US20170211057A1 - Expression Vector Comprising a Polynucleotide Encoding a Modified Glutamine Synthetase and a Method for Preparing a Target Protein Employing the Same - Google Patents

Expression Vector Comprising a Polynucleotide Encoding a Modified Glutamine Synthetase and a Method for Preparing a Target Protein Employing the Same Download PDF

Info

Publication number
US20170211057A1
US20170211057A1 US15/399,432 US201715399432A US2017211057A1 US 20170211057 A1 US20170211057 A1 US 20170211057A1 US 201715399432 A US201715399432 A US 201715399432A US 2017211057 A1 US2017211057 A1 US 2017211057A1
Authority
US
United States
Prior art keywords
vector
tnfr
gene
glutamine synthetase
expression
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.)
Abandoned
Application number
US15/399,432
Inventor
Hyun Sook Jang
Dong Heon Lee
Sun Kyu KIM
Yong HO Ahn
Sang Kyung Park
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.)
Ares Trading SA
Original Assignee
Ares Trading SA
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 Ares Trading SA filed Critical Ares Trading SA
Priority to US15/399,432 priority Critical patent/US20170211057A1/en
Assigned to HANWHA CHEMICAL CORPORATION reassignment HANWHA CHEMICAL CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: AHN, YONG HO, JANG, HYUN SOOK, KIM, SUN KYU, LEE, DONG HEON, PARK, SANG KYUNG
Assigned to ARES TRADING S.A. reassignment ARES TRADING S.A. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HANWHA CHEMICAL CORPORATION
Publication of US20170211057A1 publication Critical patent/US20170211057A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/93Ligases (6)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • 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
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • 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
    • C12N15/52Genes encoding for enzymes or proenzymes
    • 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
    • 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
    • C12YENZYMES
    • C12Y603/00Ligases forming carbon-nitrogen bonds (6.3)
    • C12Y603/01Acid-ammonia (or amine)ligases (amide synthases)(6.3.1)
    • C12Y603/01002Glutamate-ammonia ligase (6.3.1.2)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • 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
    • C12N2840/00Vectors comprising a special translation-regulating system
    • C12N2840/20Vectors comprising a special translation-regulating system translation of more than one cistron
    • C12N2840/203Vectors comprising a special translation-regulating system translation of more than one cistron having an IRES

Definitions

  • the present invention relates to a vector comprising a polynucleotide encoding a modified glutamine synthetase (GS), and a method for preparing a target protein employing the same. More particularly, the present invention relates to a modified GS having an increased sensitivity to a glutamine synthetase (GS) inhibitor, a polynucleotide encoding the modified GS, a vector comprising the polynucleotide, a transformant comprising the vector, and a method for preparing a target protein using the transformant.
  • GS glutamine synthetase
  • Recombinant proteins can be expressed in different types of host cells including prokaryotic and eukaryotic cells.
  • glycoproteins such as antibodies and Fc fusion proteins, consist of a polypeptide linked to a carbohydrate moiety which influences the safety and efficacy thereof, and thus they are usually expressed in animal cells that are capable of glycosylation during post-translational modification.
  • the difference in their sugar chains with those of human (native) glycoproteins is associated with immunogenicity of the protein drugs.
  • animal cells such as hybridoma, mouse myeloma, and CHO cells have been commonly used in the expression and production of recombinant protein drugs.
  • the protein expression in these animal cells is appropriate for producing proteins similar to human proteins, but there are disadvantages of a significantly low expression yield and difficulty in scale-up of the production.
  • therapeutic antibodies need to be produced in kilogram quantities, and thus animal cell culturing is not suitable for a large-scale production of the therapeutic antibodies. Therefore, for a high level expression of a target gene in the host cells, the target gene needs to be integrated into a transcriptionally active region of the genome when the target DNA is randomly introduced to the animal cell. The introduced foreign gene replicates along with the genome of the host cell.
  • a homologous recombination technique for integration of the gene into transcriptionally active regions is not generalized for common use yet.
  • a DHFR system (Takeshi omasa, gene amplification and its application in cell and tissue engineering, J. Bios. and Bioe (2002), Vol. 94, No. 6, 600-605) is a common gene amplification system, and this system increases the protein expression level by co-amplification of a target gene and DHFR using methotrexate (MTX) which is an inhibitor of dihydrofolate reductase (DHFR).
  • MTX methotrexate
  • DHFR is an enzyme involved in nucleotide biosynthesis, and thus inhibition of DHFR activity can effectively interrupt DNA synthesis which is essential for cell maintenance, thereby leading to cell death. Therefore, only those clones having exogenous DHFR gene inserted in their genome can survive under this condition. Furthermore, when the concentration of MTX being added is increased and a strong promoter is used, DHFR gene can be amplified to hundreds to thousands of copies. That is, the more MTX, a DHFR inhibitor, is added to the cell culture, in order for them to survive they increase the expression of DHFR along with the introduced target gene. Consequently, several copies of DNA will be incorporated, leading to the generation of various molecular variants.
  • a DHFR system using CHO cell line has been reported and commercialized as an expression system for various protein drugs, verifying its safety and efficacy in use.
  • the DHFR system has a disadvantage in that it requires several months to isolate a single cell line that shows the expression level higher than the normal level.
  • the target gene cannot be amplified anymore.
  • the CHO DUKX cells used for DHFR system revertants may appear easily. As a result, there has been a high demand for the development of a high-level gene expression system for protein production other than the DHFR system.
  • GS system is a high-level gene expression system that was first developed by Celltech (U.S. Pat. No. 5,122,464), and it overcame the limitations of the DHFR-based gene expression system, that is, low time-efficiency for isolating the single cell line of interest and low productivity of target protein.
  • the GS system utilizes glutamine synthetase (GS) which is an enzyme involved in the sole synthetic pathway for producing glutamine from glutamate and ammonia, based on the fact that animal cells cannot grow properly in the glutamine-deficient condition.
  • GS glutamine synthetase
  • the GS system has advantage in that it requires less number of DNA copies per cell compared to the DHFR system and allows for the selection of single cell line having high expression rate at the early stage of screening.
  • NS0 cell line and CHO cell line are the most commonly used cell lines for GS system. Between two, NS0 cell line which is a mouse myeloma cell line cannot express sufficient amount of GS, and thus in the glutamine-deficient condition, those cells where the target gene is inserted into their genome can be easily selected. Unlike the NSO cell line, a CHO cell line can express sufficient amount of GS that they can survive even in the glutamine-deficient medium.
  • the CHO cells are treated with a high concentration of GS-specific inhibitor such as methionine sulphoximine (MSX), the cells cannot survive only with the endogenous GS activity, and thus only those cells introduced with the vector comprising the GS gene and the gene for a target protein can survive.
  • MSX methionine sulphoximine
  • the cells inserted with the gene for target protein can be isolated, and the target protein can be produced at high yield.
  • more GS-specific inhibitor is added to the cells, in order for them to survive, they will amplify exogenous GS gene as well as the target gene which is introduced together with the GS gene, thereby increasing the amount of target protein in the cell.
  • this GS system has a limitation in production amount when the cells transfected with the vector comprising the genes for target protein and GS are treated with the GS-specific inhibitor for producing the target protein. Also when the cells were cultured for a long time, the production amount of the target protein was reduced. Due to these limitations, there has been a high demand for the development of a modified GS protein that responses more sensitively to the GS inhibitor and thus can amplify the target gene introduced with GS to the greater level.
  • the present inventors In an effort to develop a modified GS protein that has a significantly higher sensitivity to GS inhibitor compared to the wildtype GS protein, the present inventors have developed a modified GS with a significantly higher sensitivity to GS inhibitor having one amino acid substituted compared to the wildtype GS protein. Then, the present inventors confirmed the production of the target protein in the presence of GS inhibitor after transfecting the animal cells with the vector comprising the gene encoding the modified GS and target protein, and further confirmed that the modified GS demonstrates high sensitivity towards GS inhibitor, showing the remarkably higher level of target protein production compared to the vector system comprising the wildtype GS protein and also no reduction in the production level of target protein even after long period of culturing, thereby completing the present invention.
  • An object of the present invention is to provide a modified glutamine synthetase (GS), wherein glycine (Gly, G) at position 299 of a glutamine synthetase having an amino acid sequence shown in SEQ ID NO. 4 is substituted with arginine (Arg, R).
  • GS modified glutamine synthetase
  • Another object of the present invention is to provide a polynucleotide encoding the modified glutamine synthetase.
  • Still another object of the present invention is to provide a vector for the expression of a target protein, comprising the polynucleotide and the gene encoding the target protein.
  • Still another object of the present invention is to provide a transformant comprising the vector.
  • Still another object of the present invention is to provide a method for the preparation of a target protein comprising culturing of the transformant and to provide the target protein prepared by said method.
  • the expression vector comprising a modified GS gene of the present invention allows for the selection of the host cells introduced with the expression vector comprising the modified GS gene even under the condition where CHO cells are used as a host cell and the cells are treated with a GS inhibitor. Therefore, the present expression vector can be widely used for the efficient production of a target protein.
  • FIG. 1 is a schematic diagram showing the cloning method for preparing pcDNA3.1-Kozak-TNFR-Fc-IRES-GS or pcDNA3.1-kozak-TNFR-Fc-IRES-GS PM of the present invention
  • FIG. 2 is a cleavage map of the recombinant expression vector, pcDNA3.1-kozak-TNFR-Fc-IRES-GS or pcDNA3.1-kozak-TNFR-Fc-IRES-GS PM, comprising a gene encoding a TNFR-Fc fusion protein which is a representative target protein of the present invention;
  • FIG. 3 is a schematic diagram showing the cloning method for preparing the recombinant expression vector, pcDNA3.1-kozak-TNFR-Fc-SV40-GS or pcDNA3.1-kozak-TNFR-Fc-SV40-GS PM, comprising a gene encoding a TNFR-Fc fusion protein which is a representative target protein of the present invention;
  • FIG. 4 is a cleavage map of the recombinant expression vector, pcDNA3.1-kozak-TNFR-Fc-SV40-GS or pcDNA3.1-kozak-TNFR-Fc-SV40-GS PM, comprising a gene encoding a TNFR-Fc fusion protein which is a representative target protein of the present invention;
  • FIG. 5 is a graph showing the expression level of TNFR-Fc in CHO K-1 cells transfected with one of the three types of TNFR-Fc expression vectors (IRES-GS PM, IRES-GS, or SV40-GS);
  • FIG. 6 is a graph showing the changes in expression level of TNFR-Fc protein with MSX treatment over time after transfection of CHO K-1 cells with one of the three types of TNFR-Fc expression vectors (IRES-GS PM, IRES-GS, or SV40-GS);
  • FIG. 7 is a graph demonstrating the changes in TNFR-Fc expression level confirmed by the secondary transient transfection, showing a high expression level in the SV40-GS vector-transfected group during the early phase of culturing, but after longer period of culturing with the MSX treatment the highest expression level was observed in IRES-GS PM vector-transfected group;
  • FIG. 8 is a graph showing the TNFR-Fc expression level in two groups of stable CHO-S cell line each transfected with either IRES-GS PM vector or SV40-GS PM vector.
  • the present invention provides a modified glutamine synthetase (GS), wherein glycine (Gly, G) at position 299 of a glutamine synthetase having an amino acid sequence shown in SEQ ID NO. 4 is substituted with arginine (Arg, R).
  • GS modified glutamine synthetase
  • GS glutamine synthetase
  • GS glutamine synthetase
  • GS glutamine synthetase
  • divalent metal ions are required, and its enzymatic activity is inhibited by the presence of glycine, alanine, tryptophan, histidine, glucosamine-6-phosphate, cytidine triphosphate, etc.
  • the glutamine synthetase refers to an enzyme that can be used to select the cells transfected with the vector comprising the gene encoding a target protein or to enhance the expression of the target protein by treatment with GS inhibitor, but is not limited thereto.
  • the information on the glutamine synthetase can be obtained from the common database such as NCBI GenBank.
  • the information on glutamine synthetase derived from hamster can be found from GenBank with the accession number X03495.1, but is not limited thereto.
  • GenBank accession number X03495.1
  • SEQ ID NO. 3 The nucleotide sequence of the representative wild type glutamine synthetase is shown as SEQ ID NO. 3 and its amino acid sequence is shown as SEQ ID NO. 4.
  • modified glutamine synthetase refers to an enzyme wherein glycine (Gly, G) at position 299 of a glutamine synthetase modified glutamine synthetase (GS), wherein glycine (Gly, G) at position 299 of a glutamine synthetase having an amino acid sequence shown in SEQ ID NO. 4 is substituted with arginine (Arg, R).
  • the modified glutamine synthetase may be used as a selection marker by transfection of the cells that can or cannot produce GS endogenously with the target protein expression vector comprising the gene encoding the modified glutamine synthetase, or preferably refers to the protein that can enhance the target protein expression by treatment with GS inhibitor through polycistronic translation with the gene encoding the target protein in the form of ‘polynucleotide encoding the promoter-target protein encoding gene-IRES-modified glutamine synthetase’ or ‘polynucleotide encoding the promoter-modified glutamine synthetase-IRES-target protein encoding gene’, but is not limited thereto.
  • the inventor of the present invention has identified for the first time that if guanosine (G) in the codon of glycine at position 299 of the glutamine synthetase with SEQ ID NO. 4 is substituted with cytidine (C), the sensitivity of glutamine synthetase towards GS inhibitor is remarkably increased, as compared to the wild type GS.
  • the modified GS of the present invention has a significantly higher sensitivity towards GS inhibitor, it can be effectively used in the target protein expression system.
  • the modified glutamine synthetase comprises the amino acid sequence wherein glycine (Gly, G) at position 299 of a glutamine synthetase consisting of an amino acid sequence of SEQ ID NO. 4 is substituted with arginine (Arg, R), and as long as the enzyme demonstrates increased sensitivity towards GS inhibitor compared to the wildtype GS, the modified glutamine synthetase of the present invention may comprise the amino acid sequence having a sequence homology of 70% or higher, preferably 80% or higher, more preferably 90% or higher, even more preferably 95% or higher, even much more preferably 98% or higher, and most preferably 99% or higher to the above-described amino acid sequence of modified GS.
  • the term ‘GS inhibitor’ refers to an external factor that is capable of inhibiting the GS activity.
  • examples of such inhibitor include glycine, alanine, tryptophan, histidine, glucosamine-6-phosphate, cytidine triphosphate, and methionine sulphoximine (MSX), but is not limited thereto.
  • MSX methionine sulphoximine
  • the GS inhibitor is preferably MSX, but is not limited thereto.
  • sensitivity generally refers to the feature of responding to external stimuli.
  • sensitivity refers to the feature of enhancing or reducing enzymatic activity in response to external factors that regulate enzymatic activity.
  • the sensitivity refers to the suppression of the GS activity in response to the GS inhibitor, but is not limited thereto.
  • the gene encoding the modified glutamine synthetase was to be obtained through cloning the glutamine synthetase from RNA of hamster cells via PCR.
  • IRES-GS PM was prepared by linking the polynucleotide encoding the modified glutamine synthetase with IRES, which was further connected to the gene encoding the target protein in order to prepare the vector comprising the expression cassette wherein the target protein encoding gene-IRES-GS PM are operably connected together (Example 4). Furthermore, when the target protein was expressed using the vector comprising the modified glutamine synthetase of the present invention, the production level of the target protein was significantly higher than when the wildtype glutamine synthetase was used (Examples 6 and 7).
  • the present invention provides a polynucleotide encoding the modified glutamine synthetase.
  • the modified glutamine synthetase of the present invention is characterized by having a substitution of the amino acid at position 299 of wild type GS from glycine to arginine.
  • DNA codon encoding the amino acid at position 299 of the modified glutamine synthetase may be selected from the group consisting of CGT, CGC, CGA, CGG, AGA and AGG, but is not limited thereto.
  • polynucleotide encoding the modified glutamine synthetase of the present invention may be preferably for enhancing the expression of target protein.
  • the polynucleotide encoding the modified glutamine synthetase of the present invention may be present in the vector to be translated by polycistronic translation with the gene encoding target protein. After transfection of the host cells with the above vector, when the cells are treated with GS inhibitor, the activity of the modified glutamine synthetase expressed from the introduced vector is inhibited, thereby reducing the synthesis of glutamine. However, in order for the cells to survive, glutamine is essential, and thus under the suppression by GS inhibitor those cells tend to synthesize more of the glutamine synthetase.
  • the target protein-encoding gene introduced along with the polynucleotide encoding the modified GS gets amplified as well, and through this mechanism the expression of the target protein can be increased.
  • the present invention provides a vector for expression of a target protein, comprising the polynucleotide encoding the modified GS and a gene encoding the target protein.
  • the polynucleotide encoding the modified GS is the same as described above.
  • target protein refers to the protein of interest to be produced in the host cells.
  • it refers to the protein whose expression is enhanced by the modified glutamine synthetase, but is not limited thereto.
  • the type of target protein is not specifically limited as long as it can be expressed by the vector of the present invention.
  • tumor necrosis factor receptor (TNFR)-Fc fusion protein was used as a representative target protein that can be expressed by the modified glutamine synthetase of the present invention.
  • TNFR-Fc fusion protein refers to the product prepared by connecting the entire or a part of TNFR protein with immunoglobulin Fc region by enzymatic action, or the product prepared by expressing two polypeptides into a single polypeptide by genomic manipulation.
  • the TNFR protein and the immunoglobulin Fc region may be directly linked with each other, or linked via a peptide linker, but is not limited thereto.
  • the polynucleotide encoding the TNFR-Fc fusion protein may be a polynucleotide of SEQ ID NO. 5, but is not limited thereto.
  • the term ‘expression vector’ refers to a DNA construct comprising an essential control component which is operably linked to an insert gene so that the insert gene is only expressed when introduced into the host cell.
  • the expression vector may be prepared and purified by a standard recombinant DNA technology.
  • the type of the expression vector is not particularly limited, as long as it expresses and produces a target gene in a variety of host cells of prokaryotic and eukaryotic cells.
  • the expression vector is a vector capable of producing a large amount of a foreign protein in a similar form to the native protein while it retains a strong promoter activity and a strong expression ability.
  • the expression vector is preferably a vector comprising at least a promoter, a start codon, a gene encoding a target protein, a stop codon, and a terminator.
  • it may comprise a DNA encoding a signal peptide, an enhancer sequence, untranslated regions at the 5′ and 3′ ends of a target gene, a selectable marker region or a replicable unit, etc., if desired.
  • the type of the expression vector may be a mono-cistronic vector including a polynucleotide encoding one recombinant protein, a bi-cistronic vector including a polynucleotide encoding two recombinant proteins, a poly-cistronic vector including a polynucleotide encoding three recombinant proteins or more.
  • the expression vector is preferably a mono-cistronic vector including a SV40 promoter or a bi-cistronic vector including an IRES sequence, more preferably, an expression vector including a promoter, a gene encoding a target protein, IRES, and a modified GS gene in this order or an expression vector including a promoter, a modified GS gene, IRES, and a gene encoding a target protein, but is not limited thereto.
  • a polynucleotide encoding GS PM was acquired from CHO DG44, and a pGEMT-GS PM vector was obtained by connecting each of the acquired polynucleotides to a pGEMT vector. Subsequently, the pGEMT-GS PM vector was inserted into a cleaved TOPO-IRES-DHFR vector to obtain a pCR2.1-TOPO-IRES-GS PM vector, and an IRES-GS PM fragment was obtained from the vector.
  • the obtained IRES-GS PM fragment was inserted into a cleaved pcDNA-Kozak-TNFR-Fc-IRES-DHFR vector so as to construct a kozak-TNFR-Fc-IRES-GS PM vector ('IRES-GS PM vector') ( FIG. 1 ).
  • the polynucleotide encoding GS PM was inserted into the cleaved pcDNA3.1-TNFR-Fc-SV40-DHFR vector so as to obtain a pcDNA3.1-TNFR-Fc-SV40-GS PM vector. Thereafter, the pcDNA3.1-TNFR-Fc-SV40-GS PM vector was cleaved, and a TNFR-Fc fragment from the pcDNA3.1-Kozak-TNFR-Fc-IRES-DHFR vector was inserted into the cleaved region so as to construct a pcDNA-Kozak-TNFR-Fc-SV40-GS PM vector ('SV40-GS PM vector') ( FIG. 3 ).
  • the present invention provides a transformant comprising the vector.
  • the term ‘transformant’ refers to the cell transformed with the expression vector so as to express the polynucleotide encoding the recombinant protein included in the expression vector. It may be recombinant mammalian cells, rodent cells, preferably animal cells or animal-derived cells, and most preferably NS0 or CHO cells, but is not limited thereto. With respect to the objects of the present invention, the transformant is preferably a transformant prepared by introducing the expression vector into a NS0 or CHO cell line, but is not limited thereto.
  • the present invention provides a method for preparing a target protein, comprising culturing the transformant.
  • the transformant and target protein are the same as described above.
  • the above method comprises (a) culturing the transformant; and (b) adding a GS inhibitor to a culture medium.
  • the method further comprises (c) isolating the target protein from the culture medium.
  • the expression system using the modified GS protein of the present invention which has an enhanced sensitivity to GS inhibitor can lead to the increased expression of a target protein as compared to the expression system using the wildtype GS.
  • the present invention provides a target protein prepared by the above described method.
  • the method and the target protein are the same as described above.
  • a TNFR-Fc fusion protein was used as a representative target protein.
  • the fusion protein-encoding gene (SEQ ID NO. 5) was synthesized by GeneArt Inc., so as to meet the following criteria: (1) it must include a TNFR signal sequence (2) it must express the TNFR amino acids at position 1 to 235 (3) it must be codon-optimized for CHO cells in order to be transfected into CHO cells (4) it must have a NheI restriction site at 5′-end and a NotI restriction site at 3′-end, considering insertion into a pcDNA3.1 vector of Invitrogen.
  • the nucleotide sequence of the synthesized fusion protein-encoding gene was finally analyzed using the VectorNTl program.
  • DHFR hamster dihydrofolate reductase
  • a pSVA3 vector (ATCC 77273) having a mutant type of hamster DHFR gene was purchased, and then a wild type of DHFR gene was obtained by performing point mutation using the DHFR gene as a template.
  • an IRES sequence was obtained by PCR from a Clontech vector (Cat. #6029-1, PT3267-5) having the corresponding DNA sequence.
  • DHFR gene and internal ribosome entry site (IRES) sequence were cloned into a pCR2.1 vector so as to construct a pCR2.1-IRES-DHFR expression vector.
  • Each of the TNFR-Fc-inserted pcDNA3.1-TNFR-Fc vector obtained in Example 1 and the obtained pCR2.1-IRES-DHFR vector was digested with restriction enzymes, SalI and XbaI, and ligated so as to obtain a TNFR-Fc-inserted pcDNA3.1-TNFR-Fc-IRES-DHFR expression vector.
  • the kanamycin-resistant gene was obtained from a pAC-GFP vector (#632483) of Clontech, so as to introduce a Kan/Neo gene.
  • the vector is a vector having a Kozak sequence at a transcription initiation sequence of the TNFR-Fc gene and the Kan/Neo gene as an antibiotic selection marker, and it was used as a basic frame for cloning 4 different expression vector systems in order to compare the expression levels of the recombinant protein using CHO cells.
  • a hamster cell line CHO DG44 (Invitrogen, 12609-012) was cultured, and then total RNA was isolated using a TRIZOL reagent (Invitrogen). After that, RT-PCR was performed using the obtained total RNA so as to obtain cDNA.
  • PCR 25 cycles of denaturation at 94° C. for 5 minutes; denaturation at 94° C. for 30 seconds, annealing at 50° C. for 30 seconds, elongation at 72° C. for 90 seconds; and elongation at 72° C. for 7 minutes
  • PCR 25 cycles of denaturation at 94° C. for 5 minutes; denaturation at 94° C. for 30 seconds, annealing at 50° C. for 30 seconds, elongation at 72° C. for 90 seconds; and elongation at 72° C. for 7 minutes
  • GS SalI-F primer and GS XbaI-R primer for acquisition of the following GS PM gene, so as to obtain a
  • GS SalI-F (Forward primer): (SEQ ID NO. 6) 5′-gtcgacatggccacctcagcaagttccc-3′ GS XbaI-R (Reverse primer): (SEQ ID NO. 7) 5′-tctagattagtttttgtattggaaaggg-3′
  • the obtained PCR product was electrophoresed on a 0.8% agarose gel, and then the corresponding band was cut, followed by clean-up using a Quiagen Cleaning kit (#28204). Then, the resultant was inserted into a gene cloning vector, pGEMT vector (Promega, USA).
  • the PCR product-inserted pGEMT vector was introduced into a TOP10 cell so as to obtain a total of 10 colonies. After that, a nucleotide sequence and an amino acid sequence encoded by the nucleotide sequence of each colony were analyzed.
  • a modified GS gene showing a difference in one amino acid at position 299 as compared to the amino acid sequence of the wild type hamster GS (NCBI GenBank: X03495.1), was acquired.
  • this difference is attributed to the alteration of the 895 th nucleotide of the wildtype GS gene (SEQ ID NO. 3) from G (Guanosine) to C (Cytidine). That is, the modified GS has the characteristic of altered amino acids at position 299 from glycine (Gly, G) to arginine (Arg, R).
  • the acquired modified GS was named ‘GS PM’.
  • the 895 th C in the nucleotide sequence of GS-PM was substituted with G. Specifically, cloning was performed by point mutation for replacement of one amino acid. More specifically, in order to obtain the wild type GS having one amino acid different from those of GS PM of the present invention, PCR (30 cycles of denaturation at 94° C. for 5 minutes; denaturation at 94° C. for 30 seconds, annealing at 54° C. for 30 seconds, elongation at 72° C. for 30 seconds; and elongation at 72° C.
  • KpnI F-primer (Forward primer): (SEQ ID NO. 8) 5′-caccggtaccacattcgagcctacgatcccaaggggggcctggacaa tgcccgtggtctg-3′
  • XbaI R-primer (Reverse primer): (SEQ ID NO. 9) 5′-tctagattagtttttgtattggaaggg-3′
  • the nucleotide sequence of the PCR product was analyzed. As a result, a point mutation from CGT to GGT was observed.
  • the corresponding PCR product was digested with KpnI and XbaI, and then ligated with a pGEMT-GS PM vector treated with KpnI and XbaI, so as to obtain a pGEMT-GS vector.
  • the GS PM gene included in the pGEMT-GS PM vector obtained in Example 3 was cloned to have SalI and XbaI restriction sites at its N- and C-terminals, respectively. Therefore, in order to obtain IRES-GS PM, a fragment obtained by treating pGEMT-GS PM with SalI and XbaI restriction enzymes was inserted into a TOPO-IRES-DHFR vector that was previously digested with SalI and XbaI restriction enzymes, so as to obtain a pCR2.1-TOPO-IRES-GS PM gene.
  • FIG. 1 is a schematic diagram showing the cloning method of pcDNA3.1-kozak-TNFR-Fc-IRES-GS PM of the present invention.
  • kozak-TNFR-Fc-IRES-GS PM vector and the TOPO-GS vector were used to construct a Kozak-TNFR-Fc-IRES-GS vector (“IRES-GS vector”) ( FIG. 2 ).
  • FIG. 2 is a cleavage map showing pcDNA3.1-kozak-TNFR-Fc-IRES-GS or pcDNA3.1-kozak-TNFR-Fc-IRES-GS PM, which is a recombinant expression vector including the TNFR-Fc-encoding gene of the present invention.
  • the cloned pcDNA3.1-TNFR-Fc-SV40-DHFR vector was used in order to prepare a SV40 promoter-GS system identical to Lonza's GS system among GS systems. Since the pcDNA3.1-TNFR-Fc-SV40-DHFR has no restriction sites suitable for GS gene insertion, new restriction sites were first inserted into both ends of the GS gene.
  • a pair of primers (GS-BsaBI-F primer and GS-BstBI-R primer) containing BsaBI at the N-terminal of GS gene and BstBI at C-terminal of GS gene were synthesized to perform PCR.
  • the BsaBI and BstBI sites were inserted into both ends of the GS gene, and DHFR was removed from pcDNA3.1-TNFR-FC-SV40-DHFR by treatment with BsaBI and BstBI.
  • the GS gene digested with BsaBI/BstBI was inserted thereto so as to construct a pcDNA3.1-TNFR-Fc-SV40-GS PM vector.
  • GS-BsaBI-F Primer (forward primer): (SEQ ID NO. 10) 5′-gatgaggatcatggccacctcagcaag-3′
  • GS-BstBI-R reverse primer: (SEQ ID NO. 11) 5′-ttcgaattagtttttgtattggaaggg-3′
  • the pcDNA3.1-TNFR-Fc-SV40-GS PM vector has no Kozak sequence prior to the TNFR-Fc gene, unlike the pcDNA3.1-TNFR-Fc-IRES-GS PM vector. Therefore, a second cloning step of inserting the Kozak sequence into the vector was performed. For the Kozak sequence, a Kozak sequence of the previously prepared pcDNA3.1-Kozak-TNFR-Fc-IRES-DHFR vector was used. Instead of TNFR-Fc of the pcDNA3.1-TNFR-Fc-SV40-GS PM vector, the Kozak-TNFR-Fc was inserted to prepare pcDNA3.1-Kozak-TNFR-FC-SV40-GS PM.
  • the restriction enzymes to be used for the cloning may include NdeI and NheI at N-terminal of TNFR-Fc and BstXI, SgrAI, and NotI at C-terminal of TNFR-Fc. Available restriction enzymes were selected from them, so as to construct a pcDNA-Kozak-TNFR-Fc-SV 40-GS PM vector (“SV40-GS PM vector”) ( FIG. 3 ).
  • SV40-GS PM vector pcDNA-Kozak-TNFR-Fc-SV 40-GS PM vector
  • FIG. 3 is a schematic diagram showing the cloning method of pcDNA3.1-kozak-TNFR-Fc-SV40-GS or pcDNA3.1-kozak-TNFR-Fc-SV40-GS PM, which is a recombinant expression vector including the TNFR-Fc-encoding gene of the present invention.
  • the constructed SV40-GS PM vector includes an antibiotic resistance gene, ampicillin resistance gene for cell line selection, and thus a third cloning step was performed to replace the gene with a kanamycin resistance gene.
  • the antibiotic resistance gene was replaced by an antibiotic with a low frequency of use on grounds of safety, because ampicillin is one of the antibiotics frequently used by patients.
  • the kanamycin resistance gene was obtained from the pAC-GFP vector (#632483) of Clontech, and introduced into a SV40-GS PM vector. Furthermore, the SV40-GS PM vector was used to construct a pcDNA-Kozak-TNFR-Fc-SV 40-GS vector (‘SV40-GS vector’) ( FIG. 4 ).
  • FIG. 4 a pcDNA-Kozak-TNFR-Fc-SV 40-GS vector
  • FIG. 4 is a cleavage map showing pcDNA3.1-kozak-TNFR-Fc-SV40-GS or pcDNA3.1-kozak-TNFR-Fc-SV40-GS PM, which is a recombinant expression vector including the TNFR-Fc-encoding gene of the present invention.
  • IRES-GS IRES-GS PM
  • SV40-GS SV40-GS PM
  • SV40-GS PM 4 different expression vectors of IRES-GS, IRES-GS PM, SV40-GS, and SV40-GS PM were constructed as a protein expression vector system for expressing a target protein in mammalian cells.
  • each of the expression vectors constructed in Example 4 was introduced into CHO K-1 cells so as to prepare each transformant.
  • CHO K-1 cells were cultured in a DMEM/F12 medium supplemented with 10% FBS, and 3 to 4 ⁇ 10 5 cells/well were inoculated into a 6 well-plate, followed by cultivation overnight. When the cells reached 80 to 90% confluence, each of the expression vectors was introduced thereto.
  • the culture medium in the 6-well plate was replaced with 2 ml of Opti-MEM I media, and 500 ⁇ l of the final mixture was added to each well of the 6-well plate, followed by cultivation at 37° C. for 4 to 6 hours.
  • the medium was replaced with the original culture medium (DMEM/F12 medium supplemented with 10% FBS), followed by further cultivation overnight.
  • the culture medium in the 6-well plate was replaced with a GS selection medium, and subculture was performed according to the growth rate of the cells introduced with each of the expression vectors.
  • the GS selection medium was glutamine-free DMEM or glutamine-free IMDM containing 10% FBS, 1 ⁇ GS supplement, and the GS inhibitor, MSX.
  • the 1 ⁇ GS supplement was prepared by including adenosine (500 ⁇ , 15 mM), cytidine (1000 ⁇ , 30 mM), uridine (1000 ⁇ , 30 mM), guanosine (1000 ⁇ , 3 mM), thymidine (1000 ⁇ , 10 mM), asparagine (1000 ⁇ , 500 mM) and glutamic acid (1000 ⁇ , 500 mM) or a commercially available 50 ⁇ GS supplement (SAFC) was used after dilution.
  • SAFC commercially available 50 ⁇ GS supplement
  • Example 5 The culture broth of each transformant subcultured in Example 5 was applied to ELISA, in order to measure the expression level of each protein expressed from the transformant.
  • a 96-well plate was coated with anti-human IgG Fc antibody (Pierce, 31125), and blocked with 1% BSA.
  • anti-human IgG Fc antibody Pieris, 31125
  • BSA 1% BSA
  • the culture broth of each subcultured transformant was added to each well, and reacted.
  • a biotin-conjugated anti-human TNFR antibody R&D system
  • HRP-conjugated streptavidin was added to each well, and reacted.
  • each well was treated with TMB for color development so as to examine the expression level of each protein.
  • FIG. 5 is a graph showing the expression levels of TNFR-Fc in CHO K-1 cells transfected with three types of TNFR-Fc gene expression vector (IRES-GS PM, IRES-GS, SV40-GS).
  • CHO K-1 cells showed no changes in the expression level of TNFR-FC protein by three types of GS vectors or in the cell survival rate. Rather, three types of the GS systems were more excellent in terms of expression level than the IRES-DHFR system. Moreover, even at 5 days after transfection of the expression vectors, the cells transfected with the three types of expression vectors maintained their growth and continuously expressed the TNFR-Fc protein.
  • FIG. 6 is a graph showing the time-dependent expression level of TNFR-Fc protein according to initial MSX treatment of CHO K-1 cells, which were transfected with the three types of TNFR-Fc gene expression vectors (IRES-GS PM, IRES-GS, SV40-GS).
  • the cell line transfected with the IRES-GS PM vector of the present invention exhibited higher levels in terms of the total TNFR-Fc protein expression level and the expression level per an equal number of cells, compared to the cell lines transfected with other vectors.
  • the cells transfected with the SV40-GS vector exhibited the highest level.
  • MSX MSX
  • the cells transfected with the IRES-GS PM vector of the present invention exhibited the highest level.
  • FIG. 7 is a graph showing the TNFR-Fc expression level after a secondary transient transfection (upper graph) and the time-dependent TNFR-Fc expression level (lower graph).
  • the TNFR-Fc expression levels were mostly 6000-9000 ng/10 6 cells when the cells were cultured without MSX for 6 days after transfection, and the TNFR-Fc expression levels were relatively increased in the cells transfected with the wild type, TRES-GS and SV40-GS vectors.
  • the TNFR-Fc expression levels were 1000 ng/10 6 cells or lower, immediately after the addition of 25 ⁇ M MSX, but the expression levels increased according to time. After the addition of 200 ⁇ M MSX, only the cells transfected with the IRES-GS PM vector of the present invention showed an increase in the TNFR-Fc expression level.
  • modified GS protein of the present invention shows an enhanced sensitivity to GS inhibitors as compared to that of the wildtype GS protein, and the vector comprising the polynucleotide encoding the modified GS protein and the gene encoding the target protein can be effective to produce the target protein.
  • Example 6 showed that the higher protein expression level per an equal number of cells was observed in the cell lines transfected with the GS-containing expression vectors (IRES-GS PM and SV40-GS PM expression vectors) among the four types of GS expression vectors. Therefore, the expression levels in the stable CHO-S cell line were compared between the IRES-GS PM and SV40-GS PM expression vectors. In order to produce a large amount of recombinant protein, the CHO-S cell adapted for growth in suspension was used for the stable cell line establishment, instead of CHO K-1.
  • FIG. 8 is a graph showing the comparison in the stable cell line between the IRES-GS PM vector and the SV40-GS PM vector.
  • PCD indicates pg/cell/day, and it was calculated from the following Equation.
  • PCD expression amount (ng/mL)/(( A ⁇ B )*Culture day/ LN ( A/B ))/1000
  • the cells transfected with the IRES-GS PM expression vector showed a higher expression level of TNFR-FC protein than those transfected with the SV40-GS PM expression vector, irrespective of MSX concentration.
  • the maximum PCD at 250 ⁇ M MSX was ⁇ 8 PCD, which is higher than that of a single cell line limiting-diluted using the known IRES-DHFR system.

Abstract

The present invention relates to a vector comprising a polynucleotide encoding a modified glutamine synthetase (GS), and a method for preparing a target protein employing the same. More particulary, the present invention relates to a modified GS having an increased sensitivity to a glutamine synthetase (GS) inhibitor, a polynucleotide encoding the modified GS, a vector comprising the polynucleotide, a transformat comprising the vector, and a method for preparing a target protein using the transformat.

Description

    TECHNICAL FIELD
  • The present invention relates to a vector comprising a polynucleotide encoding a modified glutamine synthetase (GS), and a method for preparing a target protein employing the same. More particularly, the present invention relates to a modified GS having an increased sensitivity to a glutamine synthetase (GS) inhibitor, a polynucleotide encoding the modified GS, a vector comprising the polynucleotide, a transformant comprising the vector, and a method for preparing a target protein using the transformant.
  • BACKGROUND ART
  • Recombinant proteins can be expressed in different types of host cells including prokaryotic and eukaryotic cells. However, glycoproteins, such as antibodies and Fc fusion proteins, consist of a polypeptide linked to a carbohydrate moiety which influences the safety and efficacy thereof, and thus they are usually expressed in animal cells that are capable of glycosylation during post-translational modification. In the production of recombinant protein drugs, the difference in their sugar chains with those of human (native) glycoproteins is associated with immunogenicity of the protein drugs. Thus, it is important to produce glycoproteins having sugar chains identical or similar to those of human glycoproteins.
  • Until recently, animal cells such as hybridoma, mouse myeloma, and CHO cells have been commonly used in the expression and production of recombinant protein drugs. The protein expression in these animal cells is appropriate for producing proteins similar to human proteins, but there are disadvantages of a significantly low expression yield and difficulty in scale-up of the production. In particular, therapeutic antibodies need to be produced in kilogram quantities, and thus animal cell culturing is not suitable for a large-scale production of the therapeutic antibodies. Therefore, for a high level expression of a target gene in the host cells, the target gene needs to be integrated into a transcriptionally active region of the genome when the target DNA is randomly introduced to the animal cell. The introduced foreign gene replicates along with the genome of the host cell. However a homologous recombination technique for integration of the gene into transcriptionally active regions is not generalized for common use yet.
  • Another method for increasing the expression rate of randomly integrated DNA is by amplifying the integrated gene, and this method needs the step of cloning the gene into the vector engineered with gene amplification system. A DHFR system (Takeshi omasa, gene amplification and its application in cell and tissue engineering, J. Bios. and Bioe (2002), Vol. 94, No. 6, 600-605) is a common gene amplification system, and this system increases the protein expression level by co-amplification of a target gene and DHFR using methotrexate (MTX) which is an inhibitor of dihydrofolate reductase (DHFR). DHFR is an enzyme involved in nucleotide biosynthesis, and thus inhibition of DHFR activity can effectively interrupt DNA synthesis which is essential for cell maintenance, thereby leading to cell death. Therefore, only those clones having exogenous DHFR gene inserted in their genome can survive under this condition. Furthermore, when the concentration of MTX being added is increased and a strong promoter is used, DHFR gene can be amplified to hundreds to thousands of copies. That is, the more MTX, a DHFR inhibitor, is added to the cell culture, in order for them to survive they increase the expression of DHFR along with the introduced target gene. Consequently, several copies of DNA will be incorporated, leading to the generation of various molecular variants.
  • A DHFR system using CHO cell line has been reported and commercialized as an expression system for various protein drugs, verifying its safety and efficacy in use. However, the DHFR system has a disadvantage in that it requires several months to isolate a single cell line that shows the expression level higher than the normal level. In addition, when the cell becomes resistant to MTX, even with an increase in MTX concentration, the target gene cannot be amplified anymore. Furthermore, in the CHO DUKX cells used for DHFR system, revertants may appear easily. As a result, there has been a high demand for the development of a high-level gene expression system for protein production other than the DHFR system.
  • GS system is a high-level gene expression system that was first developed by Celltech (U.S. Pat. No. 5,122,464), and it overcame the limitations of the DHFR-based gene expression system, that is, low time-efficiency for isolating the single cell line of interest and low productivity of target protein. The GS system utilizes glutamine synthetase (GS) which is an enzyme involved in the sole synthetic pathway for producing glutamine from glutamate and ammonia, based on the fact that animal cells cannot grow properly in the glutamine-deficient condition. The GS system has advantage in that it requires less number of DNA copies per cell compared to the DHFR system and allows for the selection of single cell line having high expression rate at the early stage of screening. Consequently, an increasing number of organizations adopt the GS system as a protein drug expression system. NS0 cell line and CHO cell line are the most commonly used cell lines for GS system. Between two, NS0 cell line which is a mouse myeloma cell line cannot express sufficient amount of GS, and thus in the glutamine-deficient condition, those cells where the target gene is inserted into their genome can be easily selected. Unlike the NSO cell line, a CHO cell line can express sufficient amount of GS that they can survive even in the glutamine-deficient medium. However, if the CHO cells are treated with a high concentration of GS-specific inhibitor such as methionine sulphoximine (MSX), the cells cannot survive only with the endogenous GS activity, and thus only those cells introduced with the vector comprising the GS gene and the gene for a target protein can survive. Through the above mechanism, the cells inserted with the gene for target protein can be isolated, and the target protein can be produced at high yield. In other words, as more GS-specific inhibitor is added to the cells, in order for them to survive, they will amplify exogenous GS gene as well as the target gene which is introduced together with the GS gene, thereby increasing the amount of target protein in the cell. However, even this GS system has a limitation in production amount when the cells transfected with the vector comprising the genes for target protein and GS are treated with the GS-specific inhibitor for producing the target protein. Also when the cells were cultured for a long time, the production amount of the target protein was reduced. Due to these limitations, there has been a high demand for the development of a modified GS protein that responses more sensitively to the GS inhibitor and thus can amplify the target gene introduced with GS to the greater level.
  • DISCLOSURE OF INVENTION Technical Problem
  • In an effort to develop a modified GS protein that has a significantly higher sensitivity to GS inhibitor compared to the wildtype GS protein, the present inventors have developed a modified GS with a significantly higher sensitivity to GS inhibitor having one amino acid substituted compared to the wildtype GS protein. Then, the present inventors confirmed the production of the target protein in the presence of GS inhibitor after transfecting the animal cells with the vector comprising the gene encoding the modified GS and target protein, and further confirmed that the modified GS demonstrates high sensitivity towards GS inhibitor, showing the remarkably higher level of target protein production compared to the vector system comprising the wildtype GS protein and also no reduction in the production level of target protein even after long period of culturing, thereby completing the present invention.
  • Solution to Problem
  • An object of the present invention is to provide a modified glutamine synthetase (GS), wherein glycine (Gly, G) at position 299 of a glutamine synthetase having an amino acid sequence shown in SEQ ID NO. 4 is substituted with arginine (Arg, R).
  • Another object of the present invention is to provide a polynucleotide encoding the modified glutamine synthetase.
  • Still another object of the present invention is to provide a vector for the expression of a target protein, comprising the polynucleotide and the gene encoding the target protein.
  • Still another object of the present invention is to provide a transformant comprising the vector.
  • Still another object of the present invention is to provide a method for the preparation of a target protein comprising culturing of the transformant and to provide the target protein prepared by said method.
  • Advantageous Effects of Invention
  • The expression vector comprising a modified GS gene of the present invention allows for the selection of the host cells introduced with the expression vector comprising the modified GS gene even under the condition where CHO cells are used as a host cell and the cells are treated with a GS inhibitor. Therefore, the present expression vector can be widely used for the efficient production of a target protein.
  • BRIEF DESCRIPTION OF DRAWINGS
  • FIG. 1 is a schematic diagram showing the cloning method for preparing pcDNA3.1-Kozak-TNFR-Fc-IRES-GS or pcDNA3.1-kozak-TNFR-Fc-IRES-GS PM of the present invention;
  • FIG. 2 is a cleavage map of the recombinant expression vector, pcDNA3.1-kozak-TNFR-Fc-IRES-GS or pcDNA3.1-kozak-TNFR-Fc-IRES-GS PM, comprising a gene encoding a TNFR-Fc fusion protein which is a representative target protein of the present invention;
  • FIG. 3 is a schematic diagram showing the cloning method for preparing the recombinant expression vector, pcDNA3.1-kozak-TNFR-Fc-SV40-GS or pcDNA3.1-kozak-TNFR-Fc-SV40-GS PM, comprising a gene encoding a TNFR-Fc fusion protein which is a representative target protein of the present invention;
  • FIG. 4 is a cleavage map of the recombinant expression vector, pcDNA3.1-kozak-TNFR-Fc-SV40-GS or pcDNA3.1-kozak-TNFR-Fc-SV40-GS PM, comprising a gene encoding a TNFR-Fc fusion protein which is a representative target protein of the present invention;
  • FIG. 5 is a graph showing the expression level of TNFR-Fc in CHO K-1 cells transfected with one of the three types of TNFR-Fc expression vectors (IRES-GS PM, IRES-GS, or SV40-GS);
  • FIG. 6 is a graph showing the changes in expression level of TNFR-Fc protein with MSX treatment over time after transfection of CHO K-1 cells with one of the three types of TNFR-Fc expression vectors (IRES-GS PM, IRES-GS, or SV40-GS);
  • FIG. 7 is a graph demonstrating the changes in TNFR-Fc expression level confirmed by the secondary transient transfection, showing a high expression level in the SV40-GS vector-transfected group during the early phase of culturing, but after longer period of culturing with the MSX treatment the highest expression level was observed in IRES-GS PM vector-transfected group; and
  • FIG. 8 is a graph showing the TNFR-Fc expression level in two groups of stable CHO-S cell line each transfected with either IRES-GS PM vector or SV40-GS PM vector.
  • BEST MODE FOR CARRYING OUT THE INVENTION
  • As one aspect, the present invention provides a modified glutamine synthetase (GS), wherein glycine (Gly, G) at position 299 of a glutamine synthetase having an amino acid sequence shown in SEQ ID NO. 4 is substituted with arginine (Arg, R).
  • As used herein, the term ‘glutamine synthetase (GS)’ refers to an enzyme that is found in the mammalian organs and microorganisms and that catalyzes the synthesis of glutamine from glutamate and ammonia in the presence of ATP. For activation of this enzyme, divalent metal ions are required, and its enzymatic activity is inhibited by the presence of glycine, alanine, tryptophan, histidine, glucosamine-6-phosphate, cytidine triphosphate, etc. For the purpose of the present invention, the glutamine synthetase refers to an enzyme that can be used to select the cells transfected with the vector comprising the gene encoding a target protein or to enhance the expression of the target protein by treatment with GS inhibitor, but is not limited thereto.
  • The information on the glutamine synthetase can be obtained from the common database such as NCBI GenBank. For instance, the information on glutamine synthetase derived from hamster can be found from GenBank with the accession number X03495.1, but is not limited thereto. The nucleotide sequence of the representative wild type glutamine synthetase is shown as SEQ ID NO. 3 and its amino acid sequence is shown as SEQ ID NO. 4.
  • As used herein, the term ‘modified glutamine synthetase’ refers to an enzyme wherein glycine (Gly, G) at position 299 of a glutamine synthetase modified glutamine synthetase (GS), wherein glycine (Gly, G) at position 299 of a glutamine synthetase having an amino acid sequence shown in SEQ ID NO. 4 is substituted with arginine (Arg, R).
  • The modified glutamine synthetase may be used as a selection marker by transfection of the cells that can or cannot produce GS endogenously with the target protein expression vector comprising the gene encoding the modified glutamine synthetase, or preferably refers to the protein that can enhance the target protein expression by treatment with GS inhibitor through polycistronic translation with the gene encoding the target protein in the form of ‘polynucleotide encoding the promoter-target protein encoding gene-IRES-modified glutamine synthetase’ or ‘polynucleotide encoding the promoter-modified glutamine synthetase-IRES-target protein encoding gene’, but is not limited thereto.
  • The inventor of the present invention has identified for the first time that if guanosine (G) in the codon of glycine at position 299 of the glutamine synthetase with SEQ ID NO. 4 is substituted with cytidine (C), the sensitivity of glutamine synthetase towards GS inhibitor is remarkably increased, as compared to the wild type GS. As the modified GS of the present invention has a significantly higher sensitivity towards GS inhibitor, it can be effectively used in the target protein expression system.
  • Furthermore, the modified glutamine synthetase comprises the amino acid sequence wherein glycine (Gly, G) at position 299 of a glutamine synthetase consisting of an amino acid sequence of SEQ ID NO. 4 is substituted with arginine (Arg, R), and as long as the enzyme demonstrates increased sensitivity towards GS inhibitor compared to the wildtype GS, the modified glutamine synthetase of the present invention may comprise the amino acid sequence having a sequence homology of 70% or higher, preferably 80% or higher, more preferably 90% or higher, even more preferably 95% or higher, even much more preferably 98% or higher, and most preferably 99% or higher to the above-described amino acid sequence of modified GS.
  • As used herein, the term ‘GS inhibitor’ refers to an external factor that is capable of inhibiting the GS activity. Examples of such inhibitor include glycine, alanine, tryptophan, histidine, glucosamine-6-phosphate, cytidine triphosphate, and methionine sulphoximine (MSX), but is not limited thereto. With respect to the objects of the present invention, the GS inhibitor is preferably MSX, but is not limited thereto.
  • As used herein, the term ‘sensitivity’ generally refers to the feature of responding to external stimuli. With respect to enzyme, sensitivity refers to the feature of enhancing or reducing enzymatic activity in response to external factors that regulate enzymatic activity. For the purpose of the present invention, the sensitivity refers to the suppression of the GS activity in response to the GS inhibitor, but is not limited thereto.
  • In one Example of the present invention, the gene encoding the modified glutamine synthetase was to be obtained through cloning the glutamine synthetase from RNA of hamster cells via PCR. The results confirmed that the modified glutamine synthetase was obtained in which glycine (Gly, G) at position 299 of wildtype glutamine synthetase of SEQ ID NO. 4 is substituted with arginine (Arg, R), and it was named GS PM (Example 3). Subsequently, IRES-GS PM was prepared by linking the polynucleotide encoding the modified glutamine synthetase with IRES, which was further connected to the gene encoding the target protein in order to prepare the vector comprising the expression cassette wherein the target protein encoding gene-IRES-GS PM are operably connected together (Example 4). Furthermore, when the target protein was expressed using the vector comprising the modified glutamine synthetase of the present invention, the production level of the target protein was significantly higher than when the wildtype glutamine synthetase was used (Examples 6 and 7).
  • As another aspect, the present invention provides a polynucleotide encoding the modified glutamine synthetase.
  • The modified glutamine synthetase of the present invention is characterized by having a substitution of the amino acid at position 299 of wild type GS from glycine to arginine. Thus, DNA codon encoding the amino acid at position 299 of the modified glutamine synthetase may be selected from the group consisting of CGT, CGC, CGA, CGG, AGA and AGG, but is not limited thereto.
  • Furthermore, the polynucleotide encoding the modified glutamine synthetase of the present invention may be preferably for enhancing the expression of target protein.
  • The polynucleotide encoding the modified glutamine synthetase of the present invention may be present in the vector to be translated by polycistronic translation with the gene encoding target protein. After transfection of the host cells with the above vector, when the cells are treated with GS inhibitor, the activity of the modified glutamine synthetase expressed from the introduced vector is inhibited, thereby reducing the synthesis of glutamine. However, in order for the cells to survive, glutamine is essential, and thus under the suppression by GS inhibitor those cells tend to synthesize more of the glutamine synthetase. Here, the target protein-encoding gene introduced along with the polynucleotide encoding the modified GS gets amplified as well, and through this mechanism the expression of the target protein can be increased.
  • As another aspect, the present invention provides a vector for expression of a target protein, comprising the polynucleotide encoding the modified GS and a gene encoding the target protein.
  • The polynucleotide encoding the modified GS is the same as described above.
  • As used herein, the term ‘target protein’ refers to the protein of interest to be produced in the host cells. For the purpose of the present invention, it refers to the protein whose expression is enhanced by the modified glutamine synthetase, but is not limited thereto. The type of target protein is not specifically limited as long as it can be expressed by the vector of the present invention. In one Example of the present invention, tumor necrosis factor receptor (TNFR)-Fc fusion protein was used as a representative target protein that can be expressed by the modified glutamine synthetase of the present invention.
  • As used herein, the term ‘tumor necrosis factor receptor (TNFR)-Fc fusion protein’ refers to the product prepared by connecting the entire or a part of TNFR protein with immunoglobulin Fc region by enzymatic action, or the product prepared by expressing two polypeptides into a single polypeptide by genomic manipulation. In the TNFR-Fc fusion protein, the TNFR protein and the immunoglobulin Fc region may be directly linked with each other, or linked via a peptide linker, but is not limited thereto. The polynucleotide encoding the TNFR-Fc fusion protein may be a polynucleotide of SEQ ID NO. 5, but is not limited thereto.
  • As used herein, the term ‘expression vector’ refers to a DNA construct comprising an essential control component which is operably linked to an insert gene so that the insert gene is only expressed when introduced into the host cell. The expression vector may be prepared and purified by a standard recombinant DNA technology. The type of the expression vector is not particularly limited, as long as it expresses and produces a target gene in a variety of host cells of prokaryotic and eukaryotic cells. Preferably, the expression vector is a vector capable of producing a large amount of a foreign protein in a similar form to the native protein while it retains a strong promoter activity and a strong expression ability. The expression vector is preferably a vector comprising at least a promoter, a start codon, a gene encoding a target protein, a stop codon, and a terminator. In addition, it may comprise a DNA encoding a signal peptide, an enhancer sequence, untranslated regions at the 5′ and 3′ ends of a target gene, a selectable marker region or a replicable unit, etc., if desired. Moreover, the type of the expression vector may be a mono-cistronic vector including a polynucleotide encoding one recombinant protein, a bi-cistronic vector including a polynucleotide encoding two recombinant proteins, a poly-cistronic vector including a polynucleotide encoding three recombinant proteins or more. With respect to the objects of the present invention, the expression vector is preferably a mono-cistronic vector including a SV40 promoter or a bi-cistronic vector including an IRES sequence, more preferably, an expression vector including a promoter, a gene encoding a target protein, IRES, and a modified GS gene in this order or an expression vector including a promoter, a modified GS gene, IRES, and a gene encoding a target protein, but is not limited thereto.
  • According to one embodiment of the present invention, a polynucleotide encoding GS PM was acquired from CHO DG44, and a pGEMT-GS PM vector was obtained by connecting each of the acquired polynucleotides to a pGEMT vector. Subsequently, the pGEMT-GS PM vector was inserted into a cleaved TOPO-IRES-DHFR vector to obtain a pCR2.1-TOPO-IRES-GS PM vector, and an IRES-GS PM fragment was obtained from the vector. The obtained IRES-GS PM fragment was inserted into a cleaved pcDNA-Kozak-TNFR-Fc-IRES-DHFR vector so as to construct a kozak-TNFR-Fc-IRES-GS PM vector ('IRES-GS PM vector') (FIG. 1).
  • According to another embodiment of the present invention, the polynucleotide encoding GS PM was inserted into the cleaved pcDNA3.1-TNFR-Fc-SV40-DHFR vector so as to obtain a pcDNA3.1-TNFR-Fc-SV40-GS PM vector. Thereafter, the pcDNA3.1-TNFR-Fc-SV40-GS PM vector was cleaved, and a TNFR-Fc fragment from the pcDNA3.1-Kozak-TNFR-Fc-IRES-DHFR vector was inserted into the cleaved region so as to construct a pcDNA-Kozak-TNFR-Fc-SV40-GS PM vector ('SV40-GS PM vector') (FIG. 3).
  • As another aspect, the present invention provides a transformant comprising the vector.
  • As used herein, the term ‘transformant’ refers to the cell transformed with the expression vector so as to express the polynucleotide encoding the recombinant protein included in the expression vector. It may be recombinant mammalian cells, rodent cells, preferably animal cells or animal-derived cells, and most preferably NS0 or CHO cells, but is not limited thereto. With respect to the objects of the present invention, the transformant is preferably a transformant prepared by introducing the expression vector into a NS0 or CHO cell line, but is not limited thereto.
  • As another aspect, the present invention provides a method for preparing a target protein, comprising culturing the transformant.
  • The transformant and target protein are the same as described above.
  • To be specific, the above method comprises (a) culturing the transformant; and (b) adding a GS inhibitor to a culture medium. In addition, the method further comprises (c) isolating the target protein from the culture medium.
  • Preferably, when the GS inhibitor is added to the medium, the expression system using the modified GS protein of the present invention, which has an enhanced sensitivity to GS inhibitor can lead to the increased expression of a target protein as compared to the expression system using the wildtype GS.
  • As another aspect, the present invention provides a target protein prepared by the above described method.
  • The method and the target protein are the same as described above.
  • MODE FOR THE INVENTION
  • Hereinafter, the present invention will be described in more detail with reference to Examples. However, these Examples are for illustrative purposes only, and the invention is not intended to be limited by these Examples.
  • EXAMPLE 1 Synthesis of TNFR-Fc Fusion Protein-Encoding Gene
  • In order to examine the expression level of a recombinant protein produced using a recombinant protein expression vector system, of the present invention, a TNFR-Fc fusion protein was used as a representative target protein.
  • The fusion protein-encoding gene (SEQ ID NO. 5) was synthesized by GeneArt Inc., so as to meet the following criteria: (1) it must include a TNFR signal sequence (2) it must express the TNFR amino acids at position 1 to 235 (3) it must be codon-optimized for CHO cells in order to be transfected into CHO cells (4) it must have a NheI restriction site at 5′-end and a NotI restriction site at 3′-end, considering insertion into a pcDNA3.1 vector of Invitrogen.
  • The nucleotide sequence of the synthesized fusion protein-encoding gene was finally analyzed using the VectorNTl program.
  • EXAMPLE 2 Construction of Expression Vector Comprising TNFR-Fc Fusion Protein-Encoding Gene
  • In the present invention, a DHFR system that is a common recombinant protein expression system was utilized as a control for the recombinant protein expression system using the modified GS protein. For this, a hamster dihydrofolate reductase (DHFR) gene was cloned as detailed below:
  • In order to obtain the hamster DHFR gene, a pSVA3 vector (ATCC 77273) having a mutant type of hamster DHFR gene was purchased, and then a wild type of DHFR gene was obtained by performing point mutation using the DHFR gene as a template. In addition, an IRES sequence was obtained by PCR from a Clontech vector (Cat. #6029-1, PT3267-5) having the corresponding DNA sequence.
  • The obtained DHFR gene and internal ribosome entry site (IRES) sequence were cloned into a pCR2.1 vector so as to construct a pCR2.1-IRES-DHFR expression vector.
  • Each of the TNFR-Fc-inserted pcDNA3.1-TNFR-Fc vector obtained in Example 1 and the obtained pCR2.1-IRES-DHFR vector was digested with restriction enzymes, SalI and XbaI, and ligated so as to obtain a TNFR-Fc-inserted pcDNA3.1-TNFR-Fc-IRES-DHFR expression vector.
  • In order to clone it into a vector having a kanamycin-resistant gene, the kanamycin-resistant gene was obtained from a pAC-GFP vector (#632483) of Clontech, so as to introduce a Kan/Neo gene. The vector is a vector having a Kozak sequence at a transcription initiation sequence of the TNFR-Fc gene and the Kan/Neo gene as an antibiotic selection marker, and it was used as a basic frame for cloning 4 different expression vector systems in order to compare the expression levels of the recombinant protein using CHO cells.
  • EXAMPLE 3 Preparation of a Modified GS Gene
  • In order to acquire a modified GS gene which has an increased sensitivity to GS inhibitor as compared to wildtype GS and thus can be applied to the target protein expression system, the following procedures were performed.
  • To acquire the modified GS gene, a hamster cell line, CHO DG44 (Invitrogen, 12609-012) was cultured, and then total RNA was isolated using a TRIZOL reagent (Invitrogen). After that, RT-PCR was performed using the obtained total RNA so as to obtain cDNA. PCR (25 cycles of denaturation at 94° C. for 5 minutes; denaturation at 94° C. for 30 seconds, annealing at 50° C. for 30 seconds, elongation at 72° C. for 90 seconds; and elongation at 72° C. for 7 minutes) was performed using the obtained cDNA as a template and a pair of primers (GS SalI-F primer and GS XbaI-R primer) for acquisition of the following GS PM gene, so as to obtain a PCR product.
  • GS SalI-F (Forward primer):
    (SEQ ID NO. 6)
    5′-gtcgacatggccacctcagcaagttccc-3′
    GS XbaI-R (Reverse primer):
    (SEQ ID NO. 7)
    5′-tctagattagtttttgtattggaaaggg-3′
  • The obtained PCR product was electrophoresed on a 0.8% agarose gel, and then the corresponding band was cut, followed by clean-up using a Quiagen Cleaning kit (#28204). Then, the resultant was inserted into a gene cloning vector, pGEMT vector (Promega, USA). The PCR product-inserted pGEMT vector was introduced into a TOP10 cell so as to obtain a total of 10 colonies. After that, a nucleotide sequence and an amino acid sequence encoded by the nucleotide sequence of each colony were analyzed.
  • By performing the above procedures numerous times, a modified GS gene showing a difference in one amino acid at position 299 as compared to the amino acid sequence of the wild type hamster GS (NCBI GenBank: X03495.1), was acquired. As a result of sequence analysis, this difference is attributed to the alteration of the 895th nucleotide of the wildtype GS gene (SEQ ID NO. 3) from G (Guanosine) to C (Cytidine). That is, the modified GS has the characteristic of altered amino acids at position 299 from glycine (Gly, G) to arginine (Arg, R). In the present invention, the acquired modified GS was named ‘GS PM’.
  • Further, to acquire a polynucleotide encoding the wild type hamster GS protein, the 895th C in the nucleotide sequence of GS-PM was substituted with G. Specifically, cloning was performed by point mutation for replacement of one amino acid. More specifically, in order to obtain the wild type GS having one amino acid different from those of GS PM of the present invention, PCR (30 cycles of denaturation at 94° C. for 5 minutes; denaturation at 94° C. for 30 seconds, annealing at 54° C. for 30 seconds, elongation at 72° C. for 30 seconds; and elongation at 72° C. for 7 minutes) was performed using the GS PM DNA as a template and a pair of primers (KpnI F-primer and XbaI R-primer) that was synthesized to contain a point mutation region (CGT→GGT). As a result, a GS PCR fragment having an alteration from CGT to GGT was obtained.
  • KpnI F-primer (Forward primer):
    (SEQ ID NO. 8)
    5′-caccggtaccacattcgagcctacgatcccaaggggggcctggacaa 
    tgcccgtggtctg-3′
    XbaI R-primer (Reverse primer):
    (SEQ ID NO. 9)
    5′-tctagattagtttttgtattggaaggg-3′
  • In addition, the nucleotide sequence of the PCR product was analyzed. As a result, a point mutation from CGT to GGT was observed. The corresponding PCR product was digested with KpnI and XbaI, and then ligated with a pGEMT-GS PM vector treated with KpnI and XbaI, so as to obtain a pGEMT-GS vector.
  • EXAMPLE 4 Cloning of a Mammalian Cellular Protein-Expressing GS Vector
  • The GS PM gene included in the pGEMT-GS PM vector obtained in Example 3 was cloned to have SalI and XbaI restriction sites at its N- and C-terminals, respectively. Therefore, in order to obtain IRES-GS PM, a fragment obtained by treating pGEMT-GS PM with SalI and XbaI restriction enzymes was inserted into a TOPO-IRES-DHFR vector that was previously digested with SalI and XbaI restriction enzymes, so as to obtain a pCR2.1-TOPO-IRES-GS PM gene.
  • Next, in order to connect the TNFR-Fc gene and the IRES-GS PM gene, the TNFR-Fc-IRES-DHFR gene and the IRES-GS PM fragment digested with XhoI and XbaI were ligated so as to construct a kozak-TNFR-Fc-IRES-GS PM vector (“IRES-GS PM vector”) (FIG. 1). FIG. 1 is a schematic diagram showing the cloning method of pcDNA3.1-kozak-TNFR-Fc-IRES-GS PM of the present invention.
  • Further, the kozak-TNFR-Fc-IRES-GS PM vector and the TOPO-GS vector were used to construct a Kozak-TNFR-Fc-IRES-GS vector (“IRES-GS vector”) (FIG. 2).
  • FIG. 2 is a cleavage map showing pcDNA3.1-kozak-TNFR-Fc-IRES-GS or pcDNA3.1-kozak-TNFR-Fc-IRES-GS PM, which is a recombinant expression vector including the TNFR-Fc-encoding gene of the present invention.
  • Meanwhile, the cloned pcDNA3.1-TNFR-Fc-SV40-DHFR vector was used in order to prepare a SV40 promoter-GS system identical to Lonza's GS system among GS systems. Since the pcDNA3.1-TNFR-Fc-SV40-DHFR has no restriction sites suitable for GS gene insertion, new restriction sites were first inserted into both ends of the GS gene.
  • A pair of primers (GS-BsaBI-F primer and GS-BstBI-R primer) containing BsaBI at the N-terminal of GS gene and BstBI at C-terminal of GS gene were synthesized to perform PCR. Thus, the BsaBI and BstBI sites were inserted into both ends of the GS gene, and DHFR was removed from pcDNA3.1-TNFR-FC-SV40-DHFR by treatment with BsaBI and BstBI. Subsequently, the GS gene digested with BsaBI/BstBI was inserted thereto so as to construct a pcDNA3.1-TNFR-Fc-SV40-GS PM vector.
  • GS-BsaBI-F Primer (forward primer):
    (SEQ ID NO. 10)
    5′-gatgaggatcatggccacctcagcaag-3′
    GS-BstBI-R (reverse primer):
    (SEQ ID NO. 11)
    5′-ttcgaattagtttttgtattggaaggg-3′
  • However, the pcDNA3.1-TNFR-Fc-SV40-GS PM vector has no Kozak sequence prior to the TNFR-Fc gene, unlike the pcDNA3.1-TNFR-Fc-IRES-GS PM vector. Therefore, a second cloning step of inserting the Kozak sequence into the vector was performed. For the Kozak sequence, a Kozak sequence of the previously prepared pcDNA3.1-Kozak-TNFR-Fc-IRES-DHFR vector was used. Instead of TNFR-Fc of the pcDNA3.1-TNFR-Fc-SV40-GS PM vector, the Kozak-TNFR-Fc was inserted to prepare pcDNA3.1-Kozak-TNFR-FC-SV40-GS PM.
  • The restriction enzymes to be used for the cloning may include NdeI and NheI at N-terminal of TNFR-Fc and BstXI, SgrAI, and NotI at C-terminal of TNFR-Fc. Available restriction enzymes were selected from them, so as to construct a pcDNA-Kozak-TNFR-Fc-SV 40-GS PM vector (“SV40-GS PM vector”) (FIG. 3). FIG. 3 is a schematic diagram showing the cloning method of pcDNA3.1-kozak-TNFR-Fc-SV40-GS or pcDNA3.1-kozak-TNFR-Fc-SV40-GS PM, which is a recombinant expression vector including the TNFR-Fc-encoding gene of the present invention.
  • The constructed SV40-GS PM vector includes an antibiotic resistance gene, ampicillin resistance gene for cell line selection, and thus a third cloning step was performed to replace the gene with a kanamycin resistance gene. The antibiotic resistance gene was replaced by an antibiotic with a low frequency of use on grounds of safety, because ampicillin is one of the antibiotics frequently used by patients.
  • In order to perform the cloning for replacement with the kanamycin resistance gene at a low frequency of use, the kanamycin resistance gene was obtained from the pAC-GFP vector (#632483) of Clontech, and introduced into a SV40-GS PM vector. Furthermore, the SV40-GS PM vector was used to construct a pcDNA-Kozak-TNFR-Fc-SV 40-GS vector (‘SV40-GS vector’) (FIG. 4). FIG. 4 is a cleavage map showing pcDNA3.1-kozak-TNFR-Fc-SV40-GS or pcDNA3.1-kozak-TNFR-Fc-SV40-GS PM, which is a recombinant expression vector including the TNFR-Fc-encoding gene of the present invention.
  • Finally, 4 different expression vectors of IRES-GS, IRES-GS PM, SV40-GS, and SV40-GS PM were constructed as a protein expression vector system for expressing a target protein in mammalian cells.
  • EXAMPLE 5 Preparation of Transformant
  • In order to compare the productivity of TNFR-Fc fusion protein between the GS and DHFR gene expression systems, each of the expression vectors constructed in Example 4 was introduced into CHO K-1 cells so as to prepare each transformant.
  • In detail, CHO K-1 cells were cultured in a DMEM/F12 medium supplemented with 10% FBS, and 3 to 4×105 cells/well were inoculated into a 6 well-plate, followed by cultivation overnight. When the cells reached 80 to 90% confluence, each of the expression vectors was introduced thereto.
  • To achieve this, 4 μg of each of the expression vectors constructed in Example 4 and 250 μl of Opti-MEM were mixed with each other, and separately, 10 μl of Lipofectamine 2000 (Invitrogen) and 250 μl of Opti-MEM were mixed with each other. Then, each mixture was left at room temperature for 5 minutes. Subsequently, the mixtures were mixed with each other, and left at room temperature for 20 minutes.
  • Thereafter, the culture medium in the 6-well plate was replaced with 2 ml of Opti-MEM I media, and 500 μl of the final mixture was added to each well of the 6-well plate, followed by cultivation at 37° C. for 4 to 6 hours. The medium was replaced with the original culture medium (DMEM/F12 medium supplemented with 10% FBS), followed by further cultivation overnight. Then, the culture medium in the 6-well plate was replaced with a GS selection medium, and subculture was performed according to the growth rate of the cells introduced with each of the expression vectors. At this time, the GS selection medium was glutamine-free DMEM or glutamine-free IMDM containing 10% FBS, 1× GS supplement, and the GS inhibitor, MSX. The 1× GS supplement was prepared by including adenosine (500×, 15 mM), cytidine (1000×, 30 mM), uridine (1000×, 30 mM), guanosine (1000×, 3 mM), thymidine (1000×, 10 mM), asparagine (1000×, 500 mM) and glutamic acid (1000×, 500 mM) or a commercially available 50× GS supplement (SAFC) was used after dilution.
  • EXAMPLE 6 Assessment of TNFR-Fc Expression Level
  • The culture broth of each transformant subcultured in Example 5 was applied to ELISA, in order to measure the expression level of each protein expressed from the transformant.
  • In detail, a 96-well plate was coated with anti-human IgG Fc antibody (Pierce, 31125), and blocked with 1% BSA. Next, the culture broth of each subcultured transformant was added to each well, and reacted. Subsequently, a biotin-conjugated anti-human TNFR antibody (R&D system) as a detection antibody was added to each well, and reacted. Each well was treated with HRP-conjugated streptavidin, and reacted. Finally, each well was treated with TMB for color development so as to examine the expression level of each protein.
  • First, the expression levels in CHO K-1 were compared between three types of TNFR-Fc GS vector (IRES-GS PM, IRES-GS, SV40-GS) and the IRES-DHFR system, and the protein expression levels were also compared between the GS vectors (FIG. 5). FIG. 5 is a graph showing the expression levels of TNFR-Fc in CHO K-1 cells transfected with three types of TNFR-Fc gene expression vector (IRES-GS PM, IRES-GS, SV40-GS).
  • As a result, as shown in FIG. 5, even at 64 hours after transfection with the expression vectors, CHO K-1 cells showed no changes in the expression level of TNFR-FC protein by three types of GS vectors or in the cell survival rate. Rather, three types of the GS systems were more excellent in terms of expression level than the IRES-DHFR system. Moreover, even at 5 days after transfection of the expression vectors, the cells transfected with the three types of expression vectors maintained their growth and continuously expressed the TNFR-Fc protein.
  • Next, it was also examined whether the same results can be obtained when a glutamine-free selection media containing 25 μm of MSX was used to culture the cells transfected with the three types of the TNFR-Fc gene expression vectors (IRES-GS PM, IRES-GS, SV40-GS) (FIG. 6). FIG. 6 is a graph showing the time-dependent expression level of TNFR-Fc protein according to initial MSX treatment of CHO K-1 cells, which were transfected with the three types of TNFR-Fc gene expression vectors (IRES-GS PM, IRES-GS, SV40-GS).
  • As shown in FIG. 6, as the cell culture time increased, the cell line transfected with the IRES-GS PM vector of the present invention exhibited higher levels in terms of the total TNFR-Fc protein expression level and the expression level per an equal number of cells, compared to the cell lines transfected with other vectors. Immediately after transfection of the expression vectors, the cells transfected with the SV40-GS vector exhibited the highest level. However, after addition of MSX, the cells transfected with the IRES-GS PM vector of the present invention exhibited the highest level.
  • Meanwhile, the overall analysis of the results of FIG. 5 showed that there is a great difference in the expression level between the IRES-GS and IRES-GS PM, even though both of them include the identical IRES. At the beginning of the experiment, it was expected that there would be no difference in their activities, because of only one amino acid difference between the two genes. Actually, there was a difference between GS and GS PM, as the cultivation maintained in the selection media.
  • Therefore, in order to clearly examine whether one amino acid difference between GS and GS PM greatly affects the expression level, the SV40-GS PM vector was used to compare the expression levels of TNFR-Fc protein between the cell lines transfected with a total of 4 types of vectors (IRES-GS, TRES-GS PM, SV40-GS, SV40-GS PM) (FIG. 7). FIG. 7 is a graph showing the TNFR-Fc expression level after a secondary transient transfection (upper graph) and the time-dependent TNFR-Fc expression level (lower graph).
  • As a result, as shown in the upper graph of FIG. 7, the TNFR-Fc expression levels were mostly 6000-9000 ng/106 cells when the cells were cultured without MSX for 6 days after transfection, and the TNFR-Fc expression levels were relatively increased in the cells transfected with the wild type, TRES-GS and SV40-GS vectors. However, as shown in the lower graph of FIG. 7, when the cells were treated with 25 μM MSX at 3 days after transfection and 200 μm MSX at 20 days after transfection, and then cultured for further 7 days, the TNFR-Fc expression levels were 1000 ng/106 cells or lower, immediately after the addition of 25 μM MSX, but the expression levels increased according to time. After the addition of 200 μM MSX, only the cells transfected with the IRES-GS PM vector of the present invention showed an increase in the TNFR-Fc expression level.
  • The above results suggest that the modified GS protein of the present invention shows an enhanced sensitivity to GS inhibitors as compared to that of the wildtype GS protein, and the vector comprising the polynucleotide encoding the modified GS protein and the gene encoding the target protein can be effective to produce the target protein.
  • EXAMPLE 7 Comparison of TNFR-Fc Protein Expression Level in Stably Transfected CHO-S cells
  • The results of Example 6 showed that the higher protein expression level per an equal number of cells was observed in the cell lines transfected with the GS-containing expression vectors (IRES-GS PM and SV40-GS PM expression vectors) among the four types of GS expression vectors. Therefore, the expression levels in the stable CHO-S cell line were compared between the IRES-GS PM and SV40-GS PM expression vectors. In order to produce a large amount of recombinant protein, the CHO-S cell adapted for growth in suspension was used for the stable cell line establishment, instead of CHO K-1.
  • First, TNFR-Fc-expressing cell lines transfected with the IRES-GS PM and SV40-GS PM expression vectors were established. Each of the established TNFR-Fc-expressing cell lines was cultured in a medium containing 25 μM or 250 μM MSX, and the expression levels of TNFR-Fc protein expressed therefrom were compared (FIG. 8). FIG. 8 is a graph showing the comparison in the stable cell line between the IRES-GS PM vector and the SV40-GS PM vector. In this regard, PCD indicates pg/cell/day, and it was calculated from the following Equation.

  • PCD=expression amount (ng/mL)/((A−B)*Culture day/LN(A/B))/1000
  • A: Harvest cell conc.(×106 cells/mL)
  • B: Seed cell conc.(×106 cells/mL)
  • As a result, as shown in FIG. 8, the cells transfected with the IRES-GS PM expression vector showed a higher expression level of TNFR-FC protein than those transfected with the SV40-GS PM expression vector, irrespective of MSX concentration. In addition, the maximum PCD at 250 μM MSX was ˜8 PCD, which is higher than that of a single cell line limiting-diluted using the known IRES-DHFR system. When single cell lines are selected from the cell groups of 8 PCD, it was expected to obtain the expression cell lines of higher than 8 PCD.
  • The above results demonstrate that the expression system using the vector comprising the modified GS gene of the present invention, specifically, the polycistronic vector comprising the modified GS gene of the present invention, can produce the target protein with high efficiency.
  • While the present invention has been particularly shown and described with reference to the foregoing preferred and alternative embodiments, it should be understood by those skilled in the art that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention without departing from the spirit and scope of the invention as defined in the following claims. It is intended that the following claims define the scope of the invention and that the method and apparatus within the scope of these claims and their equivalents be covered thereby. This description of the invention should be understood to include all novel and non obvious combinations of elements described herein, and claims may be presented in this or a later application to any novel and non-obvious combination of these elements.

Claims (16)

1. A modified glutamine synthetase (GS), wherein glycine (Gly, G) at position 299 of a glutamine synthetase having an amino acid sequence shown in SEQ ID NO. 4 is substituted with arginine (Arg, R).
2. The modified glutamine synthetase according to claim 1, having an increased sensitivity to a glutamine synthetase inhibitor, compared to the glutamine synthetase having an amino acid sequence shown in SEQ ID NO.4.
3. The modified glutamine synthetase according to claim 2, wherein the glutamine synthetase inhibitor is selected from the group consisting of glycine, alanine, tryptophan, histidine, glucosamine-6-phosphate, cytidine triphosphate, methionine sulphoximine (MSX), and combinations thereof.
4. (canceled)
5. (canceled)
6. (canceled)
7. (canceled)
8. (canceled)
9. (canceled)
10. (canceled)
11. (canceled)
12. (canceled)
13. (canceled)
14. (canceled)
15. (canceled)
16. A target protein prepared by a method comprising culturing a transformant comprising a vector for expression of a target protein, comprising the polynucleotide encoding a modified glutamine synthetase wherein glycine (Gly, G) at position 299 of a glutamine synthetase having an amino acid sequence shown in SEQ ID NO. 4 is substituted with arginine (Arg, R).
US15/399,432 2012-03-12 2017-01-05 Expression Vector Comprising a Polynucleotide Encoding a Modified Glutamine Synthetase and a Method for Preparing a Target Protein Employing the Same Abandoned US20170211057A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/399,432 US20170211057A1 (en) 2012-03-12 2017-01-05 Expression Vector Comprising a Polynucleotide Encoding a Modified Glutamine Synthetase and a Method for Preparing a Target Protein Employing the Same

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
KR20120025197 2012-03-12
KR10-2012-0025197 2012-03-12
PCT/KR2013/001779 WO2013137583A1 (en) 2012-03-12 2013-03-05 An expression vector comprising a polynucleotide encoding a modified glutamine synthetase and a method for preparing a target protein employing the same
US201414384594A 2014-09-11 2014-09-11
US15/399,432 US20170211057A1 (en) 2012-03-12 2017-01-05 Expression Vector Comprising a Polynucleotide Encoding a Modified Glutamine Synthetase and a Method for Preparing a Target Protein Employing the Same

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/KR2013/001779 Division WO2013137583A1 (en) 2012-03-12 2013-03-05 An expression vector comprising a polynucleotide encoding a modified glutamine synthetase and a method for preparing a target protein employing the same
US14/384,594 Division US9567577B2 (en) 2012-03-12 2013-03-05 Expression vector comprising a polynucleotide encoding a modified glutamine synthetase and a method for preparing a target protein employing the same

Publications (1)

Publication Number Publication Date
US20170211057A1 true US20170211057A1 (en) 2017-07-27

Family

ID=49161429

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/384,594 Expired - Fee Related US9567577B2 (en) 2012-03-12 2013-03-05 Expression vector comprising a polynucleotide encoding a modified glutamine synthetase and a method for preparing a target protein employing the same
US15/399,432 Abandoned US20170211057A1 (en) 2012-03-12 2017-01-05 Expression Vector Comprising a Polynucleotide Encoding a Modified Glutamine Synthetase and a Method for Preparing a Target Protein Employing the Same

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US14/384,594 Expired - Fee Related US9567577B2 (en) 2012-03-12 2013-03-05 Expression vector comprising a polynucleotide encoding a modified glutamine synthetase and a method for preparing a target protein employing the same

Country Status (6)

Country Link
US (2) US9567577B2 (en)
EP (1) EP2825641A4 (en)
JP (1) JP2015513406A (en)
KR (1) KR101494072B1 (en)
BR (1) BR112014022409A2 (en)
WO (1) WO2013137583A1 (en)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013137583A1 (en) * 2012-03-12 2013-09-19 Hanwha Chemical Corporation An expression vector comprising a polynucleotide encoding a modified glutamine synthetase and a method for preparing a target protein employing the same
US11098310B2 (en) 2016-01-27 2021-08-24 Just-Evotec Biologics, Inc. Expression from transposon-based vectors and uses
US11028410B2 (en) 2016-01-27 2021-06-08 Just-Evotec Biologics, Inc. Hybrid promoter and uses thereof
US11261462B2 (en) 2016-01-27 2022-03-01 Just-Evotec Biologics, Inc. Inducible expression from transposon-based vectors and uses
US20190352631A1 (en) * 2016-11-16 2019-11-21 Agency For Science Technology And Research Attenuated Glutamine Synthetase as a Selection Marker
CA3064352A1 (en) 2017-05-24 2018-11-29 Thoeris Gmbh Use of glutamine synthetase for treating hyperammonemia
US11713468B2 (en) 2017-08-02 2023-08-01 Dna Twopointo Inc. DNA vectors and elements for sustained gene expression in eukaryotic cells
CN113005140B (en) * 2019-12-19 2024-03-15 鲁南制药集团股份有限公司 GS expression vector with double expression cassettes and application thereof
KR102198072B1 (en) * 2020-03-04 2021-01-04 씨제이제일제당 주식회사 A modified polypeptide of glutamine synthetase and a method for L-glutamine using the same

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030235536A1 (en) * 2002-03-15 2003-12-25 The Brigham And Women's Hospital, Inc. Central airway administration for systemic delivery of therapeutics
US9567577B2 (en) * 2012-03-12 2017-02-14 Ares Trading S.A. Expression vector comprising a polynucleotide encoding a modified glutamine synthetase and a method for preparing a target protein employing the same

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8601597D0 (en) 1986-01-23 1986-02-26 Wilson R H Nucleotide sequences
US7294481B1 (en) * 1999-01-05 2007-11-13 Immunex Corporation Method for producing recombinant proteins
KR100982922B1 (en) * 2002-01-17 2010-09-20 론자 바이올로직스 피엘씨 Glutamine-auxothrophic human cells capable of producing proteins and capable of growing in a glutamine-free medium
JP2003274963A (en) * 2002-03-22 2003-09-30 Japan Science & Technology Corp Gene recombinant cell strain and liver function supporting equipment using the same
KR100976098B1 (en) * 2002-07-18 2010-08-16 론자 바이올로직스 피엘씨 Method of expressing recombinant protein in cho cells
US7244616B2 (en) 2003-06-27 2007-07-17 Bayer Pharmaceuticals Corporation Use of molecular chaperones for the enhanced production of secreted, recombinant proteins in mammalian cells
KR101454316B1 (en) * 2011-08-17 2014-10-27 한화케미칼 주식회사 Method for Preparing Active Form of TNFR-Fc Fusion Protein
WO2013161958A1 (en) * 2012-04-27 2013-10-31 日本ケミカルリサーチ株式会社 Novel expression vector

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030235536A1 (en) * 2002-03-15 2003-12-25 The Brigham And Women's Hospital, Inc. Central airway administration for systemic delivery of therapeutics
US9567577B2 (en) * 2012-03-12 2017-02-14 Ares Trading S.A. Expression vector comprising a polynucleotide encoding a modified glutamine synthetase and a method for preparing a target protein employing the same

Also Published As

Publication number Publication date
KR101494072B1 (en) 2015-02-17
EP2825641A1 (en) 2015-01-21
WO2013137583A1 (en) 2013-09-19
JP2015513406A (en) 2015-05-14
US9567577B2 (en) 2017-02-14
EP2825641A4 (en) 2015-09-30
US20150044753A1 (en) 2015-02-12
KR20130105380A (en) 2013-09-25
BR112014022409A2 (en) 2018-09-25

Similar Documents

Publication Publication Date Title
US9567577B2 (en) Expression vector comprising a polynucleotide encoding a modified glutamine synthetase and a method for preparing a target protein employing the same
AU2003302689B2 (en) Novel neomycin phosphotransferase genes and method for the selection of high-producing recombinant cells
ES2421152T3 (en) New regulatory elements
JP5432117B2 (en) Recombinant expression vector element (rEVE) for enhancing expression of recombinant protein in a host cell
US9657310B2 (en) Expression vector
RU2494147C2 (en) Expression vector of mammals
US20210317435A1 (en) Double knock-out cho cell line method of its generation and producing therapeutic proteins therefrom
CN104884467A (en) Production of therapeutic proteins in genetically modified mammalian cells
US7344886B2 (en) Neomycin-phosphotransferase-genes and methods for the selection of recombinant cells producing high levels of a desired gene product
CN106255749A (en) Novel spinal zooblast and method for recombinant expressed polypeptide of interest
Mensah et al. Establishment of DHFR-deficient HEK293 cells for high yield of therapeutic glycoproteins
EP3699269A1 (en) Expression of fc-containing proteins
RU2656142C1 (en) Recombinant plasmid dna pbipr-abiga1fi6-ht for obtaining recombinant immunoglobulin a igoth iga1
KR20210141511A (en) Novel Selectable Marker-Containing Cell Lines and Their Uses for Protein Production
JP4555373B2 (en) Novel neomycin phosphotransferase gene and method for selecting highly producing recombinant cells
Zucchetti Transposon based technology in DHFR knockout CHO cell line improves generation of AMH high producing clones for industrial applications
US9315565B2 (en) Method for producing protein
Schüler Development of a novel expression system for the production of recombinant proteins in Chinese hamster ovary cells based on the selection with a metabolic enzyme

Legal Events

Date Code Title Description
AS Assignment

Owner name: HANWHA CHEMICAL CORPORATION, KOREA, REPUBLIC OF

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:JANG, HYUN SOOK;LEE, DONG HEON;KIM, SUN KYU;AND OTHERS;REEL/FRAME:042205/0808

Effective date: 20140922

Owner name: ARES TRADING S.A., SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:HANWHA CHEMICAL CORPORATION;REEL/FRAME:042384/0334

Effective date: 20150925

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION