CN114908089A - Construction method and application of 3' UTR - Google Patents

Construction method and application of 3' UTR Download PDF

Info

Publication number
CN114908089A
CN114908089A CN202110172382.6A CN202110172382A CN114908089A CN 114908089 A CN114908089 A CN 114908089A CN 202110172382 A CN202110172382 A CN 202110172382A CN 114908089 A CN114908089 A CN 114908089A
Authority
CN
China
Prior art keywords
gene
sequence
utr
mrna
polynucleotide
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.)
Granted
Application number
CN202110172382.6A
Other languages
Chinese (zh)
Other versions
CN114908089B (en
Inventor
刘韬
王猛
高海霞
邵梅琪
蒋婷
王佩
钱其军
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.)
Shanghai Cell Therapy Group Co Ltd
Original Assignee
Shanghai Cell Therapy Group 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 Shanghai Cell Therapy Group Co Ltd filed Critical Shanghai Cell Therapy Group Co Ltd
Priority to CN202110172382.6A priority Critical patent/CN114908089B/en
Priority to PCT/CN2022/074391 priority patent/WO2022166771A1/en
Publication of CN114908089A publication Critical patent/CN114908089A/en
Application granted granted Critical
Publication of CN114908089B publication Critical patent/CN114908089B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • 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
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • 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
    • 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
    • C12N15/65Introduction 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
    • C12N15/67General 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0681Cells of the genital tract; Non-germinal cells from gonads
    • C12N5/0682Cells of the female genital tract, e.g. endometrium; Non-germinal cells from ovaries, e.g. ovarian follicle cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • 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
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/106Plasmid DNA for vertebrates
    • C12N2800/107Plasmid DNA for vertebrates for mammalian

Abstract

The invention relates to 3' UTR polynucleotides capable of improving gene expression, methods of construction and uses thereof.

Description

Construction method and application of 3' UTR
Technical Field
The invention belongs to the technical field of protein expression, and particularly relates to a 3' UTR (untranslated regions) region sequence for improving mRNA (messenger ribonucleic acid) translation as well as a construction method and application thereof.
Background
Messenger ribonucleic acid (mRNA) contains, in addition to coding sequences (CDS) necessary for translation of proteins, necessary untranslated sequences such as the m7G cap, the 5'untranslated region (5' -UTR), the 3'untranslated region (3' UTR), and the poly (A) tail.
The 3 'end untranslated region is closely connected with the mRNA coding region, is closely related to post-transcriptional regulation, and influences the translation efficiency, stability and the like of the mRNA in cytoplasm through the combination of other regulation factors, for example, the combination of a small molecule ribonucleic acid (miRNA) and the 3' end untranslated region inhibits the translation of the mRNA and even induces the degradation of the mRNA; silencer sequences in the 3' untranslated region bind to RNA binding proteins and may also inhibit translation. In addition, the longer the 3' untranslated region sequence, the greater the likelihood of negatively affecting protein translation. The prior art 3' -UTRs are usually isolated from the animal itself without modification and their efficacy may be subject to host influences and thus uncertain. There remains a need in the art for a universal 3' UTR region that can enhance translation of mRNA.
Disclosure of Invention
The invention constructs 3 'end non-translated region sequences through natural gene modification or artificial design, and the 3' end non-translated region sequences ensure that the messenger ribonucleic acid of the gene keeps higher translation activity in different cells.
In a first aspect, the present invention provides a polynucleotide comprising
(1) A 3'UTR sequence which is a sequence after knockout of one or more microRNA recognition sites in the 3' UTR region of the PCBP4 gene or the PIWIL4 gene, and/or
(2) The complementary sequence of (1).
In one or more embodiments, the nucleotide sequence of the 3' UTR region of the PCBP4 gene is set forth in any one of SEQ ID No. 1 or a sequence having at least 80% sequence identity thereto.
In one or more embodiments, the nucleotide sequence of the 3' UTR region of the PIWIL4 gene is set forth in any one of SEQ ID No. 2 or a sequence having at least 80% sequence identity thereto.
In one or more embodiments, the microRNA recognition site can be obtained using http:// www.targetscan.org/vert _ 72/website search.
In one or more embodiments, the microRNA recognition site is selected from one or more microRNA recognition sites in table 1 and/or table 2.
In one or more embodiments, the one or more microRNA recognition sites in the 3' UTR region of the PCBP4 gene are selected from table 1.
In one or more embodiments, the one or more microRNA recognition sites in the 3' UTR region of the PIWIL4 gene are selected from table 2.
In one or more embodiments, the PCBP4 gene or PIWIL4 gene is a human PCBP4 gene or a human PIWIL4 gene.
In one or more embodiments, the sequence of the polynucleotide is selected from (1) the sequence set forth in any one of SEQ ID NOS: 3-7, (2) a sequence having at least 80% sequence identity to (1).
In one or more embodiments, the polynucleotide is DNA or RNA.
The first aspect of the invention also provides a method for constructing a 3'UTR sequence, which comprises the step of knocking out one or more microRNA recognition sites in the 3' UTR region of a gene.
In one or more embodiments, the gene is PCBP4 gene or PIWIL4 gene. Preferably, the PCBP4 gene or PIWIL4 gene is a human PCBP4 gene or a human PIWIL4 gene.
In one or more embodiments, the nucleotide sequence of the 3' UTR region of the PCBP4 gene is set forth in any one of SEQ ID No. 1 or a sequence having at least 80% sequence identity thereto.
In one or more embodiments, the nucleotide sequence of the 3' UTR region of the PIWIL4 gene is set forth in any one of SEQ ID No. 2 or a sequence having at least 80% sequence identity thereto.
In one or more embodiments, the microRNA recognition sites are obtained using http:// www.targetscan.org/vert _ 72/website search.
In one or more embodiments, the microRNA recognition site is selected from one or more microRNA recognition sites in table 1 and/or table 2.
In one or more embodiments, the one or more microRNA recognition sites in the 3' UTR region of the PCBP4 gene are selected from table 1.
In one or more embodiments, the one or more microRNA recognition sites in the 3' UTR region of the PIWIL4 gene are selected from table 2.
In a second aspect, the present invention provides a polynucleotide comprising
(1) A 3' UTR sequence comprising a sequence of at least 3 tandem repeats of an RNA methylation (m6A) site and optionally a linker between any two repeats, and/or
(2) The complementary sequence of (1).
In one or more embodiments, the m6A site is the recognition sequence of the m6A methylase (writer) or the m6A RNA methylation recognition protein (reader). In one or more embodiments, the m6A methylase is selected from METTL3, METTL14, METTL16, WTAP, VIRMA, ZC3H13, RBM15, RBM158, HAKAI and KIAA 1492. In one or more embodiments, the m6A RNA methylation recognition protein is selected from the group consisting of YTHDC1-2, YTHDF1-3, IGF2BP1-3, Prrc2a, HNRNPC, HNRNPG, FMR1, SRSF2, HuR, LRPRC, eIF3, and HNRNPA2B 1.
In one or more embodiments, the m6A position is selected from: AAABH, AGABH, GAABH, GGABH, wherein B ═ T or C or G, H ═ T or a or C.
In one or more embodiments, the m6A position is selected from: GGACT, GGACC, GAACT.
In one or more embodiments, the 3' UTR sequence comprises a sequence of at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10 tandem repeats of the m6A position.
In one or more embodiments, the linker is 1-5bp in length, preferably 3 bp.
In one or more embodiments, the linker sequence is selected from the group consisting of: TCT, TGT, GGT, CT, TT, and CA.
In one or more embodiments, the sequence of the polynucleotide is selected from (1) the sequence set forth in any one of SEQ ID NOS: 8-14, (2) a sequence having at least 80% sequence identity to (1).
In one or more embodiments, the polynucleotide is DNA or RNA.
The second aspect of the invention also provides a method of constructing a 3' UTR sequence comprising the step of ligating in series at least 3 RNA methylation (m6A) sites and optionally the step of adding restriction enzyme sites at both ends.
In one or more embodiments, the linkage is direct or through a linker.
In one or more embodiments, the linker is 1-5bp in length.
In one or more embodiments, the m6A site is a recognition sequence of the m6A methylase or m6A RNA methylation recognition protein. In one or more embodiments, the m6A methylase is selected from METTL3, METTL14, METTL16, WTAP, VIRMA, ZC3H13, RBM15, RBM158, HAKAI, and KIAA 1492. In one or more embodiments, the m6A RNA methylation recognition protein is selected from the group consisting of YTHDC1-2, YTHDF1-3, IGF2BP1-3, Prrc2a, HNRNPC, HNRNPG, FMR1, SRSF2, HuR, LRPRC, eIF3, and HNRNPA2B 1.
In one or more embodiments, the m6A position is selected from: AAABH, AGABH, GAABH, GGABH, wherein B ═ T or C or G, H ═ T or a or C.
In one or more embodiments, the m6A position is selected from: GGACT, GGACC, GAACT.
In one or more embodiments, the method comprises the step of linking at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10 RNA methylation (m6A) sites in tandem.
In one or more embodiments, the RNA methylation (m6A) sites in the 3' UTR sequences are the same.
The invention also provides a nucleic acid construct comprising a polynucleotide according to any of the embodiments herein and optionally a gene to be expressed.
In one or more embodiments, the nucleic acid construct is an mRNA containing the gene to be expressed and the polynucleotide.
In one or more embodiments, the nucleic acid construct is a vector, such as a cloning vector or an expression vector.
In one or more embodiments, the polynucleotide is located 3' to the gene to be expressed.
In one or more embodiments, the nucleic acid construct further comprises one or more elements selected from the group consisting of: promoter, 5' UTR, multiple cloning site, polyA.
In one or more embodiments, the promoter is the T7 promoter.
In one or more embodiments, the 5 'UTR is the 5' UTR of HBB mRNA.
In one or more embodiments, the nucleic acid construct comprises, in order, at least: a promoter, a multiple cloning site, a polynucleotide according to any one of the embodiments herein, and a polyA.
In one or more embodiments, the exogenous gene is an eGFP gene.
The invention also provides a host cell comprising a polynucleotide and/or nucleic acid construct according to any of the embodiments herein.
In one or more embodiments, the host cell is a CHO cell or a PBMC cell.
The invention also provides a pharmaceutical composition comprising a polynucleotide, nucleic acid construct or host cell according to any of the embodiments herein, and a pharmaceutically acceptable excipient.
In one or more embodiments, the polynucleotide is located 3' to the gene to be expressed.
The present invention also provides a method for increasing the strength of gene expression, prolonging the time of gene expression, increasing the ability of mRNA to initiate translation, or maintaining the length of translational activity, comprising the step of expressing a gene carrying a polynucleotide according to any of the embodiments described herein.
In one or more embodiments, the polynucleotide is located at the 3' end of the gene.
In one or more embodiments, the gene is an eGFP gene.
The invention also provides the use of a polynucleotide, nucleic acid construct and/or host cell according to any of the embodiments herein in the preparation of an agent for increasing the strength of gene expression, prolonging the time of gene expression, increasing the ability of mRNA to initiate translation, or maintaining the length of translational activity.
In one or more embodiments, the polynucleotide is located at the 3' end of the gene.
In one or more embodiments, the gene is an eGFP gene.
The invention also provides the use of a polynucleotide, nucleic acid construct and/or host cell according to any of the embodiments herein, said polynucleotide being located 3' to a gene whose expression is conducive to the treatment of a disease, in the manufacture of a medicament for the treatment of said disease.
The invention also provides the use of a polynucleotide according to any one of the embodiments herein for controlling gene expression.
In one or more embodiments, the polynucleotide is located at the 3' end of the gene.
In one or more embodiments, the gene is an eGFP gene.
Drawings
FIG. 1 shows an exemplary vector map containing the 3' UTR described herein.
FIG. 2 shows the proportion of eGFP expression fluorescence 24h after mRNA electrotransformation by PBMC cells with different 3' UTRs.
FIG. 3 shows the mean fluorescence intensity of PBMCs after 24h electrotransformation of mRNA containing different 3' UTRs.
FIG. 4 shows eGFP positivity after 72h of mRNA electrotransformation by PBMC for different 3' UTR.
FIG. 5 shows the mean fluorescence intensity of eGFP mRNA from different 3' UTRs after 72h of electroporation of PBMCs.
FIG. 6 shows eGFP positivity 24h after transfection of mRNA by CHO cells with different 3' UTRs.
FIG. 7 shows the mean fluorescence intensity of eGFP 24h after transfection of mRNA by CHO cells.
FIG. 8 shows eGFP positivity 48h after transfection of mRNA by CHO cells under different 3' UTR conditions.
FIG. 9 shows the mean fluorescence intensity of eGFP 48h after transfection of mRNA by CHO cells with different 3' UTRs.
FIG. 10 shows eGFP positivity after 96h transfection of CHO cells with different 3' UTRs.
FIG. 11 shows the mean eGFP fluorescence intensity 96h after transfection of mRNA by CHO cells with different 3' UTRs.
Detailed Description
It is to be understood that within the scope of the present invention, the above-described technical features of the present invention and the technical features specifically described below (e.g., embodiments) may be combined with each other to constitute a preferred embodiment.
As used herein and in the appended claims, the singular forms "a", "an", and "the" include plural referents unless the context clearly dictates otherwise. Likewise, the terms "a" (or "an"), "one or more" and "at least one" are used interchangeably herein. The terms "include" and variations thereof, where such terms appear in the specification and claims, are not to be limiting. Thus, the terms "comprising," "including," and "containing" are used interchangeably.
The invention constructs 3 'end non-translated region sequence through natural gene modification or artificial design, and the 3' end non-translated region sequence enables the messenger ribonucleic acid of the gene to maintain higher translation activity in different cells.
Thus, in one or more embodiments, the invention encompasses a 3'UTR polynucleotide obtained by natural genetic engineering comprising a 3' UTR sequence, or a complement thereof, said 3'UTR sequence being a sequence following knockout of one or more microRNA recognition sites in the 3' UTR region of a natural gene (e.g., human PCBP4 gene or PIWIL4 gene). The 3'UTR sequence of PCBP4 mRNA (NM-001174100) is shown in SEQ ID NO. 1, and the 3' UTR sequence of PIWIL4 mRNA (NM-152431) is shown in SEQ ID NO. 2. Herein, a microRNA recognition site is a site that is recognized by microRNA and affects mRNA stability. The sequences of some microRNA recognition sites can be obtained by searching with http:// www.targetscan.org/vert _ 72/website. In particular embodiments, the microRNA target sequence of the 3' UTR of PCBP4 is selected from one or more of table 1; the microRNA target sequence of the 3' UTR of PIWIL4 is selected from one or more of Table 2.
TABLE 1 microRNA target sequences in the 3' UTR of the PCBP4 gene
microRNA Seed sequence (Seed sequence)
Has-miR-6801-3p gcagggg
Has-miR-4313 agggggc
Has-miR-6745 tccaccc
Has-miR-4251 ttctcag
Has-miR-6777-3p agagtgg
Has-miR-1976 caggaga
Has-miR-92a-2-5p ccccacca
Has-miR-1205 cctgcaga
Has-miR-145-5p actggaa
hsa-miR-548g-3p acagttt
hsa-miR-548az-3p cagtttt
hsa-miR-3613-3p tttttga
TABLE 2 microRNA target sequences in the 3' UTR of the PIWIL4 gene
Figure BDA0002939172050000071
Figure BDA0002939172050000081
The method for constructing the 3' UTR sequence comprises the step of knocking out one or more microRNA recognition sites in the 3' UTR region of the gene and the optional step of adding restriction enzyme cutting sites at two ends of the 3' UTR region. Knocking out the microRNA recognition site can be achieved by any method known in the art, for example, mutating a 2-8nt target sequence complementary to a seed sequence (seed) of the microRNA in the 3' UTR. In a specific embodiment, primers are designed based on the 3' UTR of a gene (e.g., human PCBP4 gene or PIWIL4 gene), the corresponding sequence is amplified from the RNA group by RT-PCR, and the mutation is introduced by PCR for knock-out. Alternatively, the 3' UTR sequences of the invention, engineered by natural genes, can also be obtained directly synthetically. Exemplary sequences of the polynucleotides are set forth in any one of SEQ ID NOS 3-7.
In other embodiments, the invention comprises a 3'UTR polynucleotide obtained by artificial design, comprising a 3' UTR sequence, or a complement thereof, comprising a sequence of at least 3 tandem repeats of an RNA methylation (m6A) site, and optionally a linker between any two of the repeats. Herein, the m6A site is a recognition site of m6A methylase or m6A RAN methylation recognition protein. Illustratively, the m6A methylase may be METTL3, METTL14, METTL16, WTAP, VIRMA, ZC3H13, RBM15, RBM158, HAKAI and KIAA 1492; the m6A RNA methylation recognition protein can be YTHDC1-2, YTHDF1-3, IGF2BP1-3, Prrc2a, HNRNPC, HNRNPG, FMR1, SRSF2, HuR, LRPRC, eIF3 and HNRNPA2B 1. The m6A position useful in the present invention is selected from: AAABH, AGABH, GAABH, GGABH, wherein B ═ T or C or G, H ═ T or a or C. The 3' UTR sequence may contain a sequence of at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10 tandem repeats of the m6A position. Multiple RNA methylation (m6A) sites in a 3' UTR sequence can be the same or different. The method of constructing the 3' UTR sequence comprises the step of ligating at least 3 RNA methylation (m6A) sites in series and optionally the step of adding restriction enzyme sites at both ends. The linkage is either direct or via a linker (e.g., an oligonucleotide of 1-5bp in length). Alternatively, the artificially designed 3' UTR sequence of the present invention can also be directly synthesized. Exemplary sequences of the polynucleotides are shown in any one of SEQ ID NO 8-14.
Herein, the polynucleotide may be DNA or RNA. The DNA may be single-stranded or double-stranded. Thus, the polynucleotides herein are generally in the form of isolated polynucleotides. The term "variant" as used herein in reference to a nucleic acid may be a naturally occurring allelic variant or a non-naturally occurring variant. These nucleotide variants include degenerate variants, substituted variants, deletion variants, and insertion variants. A nucleic acid of the invention can comprise a nucleotide sequence having at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, at least about 99%, or 100% sequence identity to the nucleic acid sequence. The invention also relates to nucleic acid fragments hybridizing with the sequences described above. The polynucleotides described herein can generally be obtained by PCR amplification. Alternatively, the nucleic acid molecules described herein can also be synthesized directly.
The term "identity" or "percent identity" in two or more polypeptide or nucleic acid molecule sequences refers to two or more sequences or subsequences that are the same or wherein a percentage of amino acid residues or nucleotides are the same (e.g., at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical) over a window of comparison or designated region, when compared and aligned for maximum correspondence by manual alignment and visual inspection using methods known in the art, such as sequence comparison algorithms. For example, preferred algorithms suitable for determining percent sequence identity and percent sequence similarity are the BLAST and BLAST 2.0 algorithms, see Altschul et al (1977) Nucleic Acids Res.25:3389 and Altschul et al (1990) J.Mol.biol.215:403, respectively.
It is well known to those skilled in the art that in gene cloning procedures, it is often necessary to design appropriate cleavage sites, which may introduce one or more cleavage sites at the end of a polynucleotide, which do not affect the activity of the 3' UTR polynucleotide or the protein of interest. The scope of the present invention encompasses the sequences described herein and sequences with one or more additional enzyme cleavage sites at either end.
The 3' UTR polynucleotides of the invention can improve expression of cognate genes. Such improvement in expression is achieved by introducing into the cell a nucleic acid construct carrying a gene comprising a polynucleotide as described herein. Accordingly, the invention also provides nucleic acid constructs comprising said polynucleotides. The nucleic acid construct may be an expression cassette comprising a 3'UTR polynucleotide as described herein, a multiple cloning site, and one or more regulatory sequences, such as an origin of replication, multiple cloning site, promoter, marker gene, or translational control element, including enhancers, operators, termination sequences such as polyA, ribosome binding sites, 5' UTR, operably linked thereto. The coding sequence of the gene to be expressed may be inserted into the nucleic acid construct 5 'of the 3' UTR polynucleotide. The polynucleotides of the invention can be manipulated in a variety of ways to ensure expression of the gene. The nucleic acid construct may be manipulated prior to insertion into the vector, depending on the type of vector or requirements. Techniques for altering polynucleotide sequences using recombinant DNA methods are known in the art.
The nucleic acid construct can also be an mRNA containing a gene to be expressed and a 3' UTR polynucleotide described herein. The mRNA can be obtained by in vivo or in vitro transcription methods known in the art. In vitro transcribed mRNA can be obtained, for example, according to the HiScribe T7 ARCA mRNA kit instructions.
In a recombinant expression vector, "operably linked" means that the nucleotide sequence of interest is linked to the regulatory sequence(s) in a manner that allows for expression of the nucleotide sequence. Those skilled in the art are familiar with methods that can be used to construct expression vectors containing the 3' UTR polynucleotides described herein and optionally the coding sequence of the gene of interest and appropriate transcription/translation control signals. These methods include in vitro recombinant DNA techniques, DNA synthesis techniques, in vivo recombinant techniques, and the like.
The control sequence may be an appropriate promoter sequence. The promoter sequence is typically operably linked to the coding sequence of the protein to be expressed. The promoter may be any nucleotide sequence which shows transcriptional activity in the host cell of choice including mutant, truncated, and hybrid promoters, and may be obtained from genes encoding extracellular or intracellular polypeptides either homologous or heterologous to the host cell. An example of a suitable promoter is the T7 promoter sequence. The promoter sequence is a strong constitutive promoter sequence capable of driving high level expression of any polynucleotide sequence operably linked thereto. Other constitutive promoter sequences can also be used, including but not limited to the T3 promoter, SP6 promoter.
The control sequence may also be a suitable transcription termination sequence, a sequence recognized by a host cell to terminate transcription. The termination sequence is operably linked to the 3 'terminus of the 3' UTR polynucleotide described herein. Any termination sequence that is functional in the host cell of choice may be used in the present invention.
The control sequence may also be a suitable leader sequence, a 5' untranslated region of an mRNA which is important for translation by the host cell. The leader sequence is operably linked to the 5' end of the gene to be expressed. Any 5' untranslated region that is functional in the host cell of choice may be used in the present invention.
In certain embodiments, the nucleic acid construct is a vector. The vector may be a cloning vector, an expression vector, or a homologous recombinant vector. The polynucleotides of the present invention can be cloned into many types of vectors, for example, plasmids, phagemids, phage derivatives, animal viruses, and cosmids. Cloning vectors may be used to provide the coding sequences for the foreign genes and polynucleotides described herein. The expression vector may be provided to the cell in the form of a viral vector. Gene expression is typically achieved by operably linking a polynucleotide of the invention to a promoter, and incorporating the construct into an expression vector. The vector may be suitable for replication and integration of eukaryotic cells. Typical cloning vectors contain transcriptional and translational terminators, initiation sequences, and promoters that may be used to regulate the expression of the desired nucleic acid sequence. Viral vector technology is well known in the art and is described, for example, in Sambrook et al (2001, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York) and other virology and Molecular biology manuals. Viruses that can be used as vectors include, but are not limited to, retroviruses, adenoviruses, adeno-associated viruses, herpes viruses, and lentiviruses. Homologous recombinant vectors are used to integrate the expression cassettes described herein into the host genome.
Generally, suitable vectors comprise an origin of replication functional in at least one organism, a promoter sequence, a convenient restriction enzyme site and one or more selectable markers. For example, in certain embodiments, the invention uses a pT7-m5U vector comprising an origin of replication, a promoter, a polynucleotide as described herein, and optionally a selectable marker.
To assess the expression of a therapeutic protein, polypeptide, or portion thereof, the expression vector introduced into the cells may also contain either or both of a selectable marker gene or a reporter gene to facilitate identification and selection of expressing cells from the population of cells sought to be transfected or infected by the viral vector. In other aspects, the selectable marker may be carried on a separate piece of DNA and used in a co-transfection procedure. Both the selectable marker and the reporter gene may be flanked by appropriate regulatory sequences to enable expression in a host cell. Useful selectable markers include Flag, HA or V5. The reporter gene is used to identify potentially transfected cells and to evaluate the functionality of the regulatory sequences. After the DNA has been introduced into the recipient cell, the expression of the reporter gene is determined at an appropriate time. Suitable reporter genes may include genes encoding luciferase, β -galactosidase, chloramphenicol acetyltransferase, secreted alkaline phosphatase, or green fluorescent protein. Suitable expression systems are well known and can be prepared using known techniques or obtained commercially.
Methods for introducing and expressing genes into cells are known in the art. The vector may be readily introduced into a host cell by any method known in the art, e.g., mammalian, bacterial, yeast or insect cells. For example, the expression vector may be transferred into a host cell by physical, chemical or biological means.
Physical methods for introducing polynucleotides into host cells include calcium phosphate precipitation, lipofection, particle bombardment, microinjection, electroporation, and the like, for example, by directly introducing in vitro transcribed mRNA into cells by electroporation. Chemical means of introducing polynucleotides into host cells include colloidal dispersion systems such as macromolecular complexes, nanocapsules, microspheres, beads; and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes.
Biological methods for introducing polynucleotides into host cells include the use of viral vectors, particularly retroviral vectors, such as vectors derived from lentiviruses, poxviruses, herpes simplex virus I, adenoviruses and adeno-associated viruses. The selected gene can be inserted into a vector and packaged into a retroviral particle using techniques known in the art. The recombinant virus can then be isolated and delivered to the subject cells in vivo or ex vivo. Many retroviral systems are known in the art. Lentiviruses are a genus of the family retroviridae. Reagents for lentiviral packaging are well known in the art, as are conventional lentiviral vector systems including pRsv-REV, pMDlg-pRRE, pMD2G and the desired interference plasmid.
Herein, a host cell contains a polynucleotide or nucleic acid construct described herein. Host cells include both cells expressing the gene of interest, e.g., CHO or PBMC cells, and various cells used in the production of the cells for expression, e.g., E.coli cells, for use, e.g., in providing a polynucleotide of the invention or providing a vector as described herein. Cells suitable for use in the present invention can be of various types of origin, including prokaryotic and eukaryotic cells, such as bacterial cells, yeast cells, insect cells, and mammalian cells, including, but not limited to, sf9, BHK21, COS1, COS3, COS7, 293T, Vero. The host cell is preferably a variety of cells which facilitate expression of the gene product or fermentative production of such cells, which are well known and commonly used in the art.
Also included herein are cell preparations comprising cells described herein or extracts thereof. For example, the cell preparation can be a cell culture, pharmaceutical composition, kit, device, medium or system, e.g., a chip, etc., comprising a cell or extract thereof as described herein and a suitable culture medium. Suitable culture media for the cultivation of various types of cells are known in the basic field.
The function of the nucleic acid constructs or cells described herein depends on the gene to be expressed carried therein, e.g., if the gene to be expressed is a fluorescent protein, the nucleic acid constructs or cells described herein can be used for cell tracking. If the gene to be expressed is an agent that is beneficial for the treatment of a disease, the nucleic acid constructs or cells described herein can be used to treat the disease.
The invention provides pharmaceutical compositions comprising a nucleic acid construct or cell of the invention that produces a therapeutically effective amount of an active molecule (e.g., an expression product of a gene of interest in the nucleic acid construct) and a pharmaceutically acceptable diluent, carrier, solubilizer, emulsifier, preservative, and/or adjuvant. In certain embodiments, acceptable diluents, carriers, solubilizing agents, emulsifiers, preservatives, adjuvants and/or the like in the pharmaceutical composition are preferably non-toxic to the recipient at the dosages and concentrations employed. In certain embodiments, the pharmaceutical compositions may contain such agents for improving, maintaining or retaining, for example, the pH, osmolarity, viscosity, clarity, color, isotonicity, odor, sterility, stability, dissolution or release rate, absorption or permeation of the composition. These substances are known in the art and can be found, for example, in REMINGTON' S PHARMACEUTICAL SCIENCES, 18 th edition, eds. A.R.Genrmo, 1990, Mack Publishing Company. The optimal pharmaceutical composition can be determined depending on the intended route of administration, mode of delivery and the desired dosage.
The route of administration of the pharmaceutical composition is according to known methods, e.g., oral, by intravenous, intraperitoneal, intracerebral (intraparenchymal), intracerebroventricular, intramuscular, intraocular, intraarterial, portal vein or intralesional injection; by a sustained release system or by an implanted device. The pharmaceutical compositions of the present invention may be selected for parenteral delivery. Alternatively, the composition may be selected for inhalation or delivery through the digestive tract (such as orally). The preparation of such pharmaceutically acceptable compositions is within the skill of the art. Techniques for formulating a variety of other sustained or controlled delivery means, such as liposome carriers, bioerodible microparticles or porous beads, and depot injections, are also known to those skilled in the art.
Pharmaceutical compositions for in vivo administration are typically provided in the form of sterile preparations. Sterilization is achieved by filtration through sterile filtration membranes. In the case of lyophilization of the composition, sterilization can be performed using this method before or after lyophilization and reconstitution. Compositions for parenteral administration may be stored in lyophilized form or in solution. Parenteral compositions are typically placed in a container having a sterile access port, such as an intravenous solution strip or vial having a stopper pierceable by a hypodermic injection needle.
Once formulated, the pharmaceutical compositions are stored in sterile vials as solutions, suspensions, gels, emulsions, solids, crystals, or as dehydrated or lyophilized powders. The formulation may be stored in a ready-to-use form or reconstituted prior to administration (e.g., lyophilized). The invention also provides kits for producing a single dose administration unit. The kits of the invention may each contain a first container having a dried protein and a second container having an aqueous formulation. In certain embodiments of the invention, kits are provided that contain single and multi-chamber pre-filled syringes (e.g., liquid syringes and lyophilized syringes).
The invention also provides a method of treating a patient by administering a nucleic acid construct or cell according to any of the embodiments of the invention or a pharmaceutical composition thereof. Herein, the terms "patient," "subject," "individual," "subject" are used interchangeably herein and include any organism, preferably an animal, more preferably a mammal (e.g., rat, mouse, dog, cat, rabbit, etc.), and most preferably a human. "treating" refers to the subject taking a treatment regimen as described herein to achieve at least one positive therapeutic effect. The treatment regimen effective to treat a patient may vary depending on factors such as the disease state, age, weight of the patient, and the ability of the therapy to elicit an anti-cancer response in the subject.
The therapeutically effective amount of a pharmaceutical composition comprising a nucleic acid construct or cell of the invention to be employed will depend, for example, on the degree of treatment and the goal. One skilled in the art will appreciate that the appropriate dosage level for treatment will vary depending, in part, on the molecule delivered (e.g., the product of expression of the gene of interest in the nucleic acid construct), the indication, the route of administration, and the size (body weight, body surface or organ size) and/or condition (age and general health) of the patient. In certain embodiments, the clinician may titrate the dosage and alter the route of administration to achieve the optimal therapeutic effect.
The frequency of administration will depend on the pharmacokinetic parameters of the particular product of interest (e.g., the product of expression of the gene of interest in the nucleic acid construct) in the formulation used. The clinician typically administers the composition until a dosage is reached that achieves the desired effect. The composition may thus be administered as a single dose, or as two or more doses (which may or may not contain the same amount of the desired molecule) over time, or by means of an implanted device or catheter as a continuous infusion.
As demonstrated in the examples below, the 3' UTR polynucleotides described herein can increase the intensity of gene expression, extend the time of gene expression, increase the ability of mRNA to initiate translation, or maintain the length of translational activity. Thus, the invention also provides a method of increasing the strength of gene expression, prolonging the time of gene expression, increasing the ability of mRNA to initiate translation, or maintaining the length of translational activity, comprising the step of expressing a gene carrying a 3' UTR polynucleotide according to any of the embodiments described herein. Typically, the 3'UTR polynucleotide is located at the 3' end of the gene. Illustratively, expression is achieved by introducing the nucleic acid construct into a cell as described above and incubating the cell under conditions for expression of the gene. GFP is used in the examples only as an example of the gene to be expressed.
The invention also provides the use of a 3' UTR polynucleotide, nucleic acid construct and/or host cell according to any of the embodiments herein, wherein the 3' UTR polynucleotide is located at the 3' end of a gene, the expression of which contributes to the treatment of a disease, in the manufacture of a medicament for use in the treatment of said disease. A nucleic acid construct comprising the gene and the 3' UTR polynucleotide can be introduced into a cell by a method as described above, and the gene expressed by incubating the cell under conditions for expression of the gene. The expression of the gene may be useful in treating the disease by directly producing a therapeutic effect from the gene or its product, or by causing the expression or activation of other genes or proteins that produce a therapeutic effect.
Examples
Example 1, 3' end untranslated region transcription template vector construction
1) Construction of transcription template vector of 3' end untranslated region expression eGFP (eGFP) reporter gene based on human natural gene modification
Construction of vector pT7-m5U-eGFP-1#3U
Kingchi (Kingchi) was entrusted with the synthesis of a modified 3' untranslated region (SEQ ID NO:3) sequence based on human PCBP4 gene, and the vector pUC57-1#3U was digested with Sal I and Hind III to recover a 430bp fragment 1# 3U. An EGFP transcription template vector pT7-m5U-eGFP containing a T7 promoter, a 5 'UTR sequence (m5U) of HBBmRNA (NM-000518), an EGFP sequence, a HBB mRNA 3' UTR sequence and a polyA sequence (SEQ ID NO:16) was constructed. After Sal I and Hind III double enzyme digestion, the recovered fragment 1#3U is connected to pT7-m5U-eGFP vector to replace 3' UTR of original HBB mRNA, and pT7-m5U-eGFP-1#3U is obtained.
FIG. 1 exemplarily shows a map of pT7-m5U-eGFP-1#3U, from which the maps of the subsequent vectors differ by using a different 3' UTR.
Construction of vector pT7-m5U-eGFP-2#3U
The Jinwei Zhi company was entrusted with the synthesis of a sequence (SEQ ID NO:4) modified from the 3' -untranslated region of the human PCBP4 gene, and after carrying out double digestion with Sal I and Hind III double digestion vectors pUC57-2#3U, the fragment 2#3U was recovered in a size of 143bp, and pT7-m5U-eGFP vectors were subjected to double digestion with Sal I and Hind III, and the recovered fragment 2#3U was ligated to pT7-m5U-eGFP vectors, to obtain pT7-m5U-eGFP-2# 3U.
Construction of vector pT7-m5U-eGFP-3#3U
The Jinwei Zhi company was entrusted with the synthesis of a modified 3' untranslated region (SEQ ID NO:5) sequence based on human PCBP4 gene, and after Sal I and HindIII double digestion of vector pUC57-3#3U, a 443bp fragment 3#3U was recovered, and pT7-m5U-eGFP vector was subjected to Sal I and HindIII double digestion, the recovered fragment 3#3U was ligated to pT7-m5U-eGFP vector, to obtain pT7-m5U-eGFP-3# 3U.
Construction of vector pT7-m5U-eGFP-4#3U
The Jinwei Zhi company is entrusted with synthesizing a sequence (SEQ ID NO:6) based on the 3' end non-translation region modification of the human PCBP4 gene, a Sal I and Hind III double enzyme digestion vector pUC57-4#3U is used for recovering a fragment 4#3U with the size of 430bp, a pT7-m5U-eGFP vector is subjected to Sal I and Hind III double enzyme digestion, and the recovered fragment 4#3U is connected to a pT7-m5U-eGFP vector to obtain pT7-m5U-eGFP-4# 3U.
Construction of vector pT7-m5U-eGFP-5#3U
The Kingjinweizhi company was assigned to synthesize a sequence (SEQ ID NO:7) modified from the 3' -untranslated region of the adult PIWIL4 gene using a Sal I and Hind III double digestion vector pUC57-5#3U to recover a 124bp fragment 5#3U, and a pT7-m5U-eGFP vector was subjected to Sal I and Hind III double digestion, and the recovered fragment 5#3U was ligated to a pT7-m5U-eGFP vector to obtain pT7-m5U-eGFP-5# 3U.
2) Construction of eGFP reporter gene transcription template vector with 3' end untranslated region as artificial random sequence
Construction of vector pT7-m5U-eGFP-6#3U
The artificial random sequence (SEQ ID NO:8) was synthesized by Cinciderogas, and the 100bp fragment 6#3U was recovered from the plasmid pUC57-6#3U digested with Sal I and Hind III, and the recovered fragment 6#3U was ligated to the pT7-m5U-eGFP vector after the plasmid pT7-m5U-eGFP vector digested with Sal I and Hind III, to obtain pT7-m5U-eGFP-6# 3U.
Construction of vector pT7-m5U-eGFP-7#3U
The artificial random sequence (SEQ ID NO:9) was synthesized by Cinciderogas, and after the plasmid pUC57-7#3U was digested with Sal I and Hind III to recover a 116bp fragment 7#3U, the recovered fragment 7#3U was ligated to the pT7-m5U-eGFP vector after the plasmid pT7-m5U-eGFP vector was digested with Sal I and Hind III to obtain pT7-m5U-eGFP-7# 3U.
Construction of vector pT7-m5U-eGFP-8#3U
An artificial random sequence (SEQ ID NO:10) was synthesized by Jinwei Zhi Co., Ltd.A vector pUC57-8#3U was digested with Sal I and Hind III, a 86 bp-sized fragment 8#3U was recovered, and a pT7-m5U-eGFP vector was digested with Sal I and Hind III, and the recovered fragment 8#3U was ligated to a pT7-m5U-eGFP vector to obtain pT7-m5U-eGFP-8# 3U.
Construction of vector pT7-m5U-eGFP-9#3U
An artificial random sequence (SEQ ID NO:11) was synthesized by Jinwei Zhi Co., Ltd., and a vector pUC57-9#3U was digested with Sal I and Hind III, a 92 bp-sized fragment 9#3U was recovered, and a pT7-m5U-eGFP vector was digested with Sal I and Hind III, and the recovered fragment 9#3U was ligated to a pT7-m5U-eGFP vector to obtain pT7-m5U-eGFP-9# 3U.
Construction of vector pT7-m5U-eGFP-10#3U
The artificial random sequence (SEQ ID NO:12) was synthesized by Cinciderogas, and after double digestion of the vector pUC57-10#3U with Sal I and Hind III, the 103bp fragment 10#3U was recovered, and double digestion of the pT7-m5U-eGFP vector with Sal I and Hind III, the recovered fragment 10#3U was ligated to the pT7-m5U-eGFP vector, to obtain pT7-m5U-eGFP-10# 3U.
Construction of vector pT7-m5U-eGFP-11#3U
An artificial random sequence (SEQ ID NO:13) was synthesized by Jinwei Zhi Co., Ltd., and after digesting the vector pUC57-11#3U with Sal I and Hind III, the 97bp fragment 11#3U was recovered and the pT7-m5U-eGFP vector was digested with Sal I and Hind III, the recovered fragment 11#3U was ligated to the pT7-m5U-eGFP vector to obtain pT7-m5U-eGFP-11# 3U.
Construction of vector pT7-m5U-eGFP-12#3U
An artificial random sequence (SEQ ID NO:14) was synthesized by Jinwei Zhi Co., Ltd.A vector pUC57-12#3U was digested with Sal I and Hind III, a fragment 12#3U having a size of 97bp was recovered, and a pT7-m5U-eGFP vector was digested with Sal I and Hind III, and the recovered fragment 12#3U was ligated to a pT7-m5U-eGFP vector to obtain pT7-m5U-eGFP-12# 3U.
3) Construction of eGFP reporter gene transcription template vector with 3' end untranslated region as natural gene sequence
Construction of vector pT7-m5U-eGFP-PCBP4-3U
The mRNA 3' UTR sequence (SEQ ID NO:1) of PCBP4 was synthesized by Cincidere Authoris, and the vector pUC57-PCBP4#3U was digested with Sal I and HindIII, and the PCBP4#3U was recovered as a 609bp fragment, and the vector pT7-m5U-eGFP was digested with Sal I and HindIII, and the recovered fragment P4#3U was ligated to the vector pT7-m5U-eGFP to obtain pT7-m5U-eGFP-PCBP4# 3U.
Construction of vector pT7-m5U-eGFP-PIWIL4-3U
The sequence of PIWIL4 mRNA 3' UTR (SEQ ID NO:2) was synthesized by KINYZ corporation, and after Sal I and HindIII double digestion vector pUC57-PIWIL4#3U was used to recover the segment PIWIL4#3U with the size of 429bp, and pT7-m5U-eGFP vector was double digested with Sal I and HindIII, the recovered segment PIWIL4#3U was ligated to pT7-m5U-eGFP vector to obtain pT7-m5U-eGFP-PIWIL4# 3U.
Construction of vector pT7-m5U-eGFP-IL2#3U
The method comprises the steps of entrusting Jinzhi company to synthesize an IL-23' UTR sequence (SEQ ID NO:15), carrying out double enzyme digestion on a vector pUC57-IL2#3U by using Sal I and Hind III, recovering a fragment IL2#3U with the size of 285bp, carrying out double enzyme digestion on a pT7-m5U-eGFP vector by using Sal I and Hind III, connecting the recovered fragment IL2#3U to a pT7-m5U-eGFP vector, and obtaining pT7-m5U-eGFP-IL2# 3U.
Example 2 in vitro transcription of mRNA containing an artificially engineered or designed 3' UTR
The HBB mRNA 3'UTR was used as a control 3' UTR sequence, and the other 5'untranslated region (5' UTR), eGFP coding region (CDS) and poly A tail (poly (A)) template sequences were all identical. Separately digesting pT7-m5 with BsaI restriction enzymeU-eGFP, pT7-m5 7-eGFP-1 #3 7-m5 7-eGFP-2 #3 7-m5 7-eGFP-3 #3 7-m5 7-eGFP-4 #3 7-m5 7-eGFP-5 #3 7-m5 7-eGFP-6 #3 7-m5 7-eGFP-7 #3 7-m5 7-eGFP-8 #3 7-m5 7-eGFP-9 #3 7-m 5-eGFP-10 # 3# 7-m5 7-eGFP-11 #3 7-m 5-eGFP-12 #3 7-m5 7-eGFP-7 # 7-eGFP-p 7-m 5-7-eGFP-PIL # 7-p # 7-m 5-eGFP-3-7-eGFP-IL # 3-m 3-7-eGFP-U3 # 7, in vitro transcribed mRNA was obtained according to the guidelines of the hisprobe T7 ARCA mRNA kit (NEB, E2065S): adding the following reagents into a test tube without nuclease: 2 × ARCA/NTP Mix 10. mu.L, BsaI linearized plasmid template 1. mu.g, T7 RNA Polymerase Mix 2. mu.l, H 2 O to 20 μ L; incubation at 37 ℃ for 30 min; adding 2 μ L DNase, and digesting for 15 min; using LiCl solution provided with the kit for precipitation for 2 h; centrifuging at 12000rpm for 2 min; cleaning with 75% ethanol for 2 times, sucking off all supernatant, and air drying at room temperature; h 2 Dissolving and precipitating mRNA to finally obtain mRNA-eGFP # ctl, mRNA-eGFP1#, mRNA-eGFP2#, mRNA-eGFP3#, mRNA-eGFP4#, mRNA-eGFP5#, mRNA-eGFP6#, mRNA-eGFP7#, mRNA-eGFP8#, mRNA-eGFP9#, mRNA-eGFP10#, mRNA-eGFP11#, mRNA-eGFP12#, mRNA-eGFP PCBP4#, mRNA-eGFP PIWIL4#, and mRNA-eGFP IL2 #.
Example 3 maintenance of Gene expression assay of constructed 3' UTR in T cells
1) Determination of the effect of 3' untranslated region sequences on mRNA translation by eGFP 24h expression intensity:
the 3'UTR sequence of HBB mRNA was used as a control 3' UTR sequence, and the other 5'untranslated region (5' UTR), coding sequence (CDS) and poly (A) tail sequences were all identical. Resuscitated and resuspended at 5X 10 6 Freshly isolated Peripheral Blood Mononuclear Cells (PBMCs) were electrically transferred to 20. mu.g of mRNA (mRNA-eGFP # ctl, mRNA-eGFP1#, mRNA-eGFP2#, mRNA-eGFP3#, mRNA-eGFP4#, mRNA-eGFP5#, mRNA-eGFP6#, mRNA-eGFP7#, mRNA-eGFP8#, mRNA-eGFP9#, mRNA-eGFP10#, mRNA-eGFP11#, mRNA-eGFP12, mRNA-eGFP4#, mRNA-eGFP4#, mRNA-eGFPIL 2) expressing EGFP protein obtained in example 1-2 using Lonza 4D-Nucleoefector, to PBMCs (PBMCs) at 37 ℃ and 5% CO2 ℃. (37 ℃.) 2 The culture medium comprises AIM-V (Gibco) and 2% fetal bovine serum (Gibco). 24 is smallCells were counted and 3X 10 cells were taken 5 Cells were analyzed for fluorescent expression by flow cytometry.
FIG. 2 is the ratio of eGFP expression fluorescence 24h after mRNA electrotransformation by PBMC cells under different 3' UTR conditions, and it can be seen that there is no significant difference in the electrotransformation efficiency of mRNA in PBMC for each group. FIG. 3 is the average fluorescence intensity of PBMC after 24h electrotransformation of mRNA containing different 3' UTR, compared with that of mRNA containing PCBP4, PIWIL4, IL-2 and other natural 3' UTR, the eGFP expression intensity of mRNA containing artificially engineered or designed 3' UTR transcribed in vitro is found to be stronger in PBMC than that of mRNA containing PCBP4, PIWIL4, IL-2 and other natural 3' UTR, wherein mRNA-eGFP8#, mRNA-eGFP9# and mRNA-eGFP10# are significantly better than that of mRNA containing HBB 3' UTR.
2) Determination of the Effect of 3' untranslated region sequences on mRNA translation time in T cells by eGFP 72h expression intensity
The 3'UTR sequence of HBB mRNA was used as a control 3' UTR sequence, and the other 5'untranslated region (5' UTR), coding sequence (CDS) and poly (A) tail sequences were all identical. Resuscitated and resuspended 5X 10 6 Freshly isolated Peripheral Blood Mononuclear Cells (PBMCs) were used to electrically transfer 20. mu.g of mRNA (mRNA-eGFP # ctl, mRNA-eGFP1#, mRNA-eGFP2#, mRNA-eGFP3#, mRNA-eGFP4#, mRNA-eGFP5#, mRNA-eGFP6#, mRNA-eGFP7#, mRNA-eGFP8#, mRNA-eGFP9#, mRNA-eGFP10#, mRNA-eGFP11#, mRNA-eGFP12, mRNA-eGFP4#, mRNA-eGFP WIL4#, mRNA-eGFPIL 2) expressing EGFP protein obtained in example 1-2 to PBMCs, which were cultured in a 37 ℃ C, 5% CO2 incubator with medium components of M-BCO, and (M-V bco, AIbco 2) (2%) using Lonza 4D-Nucleoeofelector. After 72h, cells were counted and 3X 10 cells were taken 5 Cells were resuspended in PBS and analyzed for cell positivity and mean fluorescence intensity by flow cytometry.
FIG. 4 shows the eGFP positive rate after mRNA 72h is electrically transferred to PBMC under different 3' UTR conditions, and it can be seen that the expression time of mRNA artificially modified or designed 3' UTR in PBMC is better than that of the natural 3' UTR such as PCBP4, PIWIL4, IL-2 and the like. FIG. 5 shows the average fluorescence intensity of eGFP mRNA of different 3' UTRs after electroporation to PBMC72h, and it was found that eGFP transcribed in vitro to mRNA containing artificially modified or designed 3' UTR was more strongly expressed in PBMC than the 3' UTR of the natural sequences such as PCBP4, PIWIL4, IL-2, etc. After 72 hours, mRNA containing the artificially engineered or designed 3'UTR still had higher expression activity in PBMC, whereas mRNA containing the 3' UTR of the IL-2 native sequence had been substantially not expressed.
Example 4 maintenance of Gene expression assay of constructed 3' UTR in CHO cells
1) CHO cell transfection (Lipo2000 transfection kit):
the HBB mRNA 3'UTR was used as a control 3' UTR sequence, and the other 5'untranslated region (5' UTR), coding sequence (CDS) and poly (A) sequences were all identical.
a. Cell plating: digesting cultured CHO cells with pancreatin, counting, and collecting 3 × 10 5 The cells are resuspended in 3mL culture medium, and are spread in a 6-well plate for 24 hours, and the components of the culture medium are 45% DMEM culture medium + 45% RPMI-1640 culture medium + 10% serum + 1% L-glutamine + 1% Thymidine-Hypoxanthine (Hypoxanthine-Thymidine);
b. changing culture solution: after the CHO cells are cultured in a 6-well plate for 24 hours, discarding the original cell culture solution, adding 1mL of PBS buffer solution to wash the cells, discarding the washing solution, adding 2mL of fresh culture solution into the cell culture solution, and putting the 6-well plate back into the incubator at 37 ℃ again for culture;
c. transfection reagent (per well): mu.g (1. mu.g/. mu.L) of eGFP mRNA obtained in example 1-2 (mRNA-eGFP # ctl, mRNA-eGFP1#, mRNA-eGFP2#, mRNA-eGFP3#, mRNA-eGFP4#, mRNA-eGFP5#, mRNA-eGFP6#, mRNA-eGFP7#, mRNA-eGFP8#, mRNA-eGFP9#, mRNA-eGFP10#, mRNA-eGFP11#, mRNA-eGFP12#, mRNA-eGFP PCBP4#, mRNA-eGFP PIPIWIL 4#, mRNA-eGFPIL2#) + 6. mu.L Lipo2000 (mRNA: Lipo 2000: 1:2), and 200. mu.L of opti-MEM medium were incubated, vortexed for 5-20min at room temperature, and a complex was formed;
d. addition of transfection mix: the prepared transfection reagent was quickly dropped into a 6-well plate plated with CHO cells, the 6-well plate was shaken several times and then returned to 37 ℃ with 5% CO 2 Culturing in an incubator for 144h at the maximum.
2) Determination of the influence of 3' untranslated region sequence on translation time and intensity of mRNA in CHO cells by eGFP expression intensity
After 24h the medium was aspirated off and washed 1 time with 1 XPBSDigesting CHO cells with pancreatin, counting, and taking 3 × 10 5 Resuspending the cells into 3mL culture solution, spreading the cells into a 6-well plate for culture, wherein the components of a culture medium are 45% DMEM culture medium, 45% RPMI-1640 culture medium, 10% serum, 1% L-glutamine and 1% thymidine hypoxanthine; another 3X 10 5 Cells were resuspended in 1 × PBS, flow observed, and analyzed by Kaluza Analysis.
FIG. 6 shows eGFP positivity of CHO cells transfected with mRNA for 24h under different 3'UTR conditions, and it can be seen that there was no significant difference in the transfection efficiency of mRNA with different 3' UTR in CHO cells. FIG. 7 shows the mean fluorescence intensity of eGFP 24h after transfection of mRNA into CHO cells, and results show that eGFP expression intensity in CHO cells when mRNA containing artificially modified or designed 3'UTR is transcribed in vitro is stronger than that of 3' UTR of natural sequences such as PCBP4, PIWIL4, IL-2, etc.
After 48h the medium was aspirated, washed 1 time with 1 × PBS, the CHO cells were trypsinized, counted and 3 × 10 cells were taken 5 Resuspending the cells into 3mL culture solution, spreading the cells into a 6-well plate for culture, wherein the components of a culture medium are 45% DMEM culture medium, 45% RPMI-1640 culture medium, 10% serum, 1% L-glutamine and 1% thymidine hypoxanthine; another 3X 10 5 Cells were resuspended in 1 × PBS, flow observed, and analyzed by FlowJo X.
FIG. 8 shows eGFP positivity of CHO cells transfected with mRNA 48h after transfection with 3'UTR, and there was no difference in the expression duration of mRNA of the different 3' UTRs in CHO cells after 48 h. FIG. 9 shows the mean fluorescence intensity of eGFP 48h after CHO cell transfection of mRNA, showing that the eGFP expression intensity in CHO cells after 48h after transcription of mRNA containing artificially modified or designed 3' UTR is stronger than that of PCBP4, PIWIL4, IL-2 and other natural sequences 3' UTR, and the fluorescence intensity of mRNA of natural 3' UTR is rapidly reduced.
After 96h the medium was aspirated, washed 1 time with 1 × PBS, the CHO cells were trypsinized, counted and 3 × 10 cells were taken 5 Cells were resuspended in 1 × PBS, flow observed, and analyzed by FlowJo X.
FIG. 10 shows the eGFP positivity of CHO cells after 96h transfection with different 3' UTRs, and it can be seen that eGFP mRNA containing artificially modified or designed 3' UTRs has better expression persistence in CHO cells than mRNA containing 3' UTRs of natural sequences such as PCBP4, PIWIL4, IL-2, etc. FIG. 11 shows the mean eGFP fluorescence intensity 96h after CHO cells were transfected with mRNA, showing that eGFP expression intensity in CHO cells is stronger than that in IL-23 'UTR when mRNA containing artificially engineered or designed 3' UTR is transcribed in vitro. As can be seen, after 96 hours, the mRNA containing the artificially modified or designed 3'UTR still has higher expression activity, while the mRNA containing the IL-23' UTR has very low expression.
Sequence listing
<110> Shanghai cell therapy group Co., Ltd
Construction method and application of <120> 3' UTR
<130> 207238
<160> 16
<170> SIPOSequenceListing 1.0
<210> 1
<211> 603
<212> DNA
<213> Artificial Sequence
<400> 1
ggccagctga ggtacaggca ggggcaggca ggaccaccag cagggggctg cctctgcacc 60
ctacccgccc aaggagactc caccctgggg tcccaaacgc cgctaacgcc cagacgcatg 120
gatgcacccc ctaccctgcc tccatctatg ggagttcttt ctctcagagt gggggcagtt 180
tctggcccag gggtctgagc tgcggcagcc ccagggcagg gggccctacc tcctcagctc 240
tgtgcttgga tacagggagc agccaggaga ctccctagtg cccccaccat ggcgggtgtc 300
actcacgcac tccccatccc ttagggcttc ctggcctact gcatccttgt gggagtcagg 360
gaggagggcc cgttgggtag ctggggccag gcttctctcc ccaccacctg cagatttctt 420
gctgcttcca ctgataccct tttgactgga atgaactggc tgggcttgtc agggggcacc 480
ccaaagaggg ggcactgcca ggtagctggg ggagtggcat ggggcagggg cccagttctc 540
agcagcagac actctgtaca gttttttcaa tccctgtttt tgaataaata ttctcagcga 600
cca 603
<210> 2
<211> 429
<212> DNA
<213> Artificial Sequence
<400> 2
tggcatgaac tactggcatc actagatgga caatccaaga agaaattggt atactttgtg 60
caaatctgcc ataagctcaa ggctgtgact ggggaaaaag attgagctta gttttcatgt 120
ctaggaaaaa aagcaaaaca acttaatctg aaacagtttt aaaaaatgtg tgttattttg 180
ttttaaagag ttgtatgctt ggggtaaatt ttcattgtca tatgtggaat ttaaatatac 240
catcatctac aaagaattcc acagagttaa atatcttaag ttaaacactt aaaataagtg 300
tttgcgtgat attttgatga cagataaaca gagtctaatt cccaccccaa attttgctga 360
ggttttctta atgttgtaga gcattttgta gagtggttta aatagttgaa aataaagttc 420
agaacatca 429
<210> 3
<211> 424
<212> DNA
<213> Artificial Sequence
<400> 3
ggccagctga ggtacagcag gcaggaccac catgctcgca ccctacgcaa gagactgggg 60
tcccaaacgc cgctaacgag acgcatggat gcacccccta ccgctcatct atggagttct 120
ttcgtgtttc tggccagggg tctgagctgc ggcagcccca ggcctacctc ctcagctctg 180
tgcttggata cagggagcag cctccctagt gctggcgggt gtcactcacg cactccccat 240
cccttagggc ttcctggcct actgcatctg acagaggagg gcccgttggg tagctggggc 300
caggcttctc ttttcgctcc tgaacccttt tgtgaactgg ctgggcttgt cagctgccag 360
gtagctgggg gagtggcatg ggcccagcag cagacactct gtttcaatcc tgattacgac 420
cagg 424
<210> 4
<211> 137
<212> DNA
<213> Artificial Sequence
<400> 4
ccccaggcct acctcctcag ctctgtgctt ggatacagag cagcctccct agtgctggcg 60
ggtgtcactc acgcactccc catcccttag ggcttcctgg cctactgcat ccttgtagtc 120
agaggagggc ccgttgg 137
<210> 5
<211> 431
<212> DNA
<213> Artificial Sequence
<400> 5
tgggagcaag aaggctgagc ggcagaaatt ctcctactga ggccagctga ggtacagcag 60
gcaggaccac catgccctac caaggagact ccaccctggg gtcccaaacg ccgctaacgc 120
atggatgcac cccctactgc tatctatatc tttcgtttct aggtctgagc tgcggcagcc 180
ccaggcctac ctcctcagct ctgtgcttgg atacagggag cagcctccct agtgctggcg 240
ggtgtcactc acgcactccc catcccttag ggcttcctgg cctactgcat ctgtagtcag 300
aggagggccc gttgggtagc tggggccagg cttctctttt cttattttga actggctggg 360
cttgtcagct gccaggtagc tgggggagtg gcatgggccc agcagcagac actctgttca 420
atacgaccag g 431
<210> 6
<211> 418
<212> DNA
<213> Artificial Sequence
<400> 6
ctgcacagct tggagatggg ctggtgggcg gcctcagaac tcagctgtga ctatgaagtg 60
ggcttttgtg ttattagaat aagatttctt tcttgtcttt taaacctacg tttctgagta 120
acagctgaaa atacggtggt actgccactc tgcaggtgga gcgggtgact ctgggggacc 180
attaagacct ccagaccggg tgcggtggtt cacacctgta atccaagcac tttgggaggc 240
cgaggcgggt ggatcatgag gttcaaaccc gtctctacta aaatacaaaa aaattagccg 300
ggtgtggcgg tgcacgcctg tagtcccagc tactcaggag gctaagatcg cttgaacccg 360
ggaggtggag gttgcagtga gccgagatca cgccactgcc tgttgacaaa tctgtctc 418
<210> 7
<211> 112
<212> DNA
<213> Artificial Sequence
<400> 7
tggggtaaat tttcattgtc atatttaaat ataccatcat tatcttaagt taaacactta 60
aaataagtgt ttgcgtgatt aactaattcg ttaatgttta aatataaaca tc 112
<210> 8
<211> 88
<212> DNA
<213> Artificial Sequence
<400> 8
ttctggactt ctggacttct ggacttctgg acttctggac ttctggactt ctggacttct 60
ggacttctgg acttctggac ttctggac 88
<210> 9
<211> 104
<212> DNA
<213> Artificial Sequence
<400> 9
ttctggactt ctggacttct ggacttctgg acttctggac ttctggactt ctggacttct 60
ggacttctgg acttctggac ttctggactt ctggacttct ggac 104
<210> 10
<211> 80
<212> DNA
<213> Artificial Sequence
<400> 10
ttgtggactt gtggacttgt ggacttgtgg acttgtggac ttgtggactt gtggacttgt 60
ggacttgtgg acttgtggac 80
<210> 11
<211> 80
<212> DNA
<213> Artificial Sequence
<400> 11
tggtggactg gtggactggt ggactggtgg actggtggac tggtggactg gtggactggt 60
ggactggtgg actggtggac 80
<210> 12
<211> 91
<212> DNA
<213> Artificial Sequence
<400> 12
ctggactctg gactctggac tctggactct ggactctgga ctctggactc tggactctgg 60
actctggact ctggactctg gactctggac t 91
<210> 13
<211> 91
<212> DNA
<213> Artificial Sequence
<400> 13
ttggaccttg gaccttggac cttggacctt ggaccttgga ccttggacct tggaccttgg 60
accttggacc ttggaccttg gaccttggac c 91
<210> 14
<211> 91
<212> DNA
<213> Artificial Sequence
<400> 14
cagaactcag aactcagaac tcagaactca gaactcagaa ctcagaactc agaactcaga 60
actcagaact cagaactcag aactcagaac t 91
<210> 15
<211> 279
<212> DNA
<213> Artificial Sequence
<400> 15
taattaagtg cttcccactt aaaacatatc aggccttcta tttatttaaa tatttaaatt 60
ttatatttat tgttgaatgt atggtttgct acctattgta actattattc ttaatcttaa 120
aactataaat atggatcttt tatgattctt tttgtaagcc ctaggggctc taaaatggtt 180
tcacttattt atcccaaaat atttattatt atgttgaatg ttaaatatag tatctatgta 240
gattggttag taaaactatt taataaattt gataaatat 279
<210> 16
<211> 1055
<212> DNA
<213> Artificial Sequence
<400> 16
taatacgact cactatagga catttgcttc tgacacaact gtgttcacta gcaacctcaa 60
acagacacca tggtgagcaa gggcgaggag ctgttcaccg gggtggtgcc catcctggtc 120
gagctggacg gcgacgtaaa cggccacaag ttcagcgtgt ccggcgaggg cgagggcgat 180
gccacctacg gcaagctgac cctgaagttc atctgcacca ccggcaagct gcccgtgccc 240
tggcccaccc tcgtgaccac cctgacctac ggcgtgcagt gcttcagccg ctaccccgac 300
cacatgaagc agcacgactt cttcaagtcc gccatgcccg aaggctacgt ccaggagcgc 360
accatcttct tcaaggacga cggcaactac aagacccgcg ccgaggtgaa gttcgagggc 420
gacaccctgg tgaaccgcat cgagctgaag ggcatcgact tcaaggagga cggcaacatc 480
ctggggcaca agctggagta caactacaac agccacaacg tctatatcat ggccgacaag 540
cagaagaacg gcatcaaggt gaacttcaag atccgccaca acatcgagga cggcagcgtg 600
cagctcgccg accactacca gcagaacacc cccatcggcg acggccccgt gctgctgccc 660
gacaaccact acctgagcac ccagtccgcc ctgagcaaag accccaacga gaagcgcgat 720
cacatggtcc tgctggagtt cgtgaccgcc gccgggatca ctctcggcat ggacgagctg 780
tacaagtaag tcgacgctcg ctttcttgct gtccaatttc tattaaaggt tcctttgttc 840
cctaagtcca actactaaac tgggggatat tatgaagggc cttgagcatc tggattctgc 900
ctaataaaaa acatttattt tcattgcaag cttaaaaaaa aaaaaaaaaa aaaaaaaaaa 960
aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaggaaaaa aaaaaaaaaa aaaaaaaaaa 1020
aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaa 1055

Claims (10)

1. A polynucleotide, said polynucleotide comprising:
(1) a 3'UTR sequence which is a sequence obtained by knocking out one or more microRNA recognition sites in a 3' UTR region of a PCBP4 gene or a PIWIL4 gene,
(2) a 3' UTR sequence comprising a sequence of at least 3 tandem repeats of position m6A and optionally a linker between any two repeats, and/or
(3) The complementary sequence of (1) or (2).
2. The polynucleotide of claim 1,
the PCBP4 gene or the PIWIL4 gene is a human PCBP4 gene or a human PIWIL4 gene, and/or
The microRNA recognition site is shown in a website http:// www.targetscan.org/vert _ 72/and/or
The m6A site is a recognition sequence of m6A methylase or m6A RNA methylation recognition protein, and/or
Said 3' UTR sequence comprises a sequence of at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10 tandem repeats of said m6A position, and/or
The length of the joint is 1-5 bp;
preferably, the first and second electrodes are formed of a metal,
the nucleotide sequence of the 3'UTR region of the PCBP4 gene is shown as SEQ ID NO. 1, and/or the nucleotide sequence of the 3' UTR region of the PIWIL4 gene is shown as SEQ ID NO. 2, or
The m6A position is selected from: AAABH, AGABH, GAABH, GGABH, wherein B ═ T or C or G, H ═ T or a or C;
more preferably, the sequence of the polynucleotide is selected from (1) the sequence as set forth in any one of SEQ ID NOS: 3-14, (2) a sequence having at least 80% sequence identity to (1).
3. A method for constructing a 3'UTR sequence comprises the step of knocking out one or more microRNA recognition sites in a 3' UTR region of a gene,
preferably, the gene is the PCBP4 gene or the PIWIL4 gene,
more preferably, the PCBP4 gene or PIWIL4 gene is human PCBP4 gene or human PIWIL4 gene,
further preferably, the nucleotide sequence of the 3' UTR region of the gene is shown as SEQ ID NO 1 or 2 or any sequence with at least 80% sequence identity with the nucleotide sequence, and/or the microRNA recognition site is shown as website http:// www.targetscan.org/vert _ 72.
4. A method of constructing a 3' UTR sequence comprising the step of ligating at least 3m 6A sites in series and optionally the step of adding restriction enzyme sites at both ends,
preferably, the first and second electrodes are formed of a metal,
the connection is direct or via a joint, and/or
The m6A site is a recognition sequence of m6A methylase or m6A RNA methylation recognition protein, and/or
The method comprises the step of linking at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10 m6A sites in series;
more preferably still, the first and second liquid crystal compositions are,
the linker is 1-5bp in length, and/or
The m6A position is selected from: AAABH, AGABH, GAABH, GGABH, wherein B ═ T or C or G, H ═ T or a or C, and/or
The m6A positions in the 3' UTR sequence are identical.
5. A nucleic acid construct comprising the polynucleotide of claim 1 or 2 and optionally a gene to be expressed, said polynucleotide being located 3' of said gene to be expressed,
preferably, the nucleic acid construct further comprises one or more elements selected from the group consisting of: promoter, 5' UTR, multiple cloning site, polyA.
6. A host cell comprising the polynucleotide of claim 1 or 2 and/or the nucleic acid construct of claim 5,
preferably, the host cell is a CHO cell or a PBMC cell.
7. A pharmaceutical composition comprising the polynucleotide of claim 1 or 2, the nucleic acid construct of claim 5, or the host cell of claim 6, and a pharmaceutically acceptable excipient.
8. A method for increasing the strength of gene expression, prolonging the time of gene expression, increasing the mRNA start-up translational ability of a gene, or maintaining the length of translational activity, comprising the step of expressing a gene carrying the polynucleotide of claim 1 or 2, which is located at the 3' end of the gene.
9. Use of a polynucleotide according to claim 1 or 2, a nucleic acid construct according to claim 5 or a host cell according to claim 6, wherein the polynucleotide is located at the 3' end of a gene whose expression is useful in the treatment of a disease, in the manufacture of a medicament for the treatment of said disease.
10. Use of a polynucleotide according to claim 1 or 2 to control the expression of a gene, said polynucleotide being located at the 3' end of said gene.
CN202110172382.6A 2021-02-08 2021-02-08 Construction method and application of 3' UTR Active CN114908089B (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN202110172382.6A CN114908089B (en) 2021-02-08 2021-02-08 Construction method and application of 3' UTR
PCT/CN2022/074391 WO2022166771A1 (en) 2021-02-08 2022-01-27 3'utr construction method and application

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110172382.6A CN114908089B (en) 2021-02-08 2021-02-08 Construction method and application of 3' UTR

Publications (2)

Publication Number Publication Date
CN114908089A true CN114908089A (en) 2022-08-16
CN114908089B CN114908089B (en) 2024-03-15

Family

ID=82741976

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110172382.6A Active CN114908089B (en) 2021-02-08 2021-02-08 Construction method and application of 3' UTR

Country Status (2)

Country Link
CN (1) CN114908089B (en)
WO (1) WO2022166771A1 (en)

Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101076592A (en) * 2004-10-12 2007-11-21 洛克菲勒大学 Micro rna
US20100016411A1 (en) * 2006-08-28 2010-01-21 Peter Leedman Method of modulation of expression of epidermal growth factor receptor(egfr) involving mirna
US20140259192A1 (en) * 2011-07-12 2014-09-11 Sanofi Transgenic animal comprising a deletion or functional deletion of the 3'utr of an endogenous gene
US20160304883A1 (en) * 2013-12-30 2016-10-20 Curevac Ag Artificial nucleic acid molecules
US20160347803A1 (en) * 2015-05-26 2016-12-01 "Nextgen" Company Limited Nucleotide sequence and pharmaceutical composition based thereon with prolonged vegf transgene expression
CN107207557A (en) * 2015-01-26 2017-09-26 中国科学院动物研究所 MiRNA is to m6A modifies the regulation and control method and its application of level
US20180195077A1 (en) * 2015-07-16 2018-07-12 Cornell University Methods of enhancing translation ability of rna molecules, treatments, and kits
US20190100761A1 (en) * 2016-04-06 2019-04-04 Duke University Compositions and methods for enhanced gene expression and viral replication
CN109628489A (en) * 2019-01-07 2019-04-16 新乡医学院 A kind of method and its application improving Chinese hamster ovary celI recombinant protein expression, expression vector, expression system and preparation method thereof
US20190264214A1 (en) * 2016-10-31 2019-08-29 Cornell University Methods of enhancing translation ability and stability of rna molecules, treatments, and kits
US20190292539A1 (en) * 2016-07-14 2019-09-26 Oxford University Innovation Limited Method for modifying genes
US20200376142A1 (en) * 2018-02-19 2020-12-03 Combined Therapeutics, Inc. Compositions and methods for organ-protective expression and modulation of coding ribonucleic acids

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101076592A (en) * 2004-10-12 2007-11-21 洛克菲勒大学 Micro rna
US20100016411A1 (en) * 2006-08-28 2010-01-21 Peter Leedman Method of modulation of expression of epidermal growth factor receptor(egfr) involving mirna
US20140259192A1 (en) * 2011-07-12 2014-09-11 Sanofi Transgenic animal comprising a deletion or functional deletion of the 3'utr of an endogenous gene
US20160304883A1 (en) * 2013-12-30 2016-10-20 Curevac Ag Artificial nucleic acid molecules
CN107207557A (en) * 2015-01-26 2017-09-26 中国科学院动物研究所 MiRNA is to m6A modifies the regulation and control method and its application of level
US20160347803A1 (en) * 2015-05-26 2016-12-01 "Nextgen" Company Limited Nucleotide sequence and pharmaceutical composition based thereon with prolonged vegf transgene expression
US20180195077A1 (en) * 2015-07-16 2018-07-12 Cornell University Methods of enhancing translation ability of rna molecules, treatments, and kits
US20190100761A1 (en) * 2016-04-06 2019-04-04 Duke University Compositions and methods for enhanced gene expression and viral replication
US20190292539A1 (en) * 2016-07-14 2019-09-26 Oxford University Innovation Limited Method for modifying genes
US20190264214A1 (en) * 2016-10-31 2019-08-29 Cornell University Methods of enhancing translation ability and stability of rna molecules, treatments, and kits
US20200376142A1 (en) * 2018-02-19 2020-12-03 Combined Therapeutics, Inc. Compositions and methods for organ-protective expression and modulation of coding ribonucleic acids
CN109628489A (en) * 2019-01-07 2019-04-16 新乡医学院 A kind of method and its application improving Chinese hamster ovary celI recombinant protein expression, expression vector, expression system and preparation method thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
FRANCESCO GHINI等: "Endogenous transcripts control miRNA levels and activity in mammalian cells by target-directed miRNA degradation", 《NATURE COMMUNICATIONS》, vol. 9, pages 3119 *
JIAN ZHENG等: "CRNDE Promotes Malignant Progression of Glioma by Attenuating miR-384/PIWIL4/STAT3 Axis", 《THE AMERICAN SOCIETY OF GENE & CELL THERAPY》, vol. 24, no. 7, pages 1199 - 1215, XP055955754, DOI: 10.1038/mt.2016.71 *

Also Published As

Publication number Publication date
WO2022166771A1 (en) 2022-08-11
CN114908089B (en) 2024-03-15

Similar Documents

Publication Publication Date Title
JP6842495B2 (en) RNA preparation containing purified modified RNA for reprogramming cells
RU2752882C2 (en) Closed-end linear duplex dna for non-viral gene transfer
CN114269919B (en) Targeted RNA editing with engineered RNA using endogenous ADAR
ES2735531T3 (en) RNA containing modified nucleosides and methods of use thereof
CN101805750B (en) Construction and application of farnesyl pyrophosphoric acid synthetase RNA (Ribonucleic Acid) interference recombinant lentivirus vector
RU2693088C2 (en) Microvesicle and method for its preparation
KR20210125560A (en) Disruption of splice receptor sites of disease-associated genes using an adenosine deaminase base editor, including for treatment of hereditary diseases
AU2023237106A1 (en) Aav vectors for treatment of dominant retinitis pigmentosa
KR20220066225A (en) Compositions and methods for selective gene regulation
EA024878B1 (en) Gene encoding human glucokinase mutant characterized by enhanced stability, and use thereof for controlling blood glucose or for preventing and treating disturbances of carbonydrate metabolism
JP2022530457A (en) Genetically engineered AAV
CN104130977A (en) Antitumor medicine screening cell model and application thereof
US20230383275A1 (en) Sgrna targeting aqp1 rna, and vector and use thereof
CN116949041A (en) Artificially designed mRNA UTR nucleotide sequence and its use
CN114908089B (en) Construction method and application of 3&#39; UTR
CN106086069B (en) Transgenic compound and preparation method and application thereof
CN109266684B (en) Method for constructing animal model with pathogen infection sensitivity
KR100427259B1 (en) Novel gene delivery system consisting of cationic polymers and fusogenic peptides
CN114480309B (en) shRNA lentivirus for inhibiting ALKBH1 expression and preparation and application thereof
WO2022111637A1 (en) Nucleic acid molecule binding to yb-1 protein
CN105968211A (en) Recombinant antiviral protein as well as preparation method and application thereof
CN116694630B (en) Sequence combination for promoting overexpression of circular RNA and application thereof
CN117187215A (en) CRISPR/CasRx system capable of being reassembled under induction of chemical small molecules and application thereof
WO2022256414A1 (en) Rna recognition complex and uses thereof
CN116814688A (en) Vector carrying bispecific promoter, construction method and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant
CP03 Change of name, title or address

Address after: 201805 No. 1585 Garden National Road, Anting Town, Jiading District, Shanghai

Patentee after: Shanghai Cell Therapy Group Co.,Ltd.

Country or region after: China

Address before: 201805 No. 1585 Garden National Road, Anting Town, Jiading District, Shanghai

Patentee before: SHANGHAI CELL THERAPY GROUP Co.,Ltd.

Country or region before: China