, and then detecting in a multi-hole micropore plate luminescence detector
Figure BDA0002097844710000141
The fluorescence values were read.
As shown in FIG. 3, the experimental group HepG2-Phic31 + AD5-HBV-C-Gluc can express high-content luciferase, and the invention can well construct a system for inducing the formation of HBV cccDNA and is used for detecting the content of the HBV cccDNA; HBV cccDNA is shown in figure 4.
Finally, it should be noted that the above embodiments are intended to illustrate the technical solutions of the present invention and not to limit the scope of the present invention, and although the present invention has been described in detail with reference to the preferred embodiments, it should be understood by those skilled in the art that modifications and equivalent substitutions can be made to the technical solutions of the present invention without departing from the spirit and scope of the technical solutions of the present invention.
Figure BDA0002097844710000151
Figure BDA0002097844710000161
Figure BDA0002097844710000171
Figure BDA0002097844710000181
Figure BDA0002097844710000191
Sequence listing
<110> eighth national hospital in Guangzhou City
<120> System for inducing formation of HBV cccDNA and construction method
<130>2019.06.06
<160>4
<170>PatentIn version 3.3
<210>1
<211>3215
<212>DNA
<213>Hepatitis B virus
<400>1
ctccacaaca ttccaccaag ctctgctaga tcccagagtg aggggcctat attttcctgc 60
tggtggctcc agttccggaa cagtaaaccc tgttccgact actgcctcac ccatatcgtc 120
aatcttctcg aggactgggg accctgcacc gaacatggag aacacaacat caggattcct 180
aggacccctg ctcgtgttac aggcggggtt tttcttgttg acaagaatcc tcacaatacc 240
acagagtcta gactcgtggt ggacttctct caattttcta gggggagcac ccacgtgtcc 300
tggccaaaat tcgcagtccc caacctccaa tcactcacca acctcttgtc ctccaatttg 360
tcctggctat cgctggatgt gtctgcggcg ttttatcata ttcctcttca tcctgctgct 420
atgcctcatc ttcttgttgg ttcttctgga ctaccaaggt atgttgcccg tttgtcctct 480
acttccagga acatcaacca ccagcacggg gccatgcaag acctgcacga ttcctgctca 540
aggaacctct atgtttccct cttgttgctg tacaaaacct tcggacggaa actgcacttg 600
tattcccatc ccatcatcct gggctttcgc aagattccta tgggagtggg cctcagtccg 660
tttctcctgg ctcagtttac tagtgccatt tgttcagtgg ttcgtagggc tttcccccac 720
tgtttggctt tcagttatat ggatgatgtg gtattggggg ccaagtctgt acaacatctt 780
gagtcccttt ttacctctat taccaatttt cttttgtctt tgggtataca tttaaaccct 840
aataaaacca aacgttgggg ctactccctt aacttcatgg gatatgtaat tggaagttgg 900
ggtactttac cgcaggaaca tattgtacta aaactcaagc aatgttttcg aaaattacct 960
gtaaatagac ctattgattg gaaagtctgt caaagaattg tgggtctttt gggctttgct 1020
gcccctttta cacaatgtgg ctatcctgcc ttgatgcctt tatatgcatg tatacaatct 1080
aagcaggctt tcactttctc gccaacttac aaggcctttc tgtgtaaaca atatctgaac 1140
ctttaccccg ttgcccggca acggtcaggt ctctgccaag tgtttgctga cgcaaccccc 1200
actggatggg gcttggccat aggccatcgg cgcatgcgtg gaacctttgt ggctcctctg 1260
ccgatccata ctgcggaact cctagcagct tgtttcgctc gcagccggtc tggagcgaaa 1320
cttatcggca ccgacaactc tgttgtcctc tctcggaaat acacctcctt tccatggctg 1380
ctagggtgtg ctgccaactg gatcctgcgc gggacgtcct ttgtctacgt cccgtcggcg 1440
ctgaatcccg cggacgaccc gtctcggggc cgtttggggc tctaccgtcc ccttcttcat 1500
ctgccgttcc ggccgaccac ggggcgcacc tctctttacg cggtctcccc gtctgtgcct 1560
tctcatctgc cggaccgtgt gcacttcgct tcacctctgc acgtcgcatg gagaccaccg 1620
tgaacgccca ccaggtcttg cccaaggtct tacataagag gactcttgga ctctcagcaa 1680
tgtcaacgac cgaccttgag gcatacttca aagactgtgt gtttaaagac tgggaggagt 1740
tgggggagga gattaggtta aaggtctttg tactaggagg ctgtaggcat aaattggtct 1800
gttcaccagc accatgcaac tttttcacct ctgcctaatc atctcatgtt catgtcctac 1860
tgttcaagcc tccaagctgt gccttgggtg gctttggggc atggacattg acccgtataa 1920
agaatttgga gcatctgtgg agttactctc ttttttgcct tctgacttct ttccttctat 1980
tcgagatctc ctcgacaccg cttctgctct gtatcgggag gccttagagt ctccggaaca 2040
ttgttcacct caccatacag cactcaggca agctattctg tgttggggtg agttgatgaa 2100
tctggccacc tgggtgggaa gtaatttgga agacccagca tccagggaat tagtagtcag 2160
ctatgtcaat gttaatatgg gcctaaaaat cagacaacta ttgtggtttc acatttcctg 2220
tcttactttt ggaagagaaa ctgttcttga gtatttggta tcttttggag tgtggattcg 2280
cactcctccc gcttacagac caccaaatgc ccctatctta tcaacacttc cggaaactac 2340
tgttgttaga cgacgaggca ggtcccctag aagaagaact ccctcgcctc gcagacgaag 2400
atctcaatcg ccgcgtcgca gaagatctca atctcgggaa tctcaatgtt agtatccctt 2460
ggactcataa ggtgggaaac tttactgggc tttattcttc tactgtacct gtctttaatc 2520
ccgagtggca aactccctcc tttcctcaca ttcatttaca ggaggacatt attaatagat 2580
gtcaacaata tgtgggccct cttacagtta atgaaaaaag gagattaaaa ttaattatgc 2640
ctgctaggtt ctatcctaac cttaccaaat atttgccctt agacaaaggc attaaaccgt 2700
attatcctga acatgcagtt aatcattact tcaaaactag gcattattta catactctgt 2760
ggaaggctgg cattctatat aagagagaaa ctacacgcag cgcctcattt tgtgggtcac 2820
catattcttg ggaacaagag ctacagcatg ggaggttggt cttccaaacc tcgacaaggc 2880
atggggacga atctttctgt tcccaatcct ctgggattct ttcccgatca ccagttggac 2940
cctgcgttcg gagccaactc aaacaatcca gattgggact tcaaccccaa caaggatcac 3000
tggccagagg caaatcaggt aggagcggga gcattcgggc cagggttcac cccaccacac 3060
ggcggtcttt tggggtggag ccctcaggct cagggcatat tgacaacagt gccagcagca 3120
cctcctcctg cctccaccaa tcggcagtca ggaagacagc ctactcccat ctctccacct 3180
ctaagagaca gtcatcctca ggccatgcag tggaa 3215
<210>2
<211>1818
<212>DNA
<213> artificially synthesized sequence
<400>2
atggacacct atgctggcgc ctatgaccgc cagtccaggg agagggagaa ctcctctgct 60
gcctcccctg ccacccaacg cagcgccaat gaggacaagg ctgctgacct gcagagggag 120
gtggagaggg atggcggcag gttcaggttt gtgggccact tctctgaggc ccctggcacc 180
tctgcctttg gcacagctga gaggcctgag tttgagagga ttctgaatga gtgcagggct 240
ggcaggctga acatgatcat tgtctatgat gtctcccgct tctcccgcct gaaagtcatg 300
gatgccatcc ccattgtctc tgagctgctg gccctgggcg tgaccattgt ctccacccaa 360
gagggcgtct tcaggcaggg caatgtgatg gacctgatcc atctgatcat gaggctggat 420
gcctcccaca aggagtcctc cctgaagtct gccaagatcc tggacaccaa gaacctgcag 480
agggagctgg gcggctatgt gggcggcaag gccccatatg gctttgagct ggtctctgag 540
accaaggaga tcaccaggaa tggcaggatg gtgaatgtgg tgatcaacaa gctggcccac 600
tccaccaccc ccctgaccgg cccatttgag tttgagcctg atgtgatcag gtggtggtgg 660
agggagatca agacccacaa gcatctgcca ttcaagcctg gctcccaggc tgccatccat 720
cctggctcca tcaccggcct gtgcaagagg atggatgctg atgctgtgcc caccaggggc 780
gagaccattg gcaagaagac agcctcctct gcctgggacc ctgccacagt gatgaggatt 840
ctgagggacc ccaggattgc tggctttgct gctgaggtga tctacaagaa gaagcctgat 900
ggcaccccca ccaccaagat tgagggctac aggattcaga gggaccccat caccctgagg 960
cctgtggagc tggactgtgg ccccatcatt gagcctgccg agtggtatga gctgcaggcc 1020
tggctggatg gcaggggcag gggcaagggc ctgtccaggg gccaggccat cctgtctgcc 1080
atggacaagc tgtactgcga gtgtggcgct gtgatgacct ccaagagggg cgaggagtcc 1140
atcaaggact cctaccgctg ccgccgccgc aaggtggtgg acccatctgc ccctggccag 1200
catgagggca cctgcaatgt ctccatggct gccctggaca agtttgtggc tgagaggatc 1260
ttcaacaaga tcaggcatgc tgagggcgat gaggagaccc tggccctgct gtgggaggct 1320
gccaggaggt ttggcaagct gacagaggcc cctgagaagt ctggcgagag ggccaacctg 1380
gtggctgaga gggctgatgc cctgaatgcc ctggaggagc tgtatgagga cagggctgct 1440
ggcgcctatg atggccctgt gggcaggaag cacttcagga agcagcaggc tgccctgacc 1500
ctgaggcagc agggcgctga ggagaggctg gctgagctgg aggctgctga ggcccccaag 1560
ctgcccctgg accagtggtt ccctgaggat gctgatgctg accccaccgg ccccaagtcc 1620
tggtggggca gggcctctgt ggatgacaag agggtctttg tgggcctgtt tgtggacaag 1680
attgtggtga ccaagtccac cacaggcagg ggccagggca cccccattga gaagagggcc 1740
tccatcacct gggccaagcc ccccacagat gatgatgagg atgatgccca ggatggcaca 1800
gaggatgtgg ctgcctag 1818
<210>3
<211>134
<212>DNA
<213> artificially synthesized sequence
<400>3
gtgaatggcg tgaaggtgct gtttgccctg atctgcattg ctgtggctga ggccaagccc 60
acagagaaca atgaggactt caacattgtg gctgtggcct ccaactttgc caccaccgac 120
ctggatgccg acag 134
<210>4
<211>430
<212>DNA
<213> artificially synthesized sequence
<400>4
gggcaagctg cctggcaaga agctgcccct ggaggtgctg aaggagatgg aggccaatgc 60
caggaaggct ggctgcacca ggggctgcct gatctgcctg tcccacatca agtgcacccc 120
caagatgaag aagttcatcc ctggccggtg ccacacctat gagggcgaca aggagtctgc 180
ccagggcggc attggcgagg ccattgtgga catccctgag atccctggct tcaaggacct 240
ggagcccatg gagcagttca ttgcccaggt ggacctgtgt gtggactgca ccaccggctg 300
cctgaagggc ctggccaatg tgcagtgctc tgacctgctg aagaagtggc tgccccagag 360
atgtgccacc tttgcctcca aaatccaggg ccaggtggac aagatcaagg gcgctggcgg 420
cgatgacacc 430

Claims (10)

1. A system for inducing the formation of HBV cccDNA, which comprises a liver cancer cell capable of expressing Phoc 31 recombinase, wherein a linear genome formed by transfection is contained in the liver cancer cell, the linear genome comprises HBV genes, the 5 'end and the 3' end of the HBV genes are respectively connected with a luciferase gene fragment and a Phic31 recombinase specific recognition site, the luciferase gene fragments connected with the 5 'end and the 3' end of the HBV genes form a complete luciferase gene, and the luciferase gene fragments connected with the 5 'end and the 3' end of the HBV genes are respectively positioned at the 5 'end and the 3' end of the linear genome;
when the Phoc 31 recombinase expressed by the liver cancer cells induces the linear genome to form cccDNA, the cccDNA can be transcribed to form a complete open reading frame of a luciferase gene, and the luciferase is translated and expressed by the liver cancer cells.
2. The system of claim 1, wherein the liver cancer cell is HepG2 cell, and the nucleotide sequence of the HBV gene is shown in SEQ ID NO. 1.
3. The system of claim 1, wherein the nucleotide sequence of the Phic31 recombinase is as set forth in SEQ ID NO 2.
4. The system as claimed in claim 1, wherein the sequence of luciferase gene fragment linked to the 5 'end of HBV gene is shown in SEQ ID NO. 3, and the sequence of luciferase gene fragment linked to the 3' end of HBV gene is shown in SEQ ID NO. 4; the specific recognition sites of the Phic31 recombinase are attP and attB.
5. The system of claim 4, wherein the Phic31 recombinase induces the specific sites attP, attR sites formed by attB recombination, and luciferase gene fragments Glu-N and Glu-C, located between positions 85 and 86 after the initiation codon of the core antigen, with Glu-N and Glu-C being on the outside; the sequence of the luciferase gene fragment Glu-N is shown as SEQ ID NO. 3, and the sequence of the luciferase gene fragment Glu-C is shown as SEQ ID NO. 4; the linear genome is transfected by ligation on an adenovirus plasmid.
6. HBV cccDNA formed systematically according to any one of claims 1 to 5.
7. A method for constructing a system for inducing the formation of HBV cccDNA, comprising the following steps: constructing liver cancer cells expressing Phic31 recombinase; constructing recombinant adenovirus plasmid connected with linear genome, packaging, purifying and transfecting liver cancer cell.
8. The method of claim 7, wherein the liver cancer cell expressing Phic31 recombinase is constructed by:
s11) replacing the Phyc 31 recombinase gene with the fluorescent protein gene EGFP by taking the plasmid pRlenti-Puro-EGFP as a vector to obtain a lentiviral vector plasmid pRenti-Puro-Phyc 31; the plasmid pRlenti-Puro-EGFP is a lentiviral vector with a screening gene Puro and a fluorescent protein gene EGFP, and the Puro-EGFP is connected by a self-shearing polypeptide 2A; the nucleotide sequence of the Phic31 recombinase is shown as SEQ ID NO. 2;
s12) lentivirus vector plasmid pRenti-Puro-Phic31, then the lentivirus pRenti-Puro-Phic31 is collected, the lentivirus pRenti-Puro-Phic31 is transfected into HepG2 cells, and HePG2-Phic31 cells which stably express Phic31 recombinase are screened.
9. The method of claim 7, wherein the recombinant adenovirus plasmid is constructed by:
s21) connecting the luciferase fragment Glu-N and the luciferase fragment Glu-C to the 5 'end and the 3' end of the linear HBV gene respectively by adopting an overlap extension PCR method, thus constructing a Glu-N-HBV-Glu-C fusion fragment; the sequence of the luciferase fragment Glu-N is shown as SEQ ID NO. 3, and the sequence of the luciferase fragment Glu-C is shown as SEQ ID NO. 4;
s22) cutting an exogenous gene P2B9 of the PMC-P2B9 plasmid to obtain the PMC plasmid; the PMC-P2B9 plasmid is a T cloning vector with multiple cloning sites, and attP and attB recognition sites are respectively positioned at two ends of the multiple cloning sites; cloning the Glu-N-HBV-Glu-C fusion fragment onto a PMC plasmid, and carrying out transfection and purification to obtain a recombinant plasmid PMC-HBV-C-Gluc;
s23) taking the PGA1-empty plasmid as a skeleton vector, taking the PMC-HBV-C-Gluc as a target fragment, and cloning the HBV-C-Gluc fusion fragment to the PGA1-empty plasmid through enzyme digestion and connection to obtain a shuttle plasmid PGA 1-HBV-C-Gluc; the PGA1-empty plasmid does not carry exogenous genes;
s24) carrying out enzyme digestion on shuttle plasmid PGA1-HBV-C-Gluc and adenovirus plasmid AD5, recovering enzyme digestion products for transfection, and collecting recombinant adenovirus plasmid AD5-HBV-C-Gluc after transfection.
10. The system of any one of claims 1 to 5, and/or the use of HBV cccDNA of claim 6 in the construction of HBV drug screening system.
CN201910524757.3A 2019-06-18 2019-06-18 System for inducing formation of HBV cccDNA and construction method Pending CN110669735A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201910524757.3A CN110669735A (en) 2019-06-18 2019-06-18 System for inducing formation of HBV cccDNA and construction method

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201910524757.3A CN110669735A (en) 2019-06-18 2019-06-18 System for inducing formation of HBV cccDNA and construction method

Publications (1)

Publication Number Publication Date
CN110669735A true CN110669735A (en) 2020-01-10

Family

ID=69068753

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910524757.3A Pending CN110669735A (en) 2019-06-18 2019-06-18 System for inducing formation of HBV cccDNA and construction method

Country Status (1)

Country Link
CN (1) CN110669735A (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111705080A (en) * 2020-05-14 2020-09-25 广州市第八人民医院 Construction method and application of HBV non-human animal model
CN113025651A (en) * 2021-03-31 2021-06-25 重庆医科大学 Drug screening cell model of targeted HBV core promoter, Triciribine and new application of structural analogue
WO2021259345A1 (en) * 2020-06-24 2021-12-30 厦门大学 Drug screening model and method for targeting hbv cccdna

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102888426A (en) * 2011-07-19 2013-01-23 中国科学院深圳先进技术研究院 Minicircle gene vector and its preparation method and use
WO2016070008A1 (en) * 2014-10-30 2016-05-06 The University Of North Carolina At Chapel Hill Hepatitis b reporter viruses and cccdna-based expression vectors
US20170327797A1 (en) * 2015-01-27 2017-11-16 Hoffmann-La Roche Inc. Recombinant hbv cccdna, the method to generate thereof and the use thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102888426A (en) * 2011-07-19 2013-01-23 中国科学院深圳先进技术研究院 Minicircle gene vector and its preparation method and use
WO2016070008A1 (en) * 2014-10-30 2016-05-06 The University Of North Carolina At Chapel Hill Hepatitis b reporter viruses and cccdna-based expression vectors
US20170327797A1 (en) * 2015-01-27 2017-11-16 Hoffmann-La Roche Inc. Recombinant hbv cccdna, the method to generate thereof and the use thereof

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
QI Z等: "Recombinant covalently closed circular hepatitis B virus DNA induces prolonged viral persistence in immunocompetent mice", 《J VIROL》 *
QI Z等: "Recombinant covalently closed circular hepatitis B virus DNA induces prolonged viral persistence in immunocompetent mice", 《J VIROL》, vol. 88, no. 14, 7 May 2014 (2014-05-07), pages 8045 - 8056, XP055268374, DOI: 10.1128/JVI.01024-14 *
无: "Accession.KC875263.1, Hepatitis B virus isolate HBV61 genotype C, complete genome", 《GENBANK》 *
朱园飞 等: "应用微环DNA技术体外诱导和制备重组的乙型肝炎病毒共价闭合环状DNA", 《微生物与感染》 *
朱园飞 等: "应用微环DNA技术体外诱导和制备重组的乙型肝炎病毒共价闭合环状DNA", 《微生物与感染》, vol. 12, no. 4, 25 August 2017 (2017-08-25), pages 229 - 234 *
王超: "《基因修饰小鼠制备常用技术》", 31 December 2013, 北京:中国农业大学出版社, pages: 124 - 125 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111705080A (en) * 2020-05-14 2020-09-25 广州市第八人民医院 Construction method and application of HBV non-human animal model
WO2021259345A1 (en) * 2020-06-24 2021-12-30 厦门大学 Drug screening model and method for targeting hbv cccdna
CN113025651A (en) * 2021-03-31 2021-06-25 重庆医科大学 Drug screening cell model of targeted HBV core promoter, Triciribine and new application of structural analogue

Similar Documents

Publication Publication Date Title
CN107502608B (en) Construction method and application of sgRNA and ALDH2 gene-deleted cell strain for knocking out human ALDH2 gene
KR102061357B1 (en) HBV polymerase mutants
EP1801209B1 (en) Modified human hepatitis c virus genomic rna having autonomous replicative competence
Sureau et al. Cloned hepatitis delta virus cDNA is infectious in the chimpanzee
CN110669735A (en) System for inducing formation of HBV cccDNA and construction method
CN108697746A (en) Encode the virus of anti-tcr recombinant antibody or segment
KR20080021659A (en) Vaccine composition
CN107164409B (en) Canine distemper virus sensitive cell line SLAM-MDCK and construction method and application thereof
CN112353939B (en) Application of GTPBP4 protein as immunosuppressant and construction of cell line for knocking down or over expressing GTPBP4
CN111705080A (en) Construction method and application of HBV non-human animal model
CN113897394A (en) Recombinant adenovirus vaccine for African swine fever and construction method thereof
JP6821780B2 (en) Hepatitis B therapeutic vaccine with inactivated fully recombinant Hansenula polymorpha cells expressing HBsAg
CN116463297A (en) Recombinant serum type 4 avian adenovirus expressing chicken infectious anemia virus VP1 protein and preparation method thereof
CN112011518A (en) Construction and application of Zika virus ZG01 strain reverse genetic system
CN111793721B (en) Application of eEF1D protein in preparation of drugs for preventing or treating foot-and-mouth disease virus infection
CN117511888A (en) Recombinant serum type 4 avian adenovirus expressing chicken infectious anemia virus T1P6 strain VP2 protein based on CRISPR-Cas9 technology and preparation method thereof
CN111704674B (en) Chimeric antigen receptor targeting c-Met and autocrine PD-L1 scFv and application thereof
CN109453366B (en) Preparation method and application of anti-tumor protein
CN111593068B (en) Preparation method and application of novel gene therapy vector pIRES-Rsirt2/4-Tet-nap
CN107034232B (en) Application of ATG16L1 gene in enhancing replication capacity of Newcastle disease virus
CN106148404B (en) Human hepatitis B virus recombinant vector and application thereof
WO2022142267A1 (en) Nucleic acid, vector and host cell for preparing hbv cccdna
CN113862287B (en) 3a type hepatitis C virus full-length infectious clone mutant and application thereof
CN113897393B (en) Recombinant adenovirus vaccine for African swine fever and construction method thereof
CN106075396A (en) The application of Cul4 albumen

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