CN114908089B - Construction method and application of 3' UTR - Google Patents

Construction method and application of 3' UTR Download PDF

Info

Publication number
CN114908089B
CN114908089B CN202110172382.6A CN202110172382A CN114908089B CN 114908089 B CN114908089 B CN 114908089B CN 202110172382 A CN202110172382 A CN 202110172382A CN 114908089 B CN114908089 B CN 114908089B
Authority
CN
China
Prior art keywords
gene
mrna
utr
sequence
egfp
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
CN202110172382.6A
Other languages
Chinese (zh)
Other versions
CN114908089A (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

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 present 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 region sequence for improving mRNA translation, and a construction method and application thereof.
Background
Messenger ribonucleic acid (mRNA) contains, in addition to coding sequences (CDS) necessary for translation of the protein, also necessary untranslated sequences such as m7G caps, 5 '-terminal untranslated regions (5'untranslated region,5 '-UTR), 3' -terminal untranslated regions (3'untranslated region,3'UTR) and poly (A) tail.
The 3'untranslated region is tightly connected with the mRNA coding region, is closely related to post-transcriptional regulation, influences the translation efficiency, stability and the like of the mRNA in cytoplasm through the combination of other regulatory factors, for example, small molecule ribonucleic acid (miRNA) is combined with the 3' untranslated region, so as to inhibit the translation of the mRNA and even induce the degradation of the mRNA; the silencer sequence in the 3' untranslated region binds to the RNA binding protein and can also inhibit translation. In addition, the longer the 3' untranslated region sequence, the greater the likelihood of negatively impacting protein translation. The 3' -UTR of the prior art is usually isolated from the animal itself without modification, and its effect may be affected by the host with uncertainty. There remains a need in the art for a universal 3' UTR region that enhances translation of mRNA.
Disclosure of Invention
The invention constructs 3 'end non-translation region sequences through natural gene modification or artificial design, and the 3' end non-translation region sequences ensure that the mRNA of the gene keeps higher translation activity in different cells.
The first aspect of the present invention provides a polynucleotide comprising
(1) A 3'UTR sequence which is a sequence knocked out by one or more microRNA recognition sites in the 3' UTR region of the PCBP4 gene or PIWIL4 gene, and/or
(2) The complement of (1).
In one or more embodiments, the nucleotide sequence of the 3' UTR region of the PCBP4 gene is as shown 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 as shown 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 retrieved using the http:// www.targetscan.org/vert_72/website.
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 polynucleotide has a sequence selected from the group consisting of (1) the sequences set forth in any one of SEQ ID NOs 3-7, (2) sequences 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 of constructing a 3'UTR sequence comprising the step of knocking out recognition sites for one or more microRNAs in the 3' UTR region of a gene.
In one or more embodiments, the gene is a PCBP4 gene or a 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 as shown 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 as shown 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 is retrieved using the http:// www.targetscan.org/vert_72/website.
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 invention provides a polynucleotide comprising
(1) A 3' UTR sequence containing a sequence of at least 3 tandem repeats of an RNA methylation (m 6A) site and optionally a linker between any two repeats, and/or
(2) The complement of (1).
In one or more embodiments, the m6A site is a recognition sequence for an m6A methylase (writer) or an m6A RNA methylation recognition protein (reader). In one or more embodiments, the m6A methylase is selected from the group consisting of METTL3, METTL14, METTL16, WTAP, VIRMA, ZC H13, RBM15, RBM158, HAKAI, and KIAA1492. 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, LRPPRC, eIF3, and HNRNPA2B1.
In one or more embodiments, the m6A site is selected from: AAABH, AGABH, GAABH, GGABH where b=t or C or G, h=t or a or C.
In one or more embodiments, the m6A site is selected from: GGACT, GGACC, GAACT.
In one or more embodiments, the 3' utr sequence contains 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 site.
In one or more embodiments, the linker is 1-5bp, preferably 3bp in length.
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 polynucleotide has a sequence selected from the group consisting of (1) the sequences set forth in any one of SEQ ID NOS: 8-14, (2) sequences 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 concatenating at least 3 RNA methylation (m 6A) sites and optionally adding restriction enzyme sites at both ends.
In one or more embodiments, the connection is a direct connection or a connection 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 for an m6A methylase or an m6A RNA methylation recognition protein. In one or more embodiments, the m6A methylase is selected from the group consisting of METTL3, METTL14, METTL16, WTAP, VIRMA, ZC H13, RBM15, RBM158, HAKAI, and KIAA1492. 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, LRPPRC, eIF3, and HNRNPA2B1.
In one or more embodiments, the m6A site is selected from: AAABH, AGABH, GAABH, GGABH where b=t or C or G, h=t or a or C.
In one or more embodiments, the m6A site is selected from: GGACT, GGACC, GAACT.
In one or more embodiments, the method comprises the step of concatenating at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10 RNA methylation (m 6A) sites.
In one or more embodiments, the RNA methylation (m 6A) sites in the 3' utr sequence are identical.
The invention also provides a nucleic acid construct comprising a polynucleotide as described in 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 at the 3' end of 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: promoters, 5' UTR, multiple cloning sites, polyA.
In one or more embodiments, the promoter is a 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, at least in order: promoters, multiple cloning sites, polynucleotides as described in any of the embodiments herein, and 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 as described in 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 as described in any of the embodiments herein, and a pharmaceutically acceptable adjuvant.
In one or more embodiments, the polynucleotide is located at the 3' end of the gene to be expressed.
The invention also provides a method of increasing the intensity of gene expression, increasing the time of gene expression, increasing the ability of mRNA to initiate translation, or maintaining the length of translation activity, comprising the step of expressing a gene carrying a polynucleotide as described in any of the embodiments 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 as described in any of the embodiments herein in the preparation of an agent for increasing the intensity of gene expression, increasing 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 as described in any of the embodiments herein, said polynucleotide being 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 the treatment of a disease.
The invention also provides the use of a polynucleotide according to any 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' UTRs described herein.
FIG. 2 shows the proportion of eGFP-expressing fluorescence 24h after mRNA electrotransformation of PBMC cells with different 3' UTRs.
FIG. 3 shows the mean fluorescence intensity after 24h of PBMC electrotransformation of mRNA containing different 3' UTRs.
FIG. 4 shows eGFP positive rates after 72h mRNA electrotransformation of PBMC with different 3' UTRs.
FIG. 5 shows the mean fluorescence intensity of eGFP mRNA from different 3' UTRs after electrotransformation of PBMC for 72 h.
FIG. 6 shows eGFP positivity 24h after CHO cell transfection of mRNA for different 3' UTRs.
FIG. 7 shows the average fluorescence intensity of eGFP 24h after transfection of mRNA with CHO cells.
FIG. 8 shows eGFP positivity after 48h of mRNA transfection by CHO cells with different 3' UTRs.
FIG. 9 shows the mean fluorescence intensity of eGFP after 48h of mRNA transfection by CHO cells with different 3' UTRs.
FIG. 10 shows the eGFP positive rate 96h after transfection of CHO cells with different 3' UTRs.
FIG. 11 shows the mean fluorescence intensity of eGFP 96h after transfection of mRNA with CHO cells for different 3' UTRs.
Detailed Description
It is understood that within the scope of the present invention, the above-described technical features of the present invention and technical features specifically described below (e.g., in the examples) may be combined with each other to constitute a preferred technical solution.
As used herein and in the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. Also, the terms "a" (or "an"), "one or more" and "at least one" can be used interchangeably herein. The term "comprising" and its variants are not to be taken in a limiting sense, wherein these terms appear in the description and in the claims. Thus, the terms "comprising," "including," and "containing" are used interchangeably.
The invention constructs 3 'end non-translation region sequences through natural gene modification or artificial design, and the 3' end non-translation region sequences ensure that the mRNA of the gene keeps higher translation activity in different cells.
Thus, in one or more embodiments, the invention comprises a 3' utr polynucleotide obtained by natural genetic engineering comprising a 3' utr sequence, or a complement thereof, that is a sequence that is knocked out of one or more microRNA recognition sites in the 3' utr region of a natural gene (e.g., a human PCBP4 gene or a PIWIL4 gene). The 3'UTR sequence of PCBP4 mRNA (NM_ 001174100) is shown as SEQ ID NO. 1, and the 3' UTR sequence of PIWIL4 mRNA (NM_ 152431) is shown as SEQ ID NO. 2. Herein, a microRNA recognition site is a site recognized by micrornas and affecting mRNA stability. The sequence of some microRNA recognition sites can be obtained by searching with http:// www.targetscan.org/vert_72/website. In specific 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 sequence in 3' UTR of 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 sequence in the 3' UTR of the PIWIL4 Gene
The method for constructing the 3' UTR sequence comprises the steps of knocking out the recognition site of one or more microRNAs in the 3' UTR region of the gene and optionally adding restriction enzyme cutting sites at two ends of the 3' UTR region. Knocking out microRNA recognition sites can be accomplished by any method known in the art, such as mutating the 2-8nt target sequence in the 3' UTR that is complementary to the seed sequence (seed) of the microRNA. In a specific embodiment, primers are designed based on the 3' UTR of a gene (e.g., human PCBP4 gene or PIWIL4 gene), corresponding sequences are amplified from the RNA set by RT-PCR, and then mutations are introduced by PCR for knockout. Alternatively, the 3' UTR sequences of the invention which are engineered by natural genes can also be obtained directly synthetically. Exemplary sequences of the polynucleotides are shown in any one of SEQ ID NOs 3-7.
In other embodiments, the invention encompasses 3'utr polynucleotides obtained by artificial design, comprising a 3' utr sequence or its complement comprising a sequence of at least 3 tandem repeats of an RNA methylation (m 6A) site and optionally a linker between any two repeats. Herein, the m6A site is a recognition site for an m6A methylase or an m6A RAN methylation recognition protein. Illustratively, the m6A methylases may be METTL3, METTL14, METTL16, WTAP, VIRMA, ZC H13, RBM15, RBM158, HAKAI and KIAA1492; the m6A RNA methylation recognition protein can be YTHDC1-2, YTHDF1-3, IGF2BP1-3, prrc2a, HNRNPC, HNRNPG, FMR1, SRSF2, huR, LRPPRC, eIF and HNRNPA2B1. The m6A site useful in the present invention is selected from: AAABH, AGABH, GAABH, GGABH where 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 site. Multiple RNA methylation (m 6A) sites in a 3' utr sequence may be the same or different. The method of constructing the 3' UTR sequence includes the steps of concatenating at least 3 RNA methylation (m 6A) sites and optionally adding restriction enzyme sites at both ends. The ligation is either direct or through a linker (e.g., an oligonucleotide of 1-5bp in length). Alternatively, the 3' UTR sequences of the invention which are designed artificially can also be obtained directly synthetically. Exemplary sequences of the polynucleotides are shown in any one of SEQ ID NOs 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 when referring to a nucleic acid may be a naturally occurring allelic variant or a non-naturally occurring variant. Such nucleotide variants include degenerate variants, substitution variants, deletion variants and insertion variants. The nucleic acids of the invention may 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 which hybridize to the sequences described above. The polynucleotides described herein can generally be obtained using PCR amplification methods. Alternatively, the nucleic acid molecules described herein can also be synthesized directly.
In two or more polypeptide or nucleic acid molecule sequences, the term "identity" or "percent identity" refers to two or more sequences or subsequences that are the same or wherein a percentage of amino acid residues or nucleotides in a specified region 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) when compared and aligned for maximum correspondence over a comparison window or specified region using methods known in the art, such as sequence comparison algorithms, by manual alignment and visual inspection. For example, preferred algorithms 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 operations, it is often necessary to design suitable cleavage sites, which tend to introduce one or more cleavage sites at the end of the polynucleotide, without affecting 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 having one or more cleavage sites added at both ends thereof.
The 3' UTR polynucleotides of the invention can improve expression of the cognate gene. The improvement of expression is achieved by introducing into the cell a nucleic acid construct carrying a gene of a polynucleotide as described herein. Accordingly, the present invention also provides a nucleic acid construct of the polynucleotide. 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 operably linked thereto, 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 site, 5' UTR. The coding sequence of the gene to be expressed may be inserted into the 5 'end of the 3' UTR polynucleotide in the nucleic acid construct. The polynucleotides of the invention may be manipulated in a variety of ways to ensure expression of the genes. The nucleic acid construct may be manipulated according to the vector or requirements prior to insertion into the vector. Techniques for altering polynucleotide sequences using recombinant DNA methods are known in the art.
The nucleic acid construct may also be an mRNA containing the gene to be expressed and the 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 is obtained, for example, according to the instructions of the HiScribe T7 ARCA mRNA kit.
In recombinant expression vectors, "operably linked" refers to the attachment of a nucleotide sequence of interest to a regulatory sequence in a manner that allows expression of the nucleotide sequence. Methods are well known to those skilled in the art for constructing expression vectors containing the coding sequences of the 3' UTR polynucleotides described herein and optionally the gene of interest and appropriate transcriptional/translational control signals. These methods include in vitro recombinant DNA techniques, DNA synthesis techniques, in vivo recombinant techniques, and the like.
The regulatory sequence may be a suitable 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 that exhibits 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 levels of expression of any polynucleotide sequence operably linked thereto. Other constitutive promoter sequences may also be used, including but not limited to the T3 promoter, the SP6 promoter.
The regulatory sequences may also be suitable transcription termination sequences, sequences recognized by the host cell to terminate transcription. The termination sequence is operably linked to the 3 'end 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 sequences may also be suitable leader sequences, 5' untranslated regions of mRNA that are important for translation by the host cell. The leader sequence is operably linked to the 5' terminus 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. Polynucleotides of the invention may be cloned into many types of vectors, e.g., plasmids, phagemids, phage derivatives, animal viruses and cosmids. Cloning vectors may be used to provide coding sequences for exogenous genes and polynucleotides described herein. The expression vector may be provided to the cell as 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 useful for regulating expression of the desired nucleic acid sequence. Viral vector techniques are well known in the art and are 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 may be used as vectors include, but are not limited to, retroviruses, adenoviruses, adeno-associated viruses, herpesviruses, and lentiviruses. Homologous recombination vectors are used to integrate the expression cassettes described herein into the host genome.
In general, 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 a replication origin, a promoter, a polynucleotide as described herein, and optionally a selectable marker.
To assess expression of the therapeutic protein, polypeptide, or portion thereof, the expression vector introduced into the cell may also comprise either or both of a selectable marker gene or a reporter gene to facilitate identification and selection of the expressing cell from a population of cells sought to be transfected or infected by the viral vector. In other aspects, the selectable marker may be carried on a single 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 the 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 assayed at the appropriate time. Suitable reporter genes may include genes encoding luciferase, beta-galactosidase, chloramphenicol acetyl transferase, secreted alkaline phosphatase, or green fluorescent protein genes. Suitable expression systems are well known and can be prepared using known techniques or commercially available.
Methods for introducing genes into cells and expressing genes into cells are known in the art. The vector may be readily introduced into a host cell, e.g., a mammalian, bacterial, yeast or insect cell, by any method known in the art. For example, the expression vector may be transferred into the 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, electrotransfection, and the like, e.g., directly introducing in vitro transcribed mRNA into cells by electrotransformation. Chemical means for introducing the polynucleotide into a host cell 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 may be inserted into a vector and packaged into retroviral particles using techniques known in the art. The recombinant virus may then be isolated and delivered to a subject cell in vivo or ex vivo. Many retroviral systems are known in the art. Lentiviruses are genera under the retrovirus family. Agents for lentiviral packaging are well known in the art, as are conventional lentiviral vector systems including pRsv-REV, pMDlg-pRRE, pMD2G and interfering plasmids of interest.
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 also include various cells used in the production of cells for expression, e.g., e.coli cells, for use in, e.g., providing a polynucleotide of the invention or providing a vector as described herein. Cells suitable for use in the present invention may be of various types of cells of various origins, 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 that facilitate expression or fermentative production of the gene product, such cells being well known and commonly used in the art.
Also included herein are cell preparations comprising the cells described herein or extracts thereof. For example, the cell preparation may be a cell culture, pharmaceutical composition, kit, device, medium or system, such as a chip or the like, comprising the cells or extracts thereof described herein and a suitable medium. Suitable culture media for culturing various types of cells are well known in the art.
The function of the nucleic acid construct or cell described herein depends on the gene to be expressed carried therein, for example, if the gene to be expressed is a fluorescent protein, the nucleic acid construct or cell described herein may be used for cell tracking. If the gene to be expressed is an agent useful in the treatment of a disease, the nucleic acid constructs or cells described herein may be used to treat the disease.
The present 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 a nucleic acid construct) and a pharmaceutically acceptable diluent, carrier, solubilizer, emulsifier, preservative, and/or adjuvant. In certain embodiments, acceptable diluents, carriers, solubilizers, emulsifiers, preservatives, and/or adjuvants and the like in the pharmaceutical compositions are preferably non-toxic to the recipient at the dosages and concentrations employed. In certain embodiments, the pharmaceutical compositions may contain such materials for improving, maintaining, or retaining, for example, pH, permeability, viscosity, clarity, color, isotonicity, odor, sterility, stability, dissolution or release rate, absorption or permeation of the composition. These substances are known from the prior art and can be found, for example, in REMINGTON' S PHARMACEUTICAL SCIENCES, 18 th edition, A.R. Genrmo, code 1990,Mack Publishing Company. The optimal pharmaceutical composition can be determined depending on the intended route of administration, the mode of delivery and the dosage required.
The route of administration of the pharmaceutical composition is according to known methods, for example, by oral, intravenous, intraperitoneal, intracerebral (intraparenchymal), intracerebroventricular, intramuscular, intraocular, intraarterial, portal or intralesional route injection; either 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 particles or porous beads, and depot injections, are also known to those skilled in the art.
Pharmaceutical compositions for in vivo administration are generally provided in the form of sterile formulations. Sterilization is achieved by filtration through sterile filtration membranes. In the case of lyophilization of a composition, this method may be used to sterilize the composition either 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 tape 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 (e.g., lyophilized) prior to administration. The invention also provides kits for producing single dose administration units. 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 prefilled 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 or pharmaceutical composition thereof according to any of the embodiments of the invention. The terms "patient," "subject," "individual," "subject" are used interchangeably herein to 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 a subject employing a treatment regimen described herein to achieve at least one positive therapeutic effect. The treatment regimen effective to treat a patient can vary depending on a variety of factors, such as the disease state, age, weight, and ability of the patient to elicit an anti-cancer response in the subject by therapy.
The therapeutically effective amount of the pharmaceutical composition comprising the nucleic acid construct or cell of the invention to be employed will depend, for example, on the degree of treatment and the goal. Those 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 target gene expression in a 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 dose and alter the route of administration to obtain the optimal therapeutic effect.
The frequency of administration will depend on the pharmacokinetic parameters of the particular target product in the formulation used (e.g., the product of target gene expression in a nucleic acid construct). The clinician typically administers the composition until a dose is reached that achieves the desired effect. The composition may thus be administered as a single dose, or over time as two or more doses (which may or may not contain the same amount of the desired molecule), or as a continuous infusion through an implanted device or catheter.
As demonstrated in the examples below, the 3' UTR polynucleotides described herein can increase gene expression intensity, increase gene expression time, increase mRNA initiation translation ability, or maintain translation activity for a prolonged period of time. Thus, the invention also provides a method of increasing the intensity of gene expression, increasing the time of gene expression, increasing the ability of an mRNA to initiate translation, or maintaining the length of translation activity, comprising the step of expressing a gene carrying a 3' utr polynucleotide as described in any of the embodiments herein. Typically, the 3'utr polynucleotide is located at the 3' end of the gene. Illustratively, expression is achieved by the above-described procedure of introducing the nucleic acid construct into a cell 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 a3' utr polynucleotide, nucleic acid construct and/or host cell according to any of the embodiments herein in the manufacture of a medicament for the treatment of a disease, the 3' utr polynucleotide being located at the 3' end of a gene, expression of the gene being conducive to the treatment of the disease. The nucleic acid construct comprising the gene and the 3' UTR polynucleotide may be introduced into a cell by the methods 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 the treatment of a disease in which the gene or product thereof directly produces a therapeutic effect, or may be such that expression of the gene or product thereof causes expression or activation of other genes or proteins that produce a therapeutic effect.
Examples
EXAMPLE 1 construction of 3' untranslated region transcription template vector
1) Construction of transcription template vector for expressing eGFP (enhanced green fluorescent protein) reporter gene based on 3' -untranslated region modified from human natural gene
Construction of vector pT7-m5U-eGFP-1#3U
The modified sequence (SEQ ID NO: 3) based on the untranslated region at the 3' -end of the human PCBP4 gene was synthesized by Jin Wei Intelligence, and the vector pUC57-1#3U was digested with Sal I and HindIII, whereby a fragment 1#3U having a size of 430bp was recovered. The self-constructed EGFP transcription template vector pT7-m5U-eGFP contains a T7 promoter, a 5'UTR sequence (m 5U) of HBBmRNA (NM-000518), an EGFP sequence, a HBB mRNA3' UTR sequence and a polyA sequence (SEQ ID NO: 16). After double digestion with Sal I and HindIII, the recovered fragment 1#3U was ligated to pT7-m5U-eGFP vector to replace the original HBB mRNA3' UTR, thereby obtaining pT7-m5U-eGFP-1#3U.
FIG. 1 shows schematically the map of pT7-m5U-eGFP-1#3U, with the subsequent vector maps differing therefrom by the use of different 3' UTRs.
Construction of vector pT7-m5U-eGFP-2#3U
The method comprises the steps of (1) obtaining a sequence (SEQ ID NO: 4) modified based on an untranslated region at the 3' -end of a human PCBP4 gene by a Jin Wei intelligent company, carrying out double digestion on a vector pUC57-2#3U by Sal I and HindIII, recovering a fragment 2#3U with the size of 143bp, carrying out double digestion on a pT7-m5U-eGFP vector by Sal I and HindIII, and connecting the recovered fragment 2#3U to the pT7-m5U-eGFP-2#3U.
Construction of vector pT7-m5U-eGFP-3#3U
The method comprises the steps of (1) obtaining a sequence (SEQ ID NO: 5) modified based on an untranslated region at the 3' -end of a human PCBP4 gene by a Jin Wei intelligent company, carrying out double digestion on a vector pUC57-3#3U by Sal I and HindIII, recovering a fragment 3#3U with the size of 443bp, carrying out double digestion on a pT7-m5U-eGFP vector by Sal I and HindIII, and then connecting the recovered fragment 3#3U to the pT7-m5U-eGFP-3#3U.
Construction of vector pT7-m5U-eGFP-4#3U
The method comprises the steps of (1) obtaining a sequence (SEQ ID NO: 6) modified based on an untranslated region at the 3' -end of a human PCBP4 gene by a Jin Wei intelligent company, carrying out double digestion on a vector pUC57-4#3U by Sal I and HindIII, recovering a fragment 4#3U with the size of 430bp, carrying out double digestion on a pT7-m5U-eGFP vector by Sal I and HindIII, and connecting the recovered fragment 4#3U to the pT7-m5U-eGFP-4#3U.
Construction of vector pT7-m5U-eGFP-5#3U
The Jin Weizhi company is entrusted with synthesizing a sequence (SEQ ID NO: 7) modified based on an untranslated region at the 3' -end of the adult PIWIL4 gene, double-enzyme-cutting the vector pUC57-5#3U by Sal I and HindIII, recovering the fragment 5#3U with the size of 124bp, double-enzyme-cutting the pT7-m5U-eGFP vector by Sal I and HindIII, and connecting the recovered fragment 5#3U to the pT7-m5U-eGFP-5#3U to obtain the pT7-m5U-eGFP-5#3U.
2) Construction of transcription template vector for expressing eGFP reporter gene with 3' -terminal untranslated region as artificial random sequence
Construction of vector pT7-m5U-eGFP-6#3U
The artificial random sequence (SEQ ID NO: 8) is synthesized by Jin Wei intelligent company, sal I and HindIII are used for double enzyme digestion of vector pUC57-6#3U, 100bp fragment 6#3U is recovered, pT7-m5U-eGFP vector is subjected to double enzyme digestion by Sal I and HindIII, and the recovered fragment 6#3U is connected to pT7-m5U-eGFP vector to obtain pT7-m5U-eGFP-6#3U.
Construction of vector pT7-m5U-eGFP-7#3U
The artificial random sequence (SEQ ID NO: 9) is synthesized by Jin Wei intelligent company, sal I and HindIII are used for double enzyme digestion of vector pUC57-7#3U, 116bp fragment 7#3U is recovered, pT7-m5U-eGFP vector is subjected to Sal I and HindIII double enzyme digestion, and the recovered fragment 7#3U is connected to pT7-m5U-eGFP vector to obtain pT7-m5U-eGFP-7#3U.
Construction of vector pT7-m5U-eGFP-8#3U
The artificial random sequence (SEQ ID NO: 10) was synthesized by Jin Wei Intelligence, the Sal I and HindIII double-digested vector pUC57-8#3U was used to recover 86bp fragment 8#3U, the pT7-m5U-eGFP vector was double-digested by Sal I and HindIII, and the recovered fragment 8#3U was ligated to the pT7-m5U-eGFP vector to obtain pT7-m5U-eGFP-8#3U.
Construction of vector pT7-m5U-eGFP-9#3U
The artificial random sequence (SEQ ID NO: 11) was synthesized by Jin Wei, the vector pUC57-9#3U was digested with Sal I and HindIII, the 92bp fragment 9#3U was recovered, the pT7-m5U-eGFP vector was digested with Sal I and HindIII, and the recovered fragment 9#3U was ligated to the 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 Jin Wei Intelligence, the Sal I and HindIII double-digested vector pUC57-10#3U was used to recover the fragment 10#3U of 103bp, the pT7-m5U-eGFP vector was double-digested by Sal I and HindIII, and 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
The artificial random sequence (SEQ ID NO: 13) was synthesized by Jin Wei Italy, the Sal I and HindIII double-digested vector pUC57-11#3U was used to recover the 97bp fragment 11#3U, the pT7-m5U-eGFP vector was double-digested by Sal I and HindIII, and 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
The artificial random sequence (SEQ ID NO: 14) was synthesized by Jin Wei Intelligence, sal I and HindIII were used to double-cleave the vector pUC57-12#3U, the 97bp fragment 12#3U was recovered, pT7-m5U-eGFP vector was double-cleaved by Sal I and HindIII, and the recovered fragment 12#3U was ligated to pT7-m5U-eGFP vector to obtain pT7-m5U-eGFP-12#3U.
3) Construction of transcription template vector for expressing eGFP reporter gene with 3' -terminal untranslated region as natural gene sequence
Construction of vector pT7-m5U-eGFP-PCBP4-3U
The PCBP4 mRNA 3' UTR sequence (SEQ ID NO: 1) was synthesized by Jin Wei Intelligence, sal I and HindIII double-digested vector pUC57-PCBP4#3U was used to recover 609bp fragment PCBP4#3U, pT7-m5U-eGFP vector was digested with Sal I and HindIII double, and the recovered fragment PCBP4#3U was ligated to pT7-m5U-eGFP vector to obtain pT7-m5U-eGFP-PCBP4#3U.
Construction of vector pT7-m5U-eGFP-PIWIL4-3U
The PIWIL4 mRNA 3' UTR sequence (SEQ ID NO: 2) was synthesized by Jin Wei Intelligence, the fragment PIWIL4#3U of 429bp was recovered by double digestion of the vector pUC57-PIWIL4#3U with Sal I and HindIII, the fragment PIWIL4#3U was recovered by double digestion of the vector pT7-m5U-eGFP with Sal I and HindIII, and the recovered fragment PIWIL4#3U was ligated to the pT7-m5U-eGFP vector to obtain pT7-m5U-eGFP-PIWIL4#3U.
Construction of vector pT7-m5U-eGFP-IL2#3U
The IL-2' UTR sequence (SEQ ID NO: 15) was synthesized by Jin Wei Italy, sal I and HindIII double digestion vectors pUC57-IL2#3U were used to recover the 285bp fragment IL2#3U, pT7-m5U-eGFP vector was digested with Sal I and HindIII double digestion, and the recovered fragment IL2#3U was ligated to pT7-m5U-eGFP vector to obtain pT7-m5U-eGFP-IL2#3U.
Example 2 in vitro transcription of mRNA containing an engineered or engineered 3' UTR
The other 5 ' untranslated region (5`untranslated region,5`UTR), the eGFP coding sequence (CDS) and the poly (A)) template sequences were all identical with the HBB mRNA 3' UTR as a control 3' UTR sequence. Digestion of pT7-m5U-eGFP, pT7-m5U-eGFP-1#3U, pT7-m5U-eGFP-2#3U, pT7-m5U-eGFP-3#3U, pT7-m5U-eGFP-4#3U, pT7-m5U-eGFP-5#3U, pT7-m5U-eGFP-6#3U, pT7-m5U-eGFP-7#3U, pT7-m5U-eGFP-8#3U, pT7-m5U-eGFP-9#3U, pT7-m5U-eGFP-10#3U, pT7-m 5U-eGFP-11U, pT7-m5U-eGFP-12#3U, GFP 7-m5U-eGFP-7#3U, pT7-m 5U-eGFP-7#7U-7#3U, pT 7-m7#7U-vL#7L#7L 3#7L 3, and PIL 35-37-3In vitro transcribed mRNA was obtained by the kit (NEB, E2065S) protocol: the following reagents were added to the nuclease-free tube: 2 XARCA/NTP Mix 10. Mu.L, bsaI linearized plasmid template 1. Mu.g, T7 RNA Polymerase Mix. Mu.l, H 2 O to 20. Mu.L; incubation at 37 ℃ for 30 minutes; adding 2 mu L of DNase, and digesting for 15min; precipitating for 2h using LiCl solution provided with the kit; centrifuging at 12000rpm for 2min; washing with 75% ethanol for 2 times, sucking all supernatant, and air drying at room temperature; h 2 O dissolves and precipitates 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 detection of maintenance of Gene expression in T cells by constructed 3' UTR
1) Determination of the effect of 3' untranslated region sequences on mRNA translation by eGFP 24h expression intensity:
the other 5 ' untranslated region (5`untranslated region,5`UTR), coding sequence (CDS) and poly (A)) sequences were all identical with the HBB mRNA 3' UTR as a control 3' UTR sequence. Resuscitates and resuspension 5 x 10 6 The peripheral blood mononuclear cells (peripheral blood mononuclear cell, PBMC) obtained by fresh isolation were subjected to electric transfer of 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-eGFP PCBP4#, mRNA-eGFP PIWIL4#, mRNA-eGFPIL 2#) obtained in examples 1-2 into PBMC using Lonza 4D-nucleic acid 2 Culturing in incubator with AIM-V (Gibco) and 2% foetal calf serum (Gibco). Cell counts after 24 hours and 3X 10 were taken 5 Cells were analyzed for fluorescent expression using flow cytometry.
FIG. 2 shows the proportion of eGFP-expressing fluorescence 24h after mRNA electrotransformation of PBMC cells with different 3' UTRs, and no significant difference in the electrotransformation efficiency of each mRNA group in PBMC was seen. FIG. 3 shows the average fluorescence intensity after 24h electrotransformation of PBMC with mRNA containing different 3 'UTRs, compared with the eGFP expression intensity of in vitro transcribed mRNA containing artificially engineered or engineered 3' UTRs, which is significantly better than mRNA containing natural 3 'UTRs such as PCBP4, PIWIL4, IL-2, etc., in PBMC, wherein mRNA-eGFP8#, mRNA-eGFP9# and mRNA-eGFP10# are significantly better than mRNA containing HBB 3' UTRs.
2) Determination of the influence of the 3' untranslated region sequence on the translation time of mRNA in T cells by eGFP 72h expression intensity
The other 5 ' untranslated region (5`untranslated region,5`UTR), coding sequence (CDS) and poly (A)) sequences were all identical with the HBB mRNA 3' UTR as a control 3' UTR sequence. Resuscitates and resuspension 5 x 10 6 The peripheral blood mononuclear cells (peripheral blood mononuclear cell, PBMC) obtained by fresh isolation were electrically transferred with Lonza 4D-nucleic acid selector 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-eGFP PCBP4#, mRNA-eGFP PIWIL4#, mRNA-eGFPIL 2#) obtained in examples 1-2, and cultured in a 5% CO2 incubator at 37℃with AIM-V (Gibco) and 2% fetal bovine serum (Gibco). After 72h, the cells were counted and taken 3X 10 5 Cells were resuspended in PBS and analyzed for cell positive rate and mean fluorescence intensity using flow cytometry.
FIG. 4 shows eGFP positive rate after mRNA electrotransformation of PBMC for 72h under different 3' UTRs, and shows that the expression time of mRNA of the artificially modified or designed 3' UTR in PBMC is better than that of natural 3' UTRs such as PCBP4, PIWIL4, IL-2 and the like. FIG. 5 shows the average fluorescence intensity of eGFP mRNA in different 3' UTRs after electrotransformation of PBMC for 72h, in comparison with the expression intensity of eGFP transcribed from mRNA containing an artificially engineered or engineered 3' UTR in PBMC, which was found to be stronger than that of 3' UTR of natural sequences such as PCBP4, PIWIL4, IL-2, etc. mRNA containing the engineered or engineered 3'UTR remained highly expressed in PBMC after 72 hours, whereas mRNA containing the 3' UTR of the IL-2 native sequence had been substantially absent.
Example 4 detection of maintenance of Gene expression in CHO cells by constructed 3' UTR
1) CHO cell transfection (Lipo 2000 transfection kit):
the other 5 ' untranslated region (5`untranslated region,5`UTR), coding sequence (CDS) and poly (A)) sequences were all identical with the HBB mRNA 3' UTR as a control 3' UTR sequence.
a. Cell plating: CHO cells well cultured by pancreatin digestion were counted and taken 3×10 5 The cells are resuspended in 3mL of culture solution and spread in a 6-well plate for culturing for 24 hours, wherein the culture medium comprises 45% DMEM culture medium+45% RPMI-1640 culture medium+10% serum+1% L-glutamine+1% Thymidine (hypoxanine);
b. Changing culture solution: after CHO cells are cultured for 24 hours in a 6-hole plate, the original cell culture solution is discarded, 1mL of PBS buffer solution is added for cleaning the cells, 2mL of new culture solution is added into the washing solution, and the 6-hole plate is placed back into a 37 ℃ incubator for culture;
c. transfection reagent (per well): 3 μg (1 μg/. Mu.L) of eGFP 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-eGFP PCBP4#, mRNA-eGFP PIWIL4#, mRNA-eGFPIL 2#) +6 μL Lipo2000 (mRNA: lipo2000 = 1:2) in 200 μL opti-MEM medium, and vortexing for 10s at room temperature for 5-20min to form a complex;
d. adding a transfection mixture: the prepared transfection reagent is rapidly dripped into a 6-well plate paved with CHO cells, the 6-well plate is shaken for a plurality of times and then is put back to 37 ℃ and 5 percent CO 2 Culturing in an incubator for 144h at maximum.
2) Determination of the influence of the sequence of the 3' untranslated region on the translation time and intensity of mRNA in CHO cells by eGFP expression intensity
After 24h, the medium was aspirated, washed 1X PBS for 1 time, CHO cells were digested with pancreatin, counted and 3X 10 was taken 5 The cells are resuspended in 3mL of culture solution and spread in a 6-well plate for culture, wherein the culture medium comprises 45% of DMEM culture medium, 45% of RPMI-1640 culture medium, 10% of serum, 1% of L-glutamine and 1% of thymidine hypoxanthine; another 3X 10 5 Cells were resuspended in 1 XPBS, flow-observed, and analyzed by Kaluza Analysis.
FIG. 6 shows eGFP positivity after 24h transfection of mRNA in CHO cells with different 3 'UTRs, showing no significant difference in transfection efficiency of mRNA in CHO cells with different 3' UTRs. FIG. 7 shows the mean fluorescence intensity of eGFP 24h after transfection of mRNA in CHO cells, showing that in vitro transcription of mRNA containing an engineered or engineered 3'UTR results in CHO cells with enhanced expression of eGFP than 3' UTR of the native sequences PCBP4, PIWIL4, IL-2, etc.
After 48h the medium was aspirated, washed 1X PBS for 1 time, CHO cells were digested with pancreatin, counted and 3X 10 was taken 5 The cells are resuspended in 3mL of culture solution and spread in a 6-well plate for culture, wherein the culture medium comprises 45% of DMEM culture medium, 45% of RPMI-1640 culture medium, 10% of serum, 1% of L-glutamine and 1% of thymidine hypoxanthine; another 3X 10 5 Cells were resuspended in 1 XPBS, flow-observed, and analyzed by FlowJo X.
FIG. 8 shows eGFP positivity after 48h of mRNA transfection in CHO cells for different 3 'UTRs, showing that there is no difference in the persistence of mRNA expression in CHO cells for different 3' UTRs after 48 h. FIG. 9 shows the average fluorescence intensity of eGFP after 48h of transfection of mRNA in CHO cells, showing that the expression intensity of eGFP in CHO cells by in vitro transcription of mRNA containing artificially engineered or engineered 3' UTR is stronger than that of 3' UTR of natural sequences such as PCBP4, PIWIL4, IL-2, etc., and that the fluorescence intensity of mRNA of natural 3' UTR is rapidly decreased after 48 h.
After 96h the medium was aspirated, washed 1X PBS for 1 time, CHO cells were digested with pancreatin, counted and 3X 10 was taken 5 Cells were resuspended in 1 XPBS, flow-observed, and analyzed by FlowJo X.
FIG. 10 shows the eGFP positive rate of CHO cells transfected for 96h under different 3' UTRs, and shows that the expression persistence of eGFP mRNA containing artificially engineered or designed 3' UTRs in CHO cells is superior to that of mRNA containing 3' UTRs of natural sequences such as PCBP4, PIWIL4, IL-2, etc. FIG. 11 shows the mean fluorescence intensity of eGFP in CHO cells 96h after transfection of mRNA, which indicates that in vitro transcription of mRNA containing an engineered or engineered 3'UTR results in CHO cells with enhanced expression of eGFP than IL-2' UTR. It can be seen that after 96 hours the mRNA containing the engineered or engineered 3'UTR still had higher expression activity, whereas the mRNA containing the IL-2 3' UTR had 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 comprising a 3' UTR sequence which is a sequence from which one or more microRNA recognition sites in the 3' UTR region of a PCBP4 gene or PIWIL4 gene have been knocked out, the 3' UTR sequence being as shown in any one of SEQ ID NOs 3 to 7.
2. A method for constructing a 3' UTR sequence shown in any one of SEQ ID NO. 3-7 comprises the step of knocking out recognition sites of one or more microRNAs in a 3' UTR region of a gene, wherein the nucleotide sequence of the 3' UTR region of the gene is shown in SEQ ID NO. 1 or 2.
3. A nucleic acid construct comprising the polynucleotide of claim 1 and optionally a gene to be expressed, said polynucleotide being located at the 3' end of said gene to be expressed.
4. The nucleic acid construct of claim 3, further comprising one or more elements selected from the group consisting of: promoters, 5' UTR, multiple cloning sites, polyA.
5. A host cell comprising the polynucleotide of claim 1 and/or the nucleic acid construct of claim 3 or 4.
6. The host cell of claim 5, wherein the host cell is a CHO cell or a PBMC cell.
7. A pharmaceutical composition comprising the polynucleotide of claim 1, the nucleic acid construct of claim 3 or 4, or the host cell of claim 5 or 6, and a pharmaceutically acceptable adjuvant.
8. A method of increasing the intensity of gene expression, increasing the time of gene expression, increasing the mRNA initiation translation ability of a gene, or maintaining the length of translation activity, comprising the step of expressing a gene carrying the polynucleotide of claim 1, said polynucleotide being located at the 3' end of said gene.
9. Use of the polynucleotide of claim 1, the nucleic acid construct of claim 3 or 4 or the host cell of claim 5 or 6 in the manufacture of a medicament for the treatment of a disease, said polynucleotide being located at the 3' end of a gene whose expression facilitates the treatment of said disease.
10. Use of the polynucleotide of claim 1, said polynucleotide being located at the 3' end of a gene, for controlling expression 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 CN114908089A (en) 2022-08-16
CN114908089B true 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 (3)

* 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
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
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

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2534430T3 (en) * 2006-08-28 2015-04-22 The University Of Western Australia Modulation method of epidermal growth factor receptor (EGFR) expression that involves miRNA
WO2013007874A1 (en) * 2011-07-12 2013-01-17 Mart Saarma A transgenic animal comprising a deletion or functional deletion of the 3'utr of an endogenous gene.
WO2015101415A1 (en) * 2013-12-30 2015-07-09 Curevac Gmbh Artificial nucleic acid molecules
RU2612497C2 (en) * 2015-05-26 2017-03-09 Общество с ограниченной ответственностью "НекстГен" Optimized nucleotide sequence and pharmaceutical compositions based thereon with sustained vegf transgene expression
WO2017011766A1 (en) * 2015-07-16 2017-01-19 Cornell University Methods of enhancing translation ability of rna molecules treatments, and kits
WO2017177029A1 (en) * 2016-04-06 2017-10-12 Duke University Compositions and methods for enhanced gene expression and viral replication
GB201612214D0 (en) * 2016-07-14 2016-08-31 Univ Oxford Innovation Ltd Method
US10907165B2 (en) * 2016-10-31 2021-02-02 Cornell University Methods of enhancing translation ability and stability of RNA molecules, treatments, and kits
CA3091543A1 (en) * 2018-02-19 2019-08-22 Combined Therapeutics, Inc. Compositions and methods for organ-protective expression and modulation of coding ribonucleic acids

Patent Citations (3)

* 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
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
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
CRNDE Promotes Malignant Progression of Glioma by Attenuating miR-384/PIWIL4/STAT3 Axis;Jian Zheng等;《The American Society of Gene & Cell Therapy》;第24卷(第7期);第1199-1215页 *
Endogenous transcripts control miRNA levels and activity in mammalian cells by target-directed miRNA degradation;Francesco Ghini等;《Nature Communications》;第9卷;第3119页 *

Also Published As

Publication number Publication date
WO2022166771A1 (en) 2022-08-11
CN114908089A (en) 2022-08-16

Similar Documents

Publication Publication Date Title
JP6842495B2 (en) RNA preparation containing purified modified RNA for reprogramming cells
KR102336362B1 (en) Closed-ended linear duplex DNA for non-viral gene delivery
RU2759335C2 (en) Gene editing of deep intron mutations
US20230416787A1 (en) SYSTEMS AND METHODS FOR ONE-SHOT GUIDE RNA (ogRNA) TARGETING OF ENDOGENOUS AND SOURCE DNA
CN111278972B (en) Non-integrated DNA vectors for cytogenetic modification
ES2735531T3 (en) RNA containing modified nucleosides and methods of use thereof
US20230250407A1 (en) Silencing of dux4 by recombinant gene editing complexes
JP2022530457A (en) Genetically engineered AAV
AU2023237106A1 (en) Aav vectors for treatment of dominant retinitis pigmentosa
KR20220066225A (en) Compositions and methods for selective gene regulation
WO2022180213A1 (en) Formulations for aerosol formation and aerosols for the delivery of nucleic acid
JP2021522849A (en) Substitution with two RNA sequences introduced at its 5&#39;end of the messenger RNA cap
US20230383275A1 (en) Sgrna targeting aqp1 rna, and vector and use thereof
CN114908089B (en) Construction method and application of 3&#39; UTR
KR20240004549A (en) Linear DNA with improved resistance to exonucleases
CN109266684B (en) Method for constructing animal model with pathogen infection sensitivity
CN116949041A (en) Artificially designed mRNA UTR nucleotide sequence and its use
CN105968211A (en) Recombinant antiviral protein as well as preparation method and application thereof
WO2022256414A1 (en) Rna recognition complex and uses thereof
TW202411426A (en) Engineered class 2 type v crispr systems
CN114854791A (en) Novel CRISPR-Cas9 system vector and application thereof
KR100631488B1 (en) New synthetic peptides containing PTD domain and membrane-destabilizing motif and their use in gene delivery
NZ747016A (en) Compositions and methods of treatment for lytic and lysogenic viruses
JPH10194997A (en) Novel method of introducing gene and complex to be used therefor

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