CN108085331B - DNA framework for circular RNA overexpression and application thereof - Google Patents

DNA framework for circular RNA overexpression and application thereof Download PDF

Info

Publication number
CN108085331B
CN108085331B CN201711428211.5A CN201711428211A CN108085331B CN 108085331 B CN108085331 B CN 108085331B CN 201711428211 A CN201711428211 A CN 201711428211A CN 108085331 B CN108085331 B CN 108085331B
Authority
CN
China
Prior art keywords
sequence
overexpression
dna
circrna
circ
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
CN201711428211.5A
Other languages
Chinese (zh)
Other versions
CN108085331A (en
Inventor
罗卫峰
张腾
谭圆圆
吴翠芳
韩慧
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.)
Zhejiang free trade zone ruicai biological medicine technology Co., Ltd.
Original Assignee
Zhejiang Free Trade Zone Ruisai Biomedical Technology Co ltd
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 Zhejiang Free Trade Zone Ruisai Biomedical Technology Co ltd filed Critical Zhejiang Free Trade Zone Ruisai Biomedical Technology Co ltd
Priority to CN201711428211.5A priority Critical patent/CN108085331B/en
Publication of CN108085331A publication Critical patent/CN108085331A/en
Application granted granted Critical
Publication of CN108085331B publication Critical patent/CN108085331B/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
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • 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
    • 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
    • C12N15/67General methods for enhancing the expression
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • 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
    • C12N2330/00Production
    • C12N2330/50Biochemical production, i.e. in a transformed host cell
    • C12N2330/51Specially adapted 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
    • 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

Landscapes

  • Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Virology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The invention relates to a DNA frame for circular RNA overexpression and application thereof, wherein the artificially synthesized DNA frame sequence is inserted into eukaryotic expression vectors, lentiviral vectors, adenoviral vectors and adeno-associated viral vectors to form a circRNA special expression vector. The DNA framework sequence and the corresponding expression vector thereof can be widely applied to the expression of various circRNAs, and provide a favorable tool for researching the function and mechanism of the circRNAs and further developing gene therapy medicaments based on the circRNAs.

Description

DNA framework for circular RNA overexpression and application thereof
[ technical field ]
The present invention relates to a DNA framework for the overexpression of circular RNA and uses thereof.
[ background art ]
Circular RNA (circular RNA) is a new class of RNA distinguished from conventional linear RNA by not having the 5 'terminal cap and 3' terminal poly (a) tail of conventional linear RNA, but existing as a closed circular structure. Research shows that the circular RNA is mainly generated through atypical variable shearing processing, widely exists in various biological cells, has the characteristics of stable structure, difficult degradation by RNA enzyme, high expression abundance, good conservation among species, tissue and space specificity and the like. Research on circRNA and diseases shows that the circRNA is related to the occurrence and the progression of diseases such as tumors, neurodevelopmental diseases, atherosclerosis, myotonic dystrophy and the like. In addition, research finds that stable circRNA can be detected in human saliva and blood, and the characteristics enable the circular RNA to have wide prospects in development and application of novel disease diagnosis and treatment methods.
Research on circRNA is not deep, and the biological function and regulation mechanism of circRNA are still unknown. Although the existing research suggests that circRNA can be used as "sponge" to adsorb microRNA in cells and block the inhibition effect of the microRNA on target genes of the microRNA; the circRNA can also directly regulate and control other RNA levels through base complementary pairing; in addition, research suggests that circRNA may have the functions of binding with protein, regulating protein activity and the like. However, the biological activity of circRNA and its mechanism of action in the cell requires extensive research.
As with conventional gene function and mechanism studies, an important research approach to study the biological function and mechanism of circRNA is to overexpress circRNA intracellularly, observe its effects on cellular function and elucidate its molecular mechanisms involved in regulating cellular function. Then, a stable and reliable means for circRNA overexpression is necessary for intracellular introduction of overexpressed circRNA. The existing method for over-expressing circular RNA usually uses genome DNA as a template, a target circRNA sequence and sequences of 1000bp upstream and 200bp downstream are amplified, then a reverse complementary sequence of 800bp upstream is inserted into the downstream by means of PCR splicing, and then the obtained complete sequence is inserted into a commercial over-expression vector such as pCDNA3.1 by means of enzyme digestion connection to express the target circRNA. This method is not suitable for the overexpression of artificially designed circular RNAs. In 2014, Liang optimally constructs a circRNA expression frame by utilizing upstream and downstream sequence research on hsa _ circ _0001727, but the expression frame has limitations, is only effective for overexpression of endogenous circRNA, and has low overexpression efficiency.
[ summary of the invention ]
The invention aims to provide a DNA sequence suitable for circRNA expression and a vector thereof, wherein the sequence is generally suitable for the expression of various circRNAs, and has high and stable expression efficiency; the sequence can be integrated into various expression vectors, and can be applied to various expression systems such as common eukaryotic expression, lentivirus expression, adenovirus expression, adeno-associated virus expression, retrovirus expression and the like; the circRNA expression by using the vector containing the sequence is simple and easy to operate and easy to popularize.
In order to achieve the above object, a DNA framework for the overexpression of circular RNAs of the sequence of formula I was invented,
US-[N]n-DS
I
wherein the content of the first and second substances,
US is an upstream sequence shown as SEQ ID NO.1,
DS is a downstream sequence shown as SEQ ID NO.2,
[N]nas an intermediate sequence, N represents A, T, G, C any one of four deoxyribonucleotides, and N identical or different N are connected in sequence to form the intermediate sequence.
The above-described DNA framework for circular RNA overexpression also has the following optimized structure:
said [ N ]]nPreferably the linear DNA sequence of the circRNA to be expressed. Further preferred are linear DNA sequences of endogenous circRNA, further comprising linear DNA sequences of hsa _ circ _0001727, hsa _ circ _0001756, hsa _ circ _0000284, hsa _ circ _0000711 or hsa _ circ _ 0000268.
The optimal range of n is more than or equal to 150 and less than or equal to 3000.
The invention also comprises a DNA framework for the overexpression of the circular RNA, and the DNA sequence of the DNA framework is shown as SEQ ID NO. 4.
The invention also comprises an expression vector which is formed by inserting the DNA framework sequence for the circular RNA overexpression into a eukaryotic expression vector, a lentiviral vector, an adenoviral vector and an adeno-associated viral vector.
The DNA framework for circular RNA overexpression of the invention can be used for overexpression of circRNA.
The circRNA overexpression DNA framework sequence can be integrated into various expression vectors, and can be applied to various expression systems such as common eukaryotic expression, lentivirus expression, adenovirus expression, adeno-associated virus expression, retrovirus expression and the like.
A plurality of DNA sequences are designed in the experimental process, and reliable experimental results prove that the circRNA overexpression DNA framework sequence related to the invention has simple design, various expression vector choices, simple and convenient vector construction operation and stable and efficient circRNA overexpression (the circRNA overexpression multiple is more than hundreds of times). The sequence and the corresponding vector can be widely applied to the expression of various circRNAs, and a powerful tool is provided for the function and mechanism research of the circRNAs.
[ description of the drawings ]
FIG. 1 is a schematic representation of the universal overexpression framework for circular RNA in the examples.
FIG. 2 is a diagram showing the overexpression effect of the circRNAs after 293T cells are transfected by a plurality of circRNA overexpression plasmids constructed by the invention.
[ detailed description of the invention ]
The present invention is further explained with reference to the following examples, which are not intended to limit the present invention in any way.
In this example, the GT-AG rule of eukaryotic gene splicing was used, and based on the upstream and downstream flanking sequences of hsa _ circ _0001727, the flanking inverted repeat sequences were simplified and optimized, respectively, and optimized and designed simultaneously on both sides of the 5 'and 3' cleavage sites to obtain a universal circRNA overexpression framework (upstream sequence + middle sequence + downstream sequence). According to the general over-expression frame sequence design principle, a plurality of circRNA over-expression frame DNA sequences are designed and synthesized by a chemical synthesis mode, and then the artificial circular RNA over-expression frame sequences are constructed on commercial expression vectors (eukaryotic expression vectors, lentivirus vectors, adenovirus vectors and the like) by an enzyme digestion linking mode. Then, the constructed circRNA overexpression vector is transfected into 293T cells, and the overexpression efficiency of the circRNA overexpression framework is verified.
The technology related by the invention is conventional technical means such as gene synthesis, vector construction, cell culture, plasmid transfection, qPCR and the like, wherein related enzymes, primers, reagents, cell culture reaction conditions and cell transfection conditions can be reasonably selected according to the experience of technicians in the field under the condition of no description, related reagent consumables belong to common products sold in the market, and cells are from ATCC. The means and instruments involved are well known and well understood by those skilled in the art.
The technical solutions of the present invention are further illustrated by the following examples and test examples, but should not be construed as limiting the present invention.
The first embodiment is as follows: construction of has _ circ _0001727 overexpression vector
Hsa _ circ _0001727 overexpression framework sequence design:
linear DNA sequences encoding hsa _ circ _0001727 circular RNA were obtained from a circBase database query. The sequence is shown as SEQ ID NO. 3.
According to the above general design principle of circRNA overexpression DNA framework sequence, the inquired DNA sequence of hsa _ circ _0001727 is replaced by [ N ] in the sequence of formula I]nObtaining the special overexpression frame DNA sequence of hsa _ circ _0001727, and the sequence is shown in SEQ ID NO. 4.
Still further, in other embodiments, as shown in fig. 1, N may also be a multiple of 2, 3, 4, 5, 10, 20, 30, 40, 50, 100, 200, 300, 400, 500, 1000, 2000, 3000, or even more, which may be the same N or a different N.
2. The DNA sequence of hsa _ circ _0001727 for the proprietary overexpression framework was submitted to Kinshire, Nanjing for sequence synthesis.
3. Primer design Synthesis
Primer design was performed using Primer5 software:
Circ-F: shown as SEQ ID NO. 5.
Circ-R: as shown in SEQ ID NO. 6.
The primer sequences were synthesized by Shanghai Huada Gene.
PCR amplification of the specialized overexpression framework DNA sequence of hsa _ circ _0001727
And (3) taking a special overexpression frame DNA sequence of hsa _ circ _0001727 obtained by whole gene synthesis as a template, and carrying out PCR amplification on a target fragment, wherein an amplification system is as follows:
10×Buffer 10ul
MgSO4(50mM) 1ul
dNTP(10mM) 1.5ul
transStart Fastpfu DNA polymerase(5U/ul) 0.5ul
Circ-F(10uM) 2ul
Circ-R(10uM) 2ul
template DNA (50ng/ul) 1ul
Add ddH2To total volume of O 50ul
The PCR cycling program was as follows:
Figure GDA0002981525310000051
Figure GDA0002981525310000061
PCR product recovery
The PCR products were electrophoresed on agarose gel and recovered using a gel recovery kit (Axygen, AP-GX-50) following the detailed protocol of the kit instructions.
PCR recovery of Nhe I and HindIII products and recovery of products
Performing double enzyme digestion on the PCR recovery product by using Asc I and Pme I, wherein the enzyme digestion system is as follows:
10×Buffer 3ul
PCR product/plasmid of interest 1ug
Restriction enzyme Nhe I 0.5ul
Restriction enzyme Hind III 0.5ul
Complement ddH20 to total volume 30ul
After enzyme digestion for 4-5 hours at 37 ℃, the enzyme digestion fragments are separated by electrophoresis, and the target fragments are recovered by cutting gel.
pCDNA3.1 plasmid Nhe I and Hind III double digestion and digestion product recovery
Eukaryotic expression plasmid pCDNA3.1 was digested simultaneously with Nhe I and HindIII as follows:
10×Buffer 3ul
PCR product/plasmid of interest 1ug
Restriction enzyme Nhe I 0.5ul
Restriction enzyme Hind III 0.5ul
Complement ddH20 to total volume 30ul
After enzyme digestion for 4-5 hours at 37 ℃, enzyme digestion fragments are separated by electrophoresis, and linearized plasmids are recovered by gel cutting.
8. The target fragment was ligated to the linearized pCDNA3.1 plasmid
T4 DNA ligase is used for connecting the PCR product after enzyme digestion and a lentiviral vector, and the connection system is as follows:
Figure GDA0002981525310000062
Figure GDA0002981525310000071
the ligation was performed at room temperature for 1 h. At the same time, negative control is carried out, and water is used for replacing the gene to be connected with the vector.
9. Conversion of ligation products
1) The ligation products were added separately to 50. mu.l Tran 5. alpha. competent cells in an ice bath. Mix by gentle rotation and ice-bath for 30 min.
2) The water bath is heat-shocked for 90s at 42 ℃.
3) The tube was quickly transferred to an ice bath for 2 min.
4) Adding 500 μ l LB culture medium respectively, mixing, and shaking culturing at 37 deg.C and 150g for 40 min.
5) 150ul of the bacterial solution was applied to the surface of an LB plate containing ampicillin (Amp) (100. mu.g/ml), and left at room temperature until the liquid was absorbed. The plate was inverted and transferred to a 37 ℃ biochemical incubator overnight.
10. Positive clone PCR identification
The next day after transformation, single colony was picked for colony PCR, and PCR amplification system and cycle procedure were as follows: the amplification system was as follows:
10×Reaction Buffer 1.5ul
MgCl2(25mM) 1.5ul
dNTPs(10mM) 0.5ul
Circ F(10mM) 0.5ul
Circ R(10mM) 0.5ul
Taq(5U/ul) 0.1ul
bacterial colony suspension 1ul
Complement ddH20 to total volume 15ul
The cycling conditions were as follows:
Figure GDA0002981525310000072
11. positive clone shake bacteria and plasmid extraction
The clones which are positive in PCR identification are cultured in 1ml LB liquid culture medium containing corresponding antibiotics, the bacteria are shaken overnight at 37 ℃, and plasmids are extracted by utilizing a plasmid miniprep kit (Axygen, AP-MN-P-50) on the next day according to the detailed steps of the instruction.
12. And (3) sending the extracted plasmid to Huada gene for sequencing, and performing sequence comparison on a sequencing result. The constructed specific overexpression plasmid for hsa _ circ _0001727 was obtained.
Example two: qPCR detection of the overexpression Effect of hsa _ circ _0001727 in 293T cells transfected with hsa _ circ _0001727 overexpression plasmid
1, qPCR primer design and Synthesis
divergent primer-F:CAGGTCAAGTTCATGGACCTG
divergent primer-R:ATTCAGACTTACCTGAAGTA
The primer sequences were synthesized by Shanghai Huada Gene.
2, before transfectionDigesting 293T cells in logarithmic growth phase with pancreatin for 24h, and subculturing the cells to a six-well plate, at 37 ℃ and 5% CO2Culturing in an incubator. Can be used for transfection after the cell density reaches 70-80% in 24 h.
3, the cell culture medium was changed to serum-free medium before transfection.
4, add 25ug of the prepared hsa _ circ _0001727 overexpression plasmid DNA solution to a sterilized centrifuge tube, mix well with the corresponding volume of Opti-MEM, and adjust the total volume to 1.5 ml.
5, Lipofectamine 2000 was gently shaken, and 60. mu.l of Lipofectamine 2000 was mixed with 1.5ml of Opti-MEM in another tube and incubated at room temperature for 5 minutes.
6, the diluted DNA and the diluted Lipofectamine 2000 were mixed, and the mixture was gently inverted and mixed without shaking.
7, transferring the mixture of DNA and Lipofectamine 2000 to 293T cell culture medium, mixing, and culturing at 37 deg.C with 5% CO2Culturing in a cell culture box.
8, after 6 hours of culture, the medium containing the transfection mixture was aspirated, 10ml of cell culture medium containing 10% serum was added to each flask of cells, and the mixture was incubated at 37 ℃ with 5% CO2The incubator was allowed to incubate for 48 hours.
9, adding 1ml of Trizol into each hole of a 6-hole plate, repeatedly blowing and beating for 10 times by using a 1ml gun head, and collecting the Trizol into an EP (EP) tube; centrifuge at 12000g for 15 min and collect the supernatant.
10, 200ul of chloroform was added to the supernatant, the mixture was mixed by inverting the upper and lower forces for half a minute, and the mixture was left to stand for 3 minutes.
Centrifugation at 12000g for 15 minutes at 11,4 ℃ gave a lysate which was visualized as three layers: the upper layer is RNA of water phase; the middle layer is DNA, lipid, etc.; the lower layer is cell residue, protein, polysaccharide, etc.
12, taking 500ul of the supernatant into a new EP tube, and sucking 167ul of the supernatant for three times; adding isopropanol with the same volume, mixing uniformly, standing for 10 minutes, and centrifuging at 12000g for 10 minutes at 4 ℃.
13, carefully remove the supernatant, take care not to lose the RNA pellet, add 1ml 75% ethanol, upside down, and resuspend the pellet.
Centrifugation at 12000g for 10min at 14,4 ℃ takes care to remove the supernatant, aspirates the wall as dry as possible, takes care not to lose the RNA pellet, and can be centrifuged again if the pellet is loose. Air-dry for about 15 minutes until the tube wall is free of liquid.
15, adding a proper volume (20-30ul) of DEPC water to dissolve the RNA, and carrying out water bath at 58 ℃ for 10 minutes.
16, 2ul of the quantification was removed, buffer was measured at 10mM TrisCl (pH7.8), and reverse transcription was performed based on the quantification result. (1A 260. mu.g/ml, A260/A280. mu.g/ml, 1.8. mu.g/ml to 2.1.)
17, reverse transcription of RNA
Reverse transcription was performed according to the protocol as follows:
add to RNase-Free PCR tube (20 ul total)
RNA 3μg
DEPC·H 2O Make up to 11.0
Mixing, incubating at 65 deg.C for 10min, immediately ice-cooling, and adding
2.5U/μl Poly A Polymerase 1μl
RTase Mix 1μl
5×PAP/RT Buffer 5μl
dd H2O(RNase/Dnase free) 8μl
Incubating at 37 deg.C for 60min, 85 deg.C for 5 min; the cDNA was frozen at-20 ℃ or immediately subjected to PCR.
18, qPCR assay
1) Under the premise of finding out the optimal primer annealing temperature and template amount in an RT-PCR pre-experiment, 2 xSYBR Green Mix is used for preparing the PCR Mix, and the PCR Mix is calculated and prepared according to the number and the repetition number of samples needing to be loaded on a computer, wherein the system is as follows:
2×SYBR Green Mix 10μl
qPCR primer Mix 1μl
Form panel 5μl
Ultrapure water 4μl
Total volume 20μl
2) Subpackaging to PCR8 connecting tubes, and instantly centrifuging and mixing the PCR system by a miniature centrifuge.
3) Putting the sample into an IQ5 fluorescent quantitative PCR instrument, carrying out SYBR Green fluorescent quantitative PCR to analyze the expression of each gene, and setting the PCR program as follows:
the PCR reaction can be carried out in 3 steps: (the annealing temperature is set by itself according to the Tm value of the primer and the result of the RT-PCR preliminary experiment, and the melting curve can be set to 60-95 ℃.)
Pre-denatured Cycle 1 (1X)
Step 1:95.0℃for 02:00.
PCR Cycle 2 (40X)
Step 1:95.0℃for 00:15.
Step 2:60.0℃for 00:20.
Step 3:72.0℃for 00:20.
Data collection and real-time analysis enabled.
Dissolution curve Cycle 3 (71X)
Step 1:60.0℃-95.0℃for 00:30.
Increase set point temperature after cycle 2 by 0.5℃
Melt curve data collection and analysis enabled.
19, qPCR relative quantitative results analysis
The calculation formula of the relative expression quantity of the target gene is as follows:
Figure GDA0002981525310000111
the Ct target is the Ct value of the target gene, and the Ct internal reference is the Ct value of the housekeeping gene. Delta Ct (CtObjective-CtEngineer) represents the relative Ct value of the objective gene of each sample relative to housekeeping gene, delta Ct (delta Ct) Test- (. DELTA Ct) Control represents the normalization of the treated group relative to the Control group,
Figure GDA0002981525310000112
the expression level of the treated group relative to the control group is shown, and the relative expression fold of the target gene is shown.
Overexpression vectors of has _ circ _0001756, has _ circ _0000284, has _ circ _0000711 or has _ circ _0000268 were constructed in the same manner as described above, and the expression effect in 293T cells after transfection of these overexpression plasmids was examined by qPCR. The effect diagram is shown in fig. 1.
Sequence listing
<110> Shanghai Ruishi Biotechnology Ltd
<120> DNA framework for circular RNA overexpression and use thereof
<160> 8
<170> SIPOSequenceListing 1.0
<210> 1
<211> 89
<212> DNA
<213> artificial sequence
<400> 1
tgagattaca ggtgtgagcc accacccccg gcctcacttt ttgtaaaggt acgtactaat 60
gacttttttt ttatacttca ggtaagtct 89
<210> 2
<211> 70
<212> DNA
<213> artificial sequence
<400> 2
tctctctctc ttcaggtaag tagcaaggaa aagagttagg cccggcacgg tagctcacac 60
ctgtaatccc 70
<210> 3
<211> 889
<212> DNA
<213> artificial sequence
<400> 3
gaatagtaaa gaaacacatc ataaaacctc ccaggacata aaggtgagca cagaccctgt 60
ttggatcaag tcagttcctg gagcctgaat gatgactgct gaatcacggg aagccacggg 120
tctgtcccca caggctgcac aggagaagga tggtatcgta atagtgaagg tggaagagga 180
agatgaggaa gaccacatgt gggggcagga ttccacccta caggacacgc ctcctccaga 240
cccagagata ttccgccaac gcttcaggcg cttctgttac cagaacactt ttgggccccg 300
agaggctctc agtcggctga aggaactttg tcatcagtgg ctgcggccag aaataaacac 360
caaggaacag atcctggagc ttctggtgct agagcagttt ctttccatcc tgcccaagga 420
gctccaggtc tggctgcagg aataccgccc cgatagtgga gaggaggccg tgacccttct 480
agaagacttg gagcttgatt tatcaggaca acaggtaaaa agaggtgaaa cctattatgt 540
gtgagcaggg cacagacgtt gaaactggag ccaggagaag tattggcagg ctttaggtta 600
ttaggtggtt actctgtctt aaaaatgttc tggctttctt cctgcatcca ctggcatact 660
catggtctgt ttttaaatat tttaattccc atttacaaag tgatttaccc acaagcccaa 720
cctgtctgtc ttcaggtccc aggtcaagtt catggacctg agatgctcgc aagggggatg 780
gtgcctctgg atccagttca ggagtcctcg agctttgacc ttcatcacga ggccacccag 840
tcccacttca aacattcgtc tcggaaaccc cgcctcttac agtcacgag 889
<210> 4
<211> 1048
<212> DNA
<213> artificial sequence
<400> 4
tgagattaca ggtgtgagcc accacccccg gcctcacttt ttgtaaaggt acgtactaat 60
gacttttttt ttatacttca ggtaagtctg aatagtaaag aaacacatca taaaacctcc 120
caggacataa aggtgagcac agaccctgtt tggatcaagt cagttcctgg agcctgaatg 180
atgactgctg aatcacggga agccacgggt ctgtccccac aggctgcaca ggagaaggat 240
ggtatcgtaa tagtgaaggt ggaagaggaa gatgaggaag accacatgtg ggggcaggat 300
tccaccctac aggacacgcc tcctccagac ccagagatat tccgccaacg cttcaggcgc 360
ttctgttacc agaacacttt tgggccccga gaggctctca gtcggctgaa ggaactttgt 420
catcagtggc tgcggccaga aataaacacc aaggaacaga tcctggagct tctggtgcta 480
gagcagtttc tttccatcct gcccaaggag ctccaggtct ggctgcagga ataccgcccc 540
gatagtggag aggaggccgt gacccttcta gaagacttgg agcttgattt atcaggacaa 600
caggtaaaaa gaggtgaaac ctattatgtg tgagcagggc acagacgttg aaactggagc 660
caggagaagt attggcaggc tttaggttat taggtggtta ctctgtctta aaaatgttct 720
ggctttcttc ctgcatccac tggcatactc atggtctgtt tttaaatatt ttaattccca 780
tttacaaagt gatttaccca caagcccaac ctgtctgtct tcaggtccca ggtcaagttc 840
atggacctga gatgctcgca agggggatgg tgcctctgga tccagttcag gagtcctcga 900
gctttgacct tcatcacgag gccacccagt cccacttcaa acattcgtct cggaaacccc 960
gcctcttaca gtcacgagtc tctctctctt caggtaagta gcaaggaaaa gagttaggcc 1020
cggcacggta gctcacacct gtaatccc 1048
<210> 5
<211> 29
<212> DNA
<213> artificial sequence
<400> 5
ttagctagct gagattacag gtgtgagcc 29
<210> 6
<211> 30
<212> DNA
<213> artificial sequence
<400> 6
gctaagcttg ggattacagg tgtgagctac 30
<210> 7
<211> 21
<212> DNA
<213> Artificial Sequence
<400> 7
caggtcaagt tcatggacct g 21
<210> 8
<211> 20
<212> DNA
<213> Artificial Sequence
<400> 8
attcagactt acctgaagta 20

Claims (7)

1. A DNA framework with a sequence shown in a formula I and used for circular RNA overexpression,
US-[N]n-DS
I
wherein the content of the first and second substances,
US is an upstream sequence, the nucleotide sequence of which is shown as SEQ ID NO.1,
DS is a downstream sequence, the nucleotide sequence of the DS is shown as SEQ ID NO.2,
[N]nfor the intermediate sequence, N represents A, T, G, C any one of four deoxyribonucleotides, N is the same or different and are connected in sequence to form the intermediate sequence, and the [ N ] is]nIs the sequence of circRNA.
2. The DNA framework for overexpression of a circular RNA as claimed in claim 1, characterized in that [ N]nIs the sequence of endogenous circRNA.
3. The DNA framework for circular RNA overexpression of claim 2, characterized in that the sequence of the endogenous circRNA is hsa _ circ _0001727, hsa _ circ _0001756, hsa _ circ _0000284, hsa _ circ _0000711 or hsa _ circ _ 0000268.
4. The DNA framework for overexpression of a circular RNA as claimed in claim 1, characterized in that 150. ltoreq. n.ltoreq.3000.
5. A DNA framework for the overexpression of circular RNA, characterized in that the DNA sequence is shown in SEQ ID NO. 4.
6. An expression vector which is a eukaryotic expression vector, a lentiviral vector, an adenoviral vector or an adeno-associated viral vector, characterized in that it carries a DNA framework sequence according to any one of claims 1 to 5 for the overexpression of circular RNA.
7. Use of a DNA framework for circular RNA overexpression according to any of claims 1 to 5 for the in vitro overexpression of circRNA.
CN201711428211.5A 2017-12-26 2017-12-26 DNA framework for circular RNA overexpression and application thereof Active CN108085331B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201711428211.5A CN108085331B (en) 2017-12-26 2017-12-26 DNA framework for circular RNA overexpression and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201711428211.5A CN108085331B (en) 2017-12-26 2017-12-26 DNA framework for circular RNA overexpression and application thereof

Publications (2)

Publication Number Publication Date
CN108085331A CN108085331A (en) 2018-05-29
CN108085331B true CN108085331B (en) 2021-06-25

Family

ID=62179408

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201711428211.5A Active CN108085331B (en) 2017-12-26 2017-12-26 DNA framework for circular RNA overexpression and application thereof

Country Status (1)

Country Link
CN (1) CN108085331B (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112852946A (en) * 2021-01-15 2021-05-28 中国农业大学 Diagnostic biomarker and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105087570A (en) * 2015-09-07 2015-11-25 广州吉赛生物科技有限公司 Annular RNA artificial over-expression frame as well as expression vector and construction method thereof
CN105176981A (en) * 2015-09-17 2015-12-23 广州永诺生物科技有限公司 DNA (deoxyribonucleic acid) sequence used for circular RNA (ribonucleic acid) expression, expression vector and applications of DNA sequence and expression vector
CN107058360A (en) * 2017-04-04 2017-08-18 杨展 A kind of circular rna expression vector establishment method and its application based on quick clone technology

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105087570A (en) * 2015-09-07 2015-11-25 广州吉赛生物科技有限公司 Annular RNA artificial over-expression frame as well as expression vector and construction method thereof
CN105176981A (en) * 2015-09-17 2015-12-23 广州永诺生物科技有限公司 DNA (deoxyribonucleic acid) sequence used for circular RNA (ribonucleic acid) expression, expression vector and applications of DNA sequence and expression vector
CN107058360A (en) * 2017-04-04 2017-08-18 杨展 A kind of circular rna expression vector establishment method and its application based on quick clone technology

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Short intronic repeat sequences facilitate circular RNA production;Dongming Liang等;《GENES & DEVELOPMENT》;20141003;第28卷(第20期);第2233-2247页,参见全文 *
ZKSCAN1 gene and its related circular RNA (circZKSCAN1) both inhibit hepatocellular carcinoma cell growth, migration, and invasion but through different signaling pathways;Zhicheng Yao等;《Molecular Oncology》;20170317;第11卷(第4期);第422-437页,参见全文 *
环状RNA mmu-circ-Pax3.1表达载体的构建及初步验证;曹洋等;《中国畜牧兽医》;20171031;第44卷(第10期);第2865-2870页,参见全文 *

Also Published As

Publication number Publication date
CN108085331A (en) 2018-05-29

Similar Documents

Publication Publication Date Title
CN108165549B (en) Universal expression framework of artificial circular RNA and application thereof
CN107502608A (en) Construction method and application for sgRNA, ALDH2 gene delection cell line for knocking out people&#39;s ALDH2 genes
CN109750035B (en) sgRNA for targeting and guiding Cas9 protein to efficiently cleave TCR and B2M gene locus
CN106754904A (en) A kind of specific molecular label of cDNA and its application
CN108018301B (en) Method for determining core promoter of miR-27a gene and binding site of transcription factor Myod thereof
CN107663539B (en) Use of circular RNA circ-PTGR1
Cornelissen et al. From clinical sample to complete genome: Comparing methods for the extraction of HIV-1 RNA for high-throughput deep sequencing
CN109182562B (en) miRNA apla-mir-25-42 related to follicular development of laying ducks as well as detection primer, inhibitor and application thereof
CN109735541B (en) ACADSB gene knockout dairy cow mammary gland epithelial cell line and construction method thereof
CN108085331B (en) DNA framework for circular RNA overexpression and application thereof
WO2015067089A1 (en) Method for introducing exogenous mitochondria into mammalian cells
Cardinali et al. Time-controlled and muscle-specific CRISPR/Cas9-mediated deletion of CTG-repeat expansion in the DMPK gene
CN109487005A (en) For expanding the primer of the intranasal tumour virus whole genome sequence of goat region
CN109486816B (en) Polynucleotide for treating tumor and its application
CN116694603A (en) Novel Cas protein, crispr-Cas system and use thereof in the field of gene editing
CN110747230A (en) Method for promoting bovine skeletal muscle satellite cell myogenic differentiation
CN110804621A (en) Preparation method of escherichia coli extracellular vesicle with endogenous high-expression miRNA (micro ribonucleic acid)
CN109456995A (en) Gene knockout plasmid, cell line and preparation method and application
US20240011055A1 (en) Precise genome deletion and replacement method based on prime editing
Hagedorn et al. Handling S/MAR Vectors
CN110229816B (en) Construction method and application of sgRNA (ribonucleic acid) for knocking out RBP4 gene and RBP4 gene deletion cell strain
CN106834293B (en) Circular RNA with molecular marker and preparation method and application thereof
CN111454944B (en) Method for synthesizing separated RNA and DNA template thereof
CN111197070A (en) Method for identifying 2&#39; -O-methylation modification in RNA molecule and application thereof
CN111411162B (en) Method for rapidly detecting enterobacter shenghuai strain level

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: 20180904

Address after: Room 1501-312, Putuo Chamber of Commerce Building, 66 Shatian Street, Donggang Street, Putuo District, Zhoushan City, Zhejiang Province

Applicant after: Zhejiang free trade zone ruicai biological medicine technology Co., Ltd.

Address before: 201315 room 1701, 25 building, 3399 lane, Kang Xin Road, Pudong New Area, Shanghai.

Applicant before: Shanghai Rui Sai Bioisystech Co., Ltd

GR01 Patent grant
GR01 Patent grant