WO2005032245A1 - Method of producing transgenic pigs expressing green fluorescent protein - Google Patents

Method of producing transgenic pigs expressing green fluorescent protein Download PDF

Info

Publication number
WO2005032245A1
WO2005032245A1 PCT/KR2004/001414 KR2004001414W WO2005032245A1 WO 2005032245 A1 WO2005032245 A1 WO 2005032245A1 KR 2004001414 W KR2004001414 W KR 2004001414W WO 2005032245 A1 WO2005032245 A1 WO 2005032245A1
Authority
WO
WIPO (PCT)
Prior art keywords
pig
oocyte
fluorescent protein
somatic cell
transgenic
Prior art date
Application number
PCT/KR2004/001414
Other languages
French (fr)
Inventor
Kwang-Wook Park
Jae-Goo Seol
Ki-Nam Heo
Seung-Hyeon Lee
Seung-Pyo Hong
Ki-Myung Choi
Original Assignee
Mgenbio, Inc.
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 Mgenbio, Inc. filed Critical Mgenbio, Inc.
Publication of WO2005032245A1 publication Critical patent/WO2005032245A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/027New breeds of vertebrates
    • A01K67/0273Cloned animals
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/027New breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/8509Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
    • 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
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/873Techniques for producing new embryos, e.g. nuclear transfer, manipulation of totipotent cells or production of chimeric embryos
    • C12N15/877Techniques for producing new mammalian cloned embryos
    • C12N15/8778Swine embryos
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/108Swine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/0393Animal model comprising a reporter system for screening tests
    • 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

Definitions

  • the present invention relates to a method of producing a transgenic pig expressing green fluorescent protein (GFP) , more specifically to a method of producing a transgenic pig expressing green fluorescent protein available for the field of producing therapeutic proteins and for the cell therapy.
  • GFP green fluorescent protein
  • Cloning means producing a number of genetically identical individuals.
  • One of the cloning techniques is an attractive nuclear transfer resulting from a great progress of molecular biology.
  • a blastomere of a fertilized egg was used as a nuclear source in the early nuclear transfer (Prather, et al., Biol . Reprod. , 1987, 37:856-866; Prather, et al . , Biol . Reprod. , 1989, 41:414-418), but it is recently performed by using the nucleus of a somatic cell.
  • a cloning technique of a somatic cell is to introduce an nucleus of a differentiated somatic cell into an enucleated oocyte to develop it in the same manner as that of a general fertilized egg. It is expected that this cloning technique is not only available for the studies of basic science including developmental biology but also for production of a therapeutic protein, especially in the field of medical science, for example developing a disease model and organ transplantation etc, so that it may be greatly useful for the industrial availability.
  • the first transgenic cloned pig was then produced by using the somatic cells introduced a exogenous gene (Park, KW. et al., Anim . Biotechnol . , 2001, 12:173-181). Then, another cloned pig, in which a specific gene was knockout in a somatic cell, was produced (Lai, L. et al., Na t . Biotechnol . , 2002 Mar, 20 (3 ): 251-255 ) .
  • somatic cell cloning with pigs has been rapidly progressed, the cloning efficiency is still very low with 1-5%, and therefore further studies are required to improve the efficiency.
  • Somatic cell cloning includes the steps of eliminating a nucleus from an oocyte, injecting a somatic cell into the enucleated oocyte, followed by fusing the resulting oocyte with the somatic cell to form a oocyte-somatic cell complex and activating it under electro-stimulation.
  • the conventional method for example microinjection of DNA has a problem of low productivity of a transgenic animal.
  • GFP green fluorescent protein
  • Aequorea ictoria fluorescent jellyfish
  • somatic cells used for the cloning above were not clonal somatic cells, integration sites of GFP gene into chromosome were all different among offsprings.
  • Retrovirus was used to insert GFP gene into cells, which has problems of limitation of size of a gene to be transferred to cells and difficulty in securing stability for a clinical application.
  • the present inventors prepared a clonal somatic cell line transfected with a foreign gene using an lipid-based reagent instead of retrovirus. And the present inventors completed this invention by confirming that a transgenic cloned pig specifically expressing a green fluorescent protein in the muscle tissues may be produced using the nuclear transfer technique of the clonal somatic cells .
  • the present invention provides a method of producing a transgenic pig expressing a green fluorescent protein including the steps of; 1) separating somatic cells from a porcine fetus; 2) preparing an expression vector which includes a green fluorescent gene, followed by introducing the expression vector into the above somatic cells; 3) selecting cloned somatic cells transfected with the above expression vector and culturing those selected cells; 4) eliminating a nucleus from an oocyte of a donor pig, followed by fusing the resulting oocyte with the somatic cell to form a oocyte-somatic cell complex; and 5) transplanting the fused replicative oocyte into a surrogate pig and delivering piglets of present invention.
  • the present invention also provides a transgenic pig specifically expressing a green fluorescent protein in muscle tissues produced by the method of the present invention.
  • cloned somatic cell lines used herein means cell lines having the identical integration site of a foreign vector.
  • peripheral border means the space between zona pellucida and ooplasm of an egg.
  • oocyte-somatic cell complex means pre-cell fusion status after eliminating a nucleus from an oocyte and injecting a somatic cell into its perivitelline space.
  • a cloned egg i.e., a replicative egg
  • the present invention provides a method of producing a transgenic pig expressing a green fluorescent protein, comprising the following steps: 1) Establishing somatic cells from a porcine fetus; 2) Preparing an expression vector which includes a green fluorescent gene, followed by introducing the expression vector into the above somatic cells; 3) Selecting cloned somatic cells transfected with the above expression vector and culturing those selected cells; 4) Eliminating a nucleus from an oocyte of a donor pig, followed by fusing the resulting oocyte with the somatic cell to form a oocyte-somatic cell complex; and 5) Transplanting the fused replicative oocyte into a surrogate pig and delivering piglets of present invention.
  • a porcine fetus is preferably an embryo at day 20 - 50 after pregnancy, and is more preferably an embryo at day 30 - 40 after pregnancy.
  • the present inventors established somatic cells from a porcine fetus at day 35 after pregnancy.
  • somatic cells from the porcine fetus a conventional method was applied equally to this invention and, in the preferred embodiment of the present invention a small portion of a porcine fetus was removed by a razor blade, treated with trypsin, and then sub- cultured to establish somatic cells.
  • the fluorescent protein is preferably GFP, and enhanced GFP (EGFP) is more preferable, but it is not always limited thereto.
  • All the known expression vectors containing the fluorescent protein may be used as an expression vector only if they can be expressed in pig cells.
  • pCX-EGFP/neo was used as an expression vector including a fluorescent protein.
  • All the known promoters may be used without limitation as a promoter to express the fluorescent protein.
  • chicken ⁇ -actin promoter known to induce an specific expression of a fluorescent protein in muscle tissues, was used.
  • the expression vector containing a selection marker gene was used to select somatic cells transfected with the expression vector of the step 2) .
  • an antibiotics-resistant gene was preferably used.
  • an antibiotics-resistant gene includes neo r , pac r , bsr r , hph r , etc and neo r was preferably used in this invention.
  • somatic cells having an expression vector containing a fluorescent protein gene were selected by the treatment of G418.
  • the present inventors inserted GFP gene into porcine somatic cells in the same manner above and then separated cloned porcine somatic cell lines expressing GFP. Then, the present inventors produced a transgenic pig specifically expressing a green fluorescent protein in muscle tissues by transferring the established somatic cell lines into enucleated oocyte.
  • oocyte-somatic cell complex was successfully cultured to blstocyst after nuclear transfer with porcine somatic cells containing GFP (Uhm SJ et al . , Mol . Reprod. Dev. , 2000, 57:331-337; Koo D-B et al . , Mol . Reprod. Dev. , 2001, 58:15-21; Park KW et al . , Biol . Reprod. , 2001, 65:1681-1685), and a surrogate pig was transplanted with a replicative egg to give birth to a piglet expressing GFP (Park KW et al . , Anim, Biotech .
  • the problem of the conventional research was that retrovirus was used to transfer GFP into cells.
  • the size of a gene to be transferred into a cell is limited, causing a problem of stability in clinical use. It was one of the major problems to use a retrovirus so as to produce a transgenic animal.
  • the present inventors have overcome the problem by transferring a fluorescent protein gene into a somatic cell successfully using as a carrier a lipid instead of retrovirus.
  • a cloned somatic cell line has not been established. Cloned somatic cell lines have the same integration regions of the expression vector in the genome. If a cloned somatic cell line is established by transfection with the above expression vector, a fluorescent protein may be present only in the same regions of the somatic cells. According to a region on the chromosomes where a fluorescent protein gene is inserted, the expression pattern of the protein differs in each cell. So, when such cell lines was used to produce a transgenic pig, the expression patterns of a fluorescent protein was different in every pigs (Park KW et al . , Anim . Biotech .
  • a cloned somatic cell line was prepared by overcoming the problem of the technique.
  • This technique is expected to enhance production efficiency of a transgenic animal. That is, a gene encoding a protein of interest is randomly inserted into a genome of a somatic cell to obtain various cloned cell lines having different gene insertion regions. Those cell lines were used to produce cloned animals and then the expression patterns of a target protein from the cloned animals were examined to select individuals having high expression rate. It is because that the expression rate may become different in the cell lines having different insertion regions of the expression vector into a genome . In earlier studies, CMV promoter was used to express a foreign gene in whole body.
  • chicken- ⁇ actin promoter was used in this method of the present invention to produce a transgenic pig, showing that a fluorescent protein was strongly expressed in muscle-related tissues (i.e., hoofs, nose, myocardium, intestines, etc.) (FIGs. 3 and 4) .
  • the expression of a fluorescent protein was not detected in viscera, bone, and brain except an intestine.
  • a tissue-specific expression technique is highly useful for producing a therapeutic protein by using a transgenic animal. For example, if a gene is controlled to specifically express a target protein in the mammary gland, the target protein can be obtained from milk.
  • a transgenic pig produced by the method of the present invention expresses GFP in muscle tissues of a whole body, so that GFP cells taken from the transgenic pig can aid to understand vital phenomenon.
  • Organs of a pig are similar to those of a human in the physiological aspect. Efforts have been made to transplant an organ of a pig into a human, which is still unsuccessful because of rejection of xenograft. Recently, a transgenic pig, in which a specific gene involved in xenograft rejection was knock out, was produced, promoting possibilities of transplanting a xenograft by using a transgenic pig (Lai L et al .
  • a xenograft of a pig organ may be successfully transplanted into human
  • a porcine somatic cell may also be introduced into human. In that case, the development and differentiation of the transplanted somatic cells in a human patient may be easily observed by using the cloned somatic cell line containing the fluorescent protein gene of the present invention.
  • Fig. 1 is a schematic diagram showing the cleavage mapping of pCX-EGFP/neo vector.
  • Fig. 2 is an electrophoresis photograph showing (A) the result of PCR to investigate the presence of GFP gene in geno ic DNA of a transgenic pig, and (B) the result of FISH to analyze chromosome of a cloned pig and localize an insertion region of GFP gene on chromosome of a cloned pig.
  • Fig. 1 is a schematic diagram showing the cleavage mapping of pCX-EGFP/neo vector.
  • Fig. 2 is an electrophoresis photograph showing (A) the result of PCR to investigate the presence of GFP gene in geno ic DNA of a transgenic pig, and (B) the result of FISH to analyze chromosome of a cloned pig and localize an insertion region of GFP gene on chromosome of a clone
  • FIG. 3 is a set of photographs showing the expression patterns of GFP (green fluorescent protein) in a donor cell, an NT embryo and an offspring: A and B) Cultured fetal fibroblast cells; C and D) An NT blastocyst cultured in vi tro for 6 days; and E) a NT piglet.
  • B, D and E are photographs examining GFP expressions by using fluorescence .
  • Fig. 4 is a set of photographs showing the GFP expressions in the tissues of cloned NT piglets: A to U) Tissues under normal light; and A' to U' ) Tissues under fluorescent light.
  • a and A' Hoof
  • B and B' nose
  • C and C Ribs with skeletal muscle
  • D and D' Heart
  • E and E' the expression patterns of GFP (green fluorescent protein) in a donor cell, an NT embryo and an offspring: A and B) Cultured fetal fibroblast cells; C and D) An NT blastoc
  • Tongue, F and F' Kidney, G and G' : Skeletal muscle (non-transgenic pig, negative control) , H and H' :
  • L and L' Ear
  • M and M' Spinal cord
  • N and N' Skeletal muscle
  • O and O' Umbilical cord
  • FIG. 5 is a photograph showing two transgenic pigs.
  • a porcine fetus at day 35 after pregnancy was cut into small pieces with razor blades.
  • Cells were incubated in DMEM medium (bio-whittaka . Inc) supplemented with trypsin-EDTA for 5 minutes.
  • the cell culture was centrifuged at 1,000 rpm for 5 minutes to obtain supernatant, which was further cultured in DMEM medium supplemented with 10% FCS (Fetal Calf Serum, Hyclone . Inc) .
  • FCS Fetal Calf Serum, Hyclone . Inc
  • pCX-EGFP vector (Kato M et al . , Mol . Reprod. Dev. , 1999, 54:43-48) was connected with neo gene to construct a pCX-EGFP/neo vector (FIG. 1) .
  • the vector pCX- EGFP/neo was constructed by separating neo gene from vector "pPNT" (Tybulewicz VL et al . , Cell . 1991, 65:1153-1163) and attaching the neo gene onto HindiII site of pCX-EGFP.
  • the pCX-EGFP/neo vector was cut with Sail (Roche.
  • the transfected cells were treated with 250 / ⁇ g/m# of G418 for 2 weeks and then colony-forming cell lines were selected.
  • the resulting cloned cell lines were frozen at -70 ° C for nuclear transfer.
  • GFP expression in the selected cloned cell lines were observed under a fluorescent microscope.
  • a cell line PCX-8 expressing over 90% of GFP was selected (FIG. 3) .
  • the present inventors deposited the cloned cell line PCX-8 in the Korean Cell Line Research Foundation (KCLRF) on May 15, 2003 (Accession No: KCLRF-BP-00079) .
  • oocytes Preparation and culture of oocytes Ovaries were collected from prepubertal gilts at a local abattoir and stored in 0.9% NaCl solution at 35-39 ° C. Cumulus-oocyte complexes (COCs) were inspired from 2-6 mm diameter of antral follicles using an 18-gauge needle fixed to a 10 m# disposable syringe. COCs were washed with tissue culture medium. 50-60 COCs were added to a 500 ⁇ JL of culture solution, then cultured for 42-46 hours.
  • COCs Cumulus-oocyte complexes
  • the tissue culture medium was prepared by supplementing TCM 199 (31100035; Gibco, Grand Island, NY) with 0.1% polyvinylalcohol, 3.05 mM D-glucose, 0.91 mM sodium pyruvate, 0.57 irvM cysteine, 0.5 ⁇ g/ ⁇ l LH (L-5269, Sigma Chemical Co. St. Louis, MO), 0.5 ⁇ g/wl FSH (F-2293, Sigma), 10 ng/m£ epidermal growth factor (E-4127, Sigma), 75 ⁇ g/mi penicillin G and 50 ⁇ g/ml streptomycin.
  • Example ⁇ 3-l> Micromanipulation COCs obtained in the above Example ⁇ 3-l> were cultured in micromanipulated medium for 5-10 minutes.
  • the cloned cell line PCX-8, selected in the above Example 2 was added to the micromanipulated medium which was prepared by supplementing TCM 199 with 0.3% BSA and 7.5 ⁇ g/wl CB (cytochalasin B) .
  • the polar body of oocytes were fixed in the direction of 1 o'clock, and a fine glass pipette (30 ⁇ rn diameter) having 30-60° beveled grinded point was put into a polar body.
  • the polar body and about 10- 20% of cytoplasm near the polar body were inspired by oil pressure.
  • the injected oocyte was transferred into the medium consisting of 0.3 M mannitol, 1.0 mM CaCl 2 -H 2 0, 0.1 mM MgCl 2 -6H 2 0 and 0.5 mM HEPES, and the oocyte-somatic cell complex was placed between two platinum electrodes which are 1 mm apart in a medium.
  • Cell fusion/activation was induced simultaneously with two successive DC pulses of 1.1 kV/cm for 30 ⁇ sec using a BTX Elector-Cell Manipulator 2001 (BTX, San Diego, CA) .
  • BTX BTX, San Diego, CA
  • Example ⁇ 3-3> 20 to 30 reconstructed embryos prepared in the above Example ⁇ 3-3> were selected and cultured for 6 days in a 4-well culture dish containing 500 ⁇ i of development medium. Then, all embryos were stained with Hoechst 33342 (5 ) to determine the number of nuclei by epi-fluorescent microscopy.
  • the development medium was prepared by adding 0.4% BSA to NCSU-23 medium (North Carolina State University- 23; Petters, RM. and Wells, KD., J. Reprod. Fertil . Suppl . , 1993, 48:61-73).
  • PCX-8 cell line was used for nuclear transfer and in vitro development of each stage from external fertilization to blastocysts was investigated.
  • the replicative embryos at day 1 or 2 after the production were transplanted into 20 surrogate pigs in estrus (DO or Dl : the beginning day of estrus cycle or the next day) by surgical operation.
  • Ultrasonography was performed on the 28 th day after the transplantation, confirming early pregnancy. And the stable maintenance of pregnancy was confirmed by using ultrasonography once a week.
  • eleven healthy female piglets and one stillbirth were delivered from three surrogate sows.
  • the mean birth weight was 1094 g and ranged from 680 to 1580 g (Table 2 and FIG. 5) .
  • PCR analysis was performed to investigate the expression of GFP in the piglet above and cloned cell line PCX-8. Particularly, proteinase K (Gibco Inc.) was treated to all piglets' ear skin cells, donor cells, and surrogate skin cells to decompose proteins. Then, genomic DNA was isolated by using phenol-extraction. Two PCR primers represented by SEQ ID No. 1 and No. 2 were used. PCR was performed as follows; predenaturation at 95°C for 5 minutes, denaturation at 95°C for 1 minute, annealing at 60°C for 1 minute, polymerization at 72°C for 1 minute, 30 cycles from denaturation to polymerization, and final extension at 72°C for 10 minutes.
  • the PCR product was transferred on to agarose-gel for electrophoresis .
  • 500 bp of EGFP DNA product was present in both of piglets and PCX-8 cell lines, but not in ear tissues of surrogate pigs (FIG. 2A) .
  • Chromosome analysis of the donor cells and cultured ear cells of piglets suggested that the chromosomes isolated from all the piglets were present in the normal state (FIG. 2B) .
  • FISH analysis showed that the GFP gene was located close to the telomere of chromosome 7 (C7) of the piglet derived from the PCX-8 cell lines, showing that it was present in the same region as in the metaphase spreads (FIG. 2B) .
  • the result indicated that all the produced piglets were the transgenic cloned pigs derived from the PCX-8 clonal cell line.
  • GFP image was acquired using Las 3000
  • GFP image was taken using blue light (460 nm) for excitation and 510DF10 filter.
  • the piglets were dissected and various organs were removed. Specimens were also placed in the dark box and GFP expression was captured. As a result, strong expression of fluorescence was observed outside body of piglets especially in tongue, eye periphery, foreleg, lumbar region and hoof (FIG. 3).
  • To compare the GFP expression intensity of each organ twenty-one tissues were separated from two piglets (GFl-1, GF2-4) .
  • GFP expression was mainly detected in muscle related tissues (hoof, nose, heart, tongue, skin, ear, skeletal muscle, and intestine), but was not detected in most internal organs except bone, bone marrow, brain, intestine, etc. (FIG. 4).
  • the transgenic pig expressing green fluorescent protein (GFP) produced according to the method of the present invention can be useful in the fields of the basic studies of science including cell therapy and in the production of a therapeutic protein by the transgenic animals.
  • GFP green fluorescent protein
  • Nucelotide sequences represented by SEQ ID No. 1 and No. 2 are primer sequences used in the PCR reaction of Example 5.
  • a The Mowing manifestation is directed to the deposited microorganism or other biological materials described in the line 21 of page 8 of the present application.
  • B. Identification of tine Microorganism An additional rrfcroorganismte) is described in the next page(s) e ⁇ cepl the one D Depository Authority Korean Ceil Line Research Foundation (RCLRP) Address (Zip code and Nationality) Korean Cell Line Research Foundation (KCLRF) Cancer Research Institute, Seoul National University College of Medicine 28 Ye ⁇ ageon-dong, Jongno-gu, Seoul 110-744, Republic of K rea Date Accession number May 24 2004 I ⁇ CLRF-BP-0007S C, Additional Manifestation (filed as "blank” if there is no disclosure) To be continued to the additional n x page D
  • RCLRP Korean Ceil Line Research Foundation
  • KCLRF Korean Cell Line Research Foundation

Abstract

The present invention relates to a method of producing a transgenic pig expressing green fluorescent protein (GFP). Particularly, the present invention relates to the method of producing the above transgenic pig including the steps of; (1) separating a somatic cell from a porcine fetus; (2) preparing the expression vector which includes a green fluorescent gene, followed by introducing the above expression vector into the above somatic cell; (3) selecting a cloned somatic cell transduced with the above expression vector and culturing that selected cell; (4) eliminating a nucleus from the oocyte of the donor pig, followed by fusing the resulting oocyte with the somatic cell to form a oocyte-somatic cell complex; and (5) transplanting the fused replicative oocyte into a surrogate pig and delivering piglets of present invention. The transgenic pigs expressing green fluorescent protein produced according to the method of the present invention can be effectively used as a transgenic animal model in the fields of the basic studies of science including cell therapy and in the production of a therapeutic protein by the transgenic animals.

Description

Invention Title
METHOD OF PRODUCING TRANSGENIC PIGS EXPRESSING GREEN FLUORESCENT PROTEIN
Technical Field
The present invention relates to a method of producing a transgenic pig expressing green fluorescent protein (GFP) , more specifically to a method of producing a transgenic pig expressing green fluorescent protein available for the field of producing therapeutic proteins and for the cell therapy.
Background Art
Cloning means producing a number of genetically identical individuals. One of the cloning techniques is an attractive nuclear transfer resulting from a great progress of molecular biology. A blastomere of a fertilized egg was used as a nuclear source in the early nuclear transfer (Prather, et al., Biol . Reprod. , 1987, 37:856-866; Prather, et al . , Biol . Reprod. , 1989, 41:414-418), but it is recently performed by using the nucleus of a somatic cell. A cloning technique of a somatic cell is to introduce an nucleus of a differentiated somatic cell into an enucleated oocyte to develop it in the same manner as that of a general fertilized egg. It is expected that this cloning technique is not only available for the studies of basic science including developmental biology but also for production of a therapeutic protein, especially in the field of medical science, for example developing a disease model and organ transplantation etc, so that it may be greatly useful for the industrial availability.
Since the cloning of the first somatic cell cloned animal "Dolly" (Willmut, I. et al . , Na ture, 1997, 385:810-813), progress has been made in cloning animals, leading to the successful cloning of a cow (Cibelli, JB . et al . , Science, 1998, 280:1256-1258; Wells, DN. et al . , Reprod. Fertil . Dev. , 1998, 10:369-378), a rat (Wakayama, T. et al . , Na ture, 1998, 394:369-374) and a goat (Bagusi, A. et al., Na t . Biotechnol . , 1999, 17:456-461). But in the case of pig cloning, the studies have been nearly made about the fertilized egg of pig and its cloning has succeeded just recently because of difficult physiological characteristics of pigs, for example their pregnancy capable of being maintained only when at least 4 embryos are implanted in their uteruses, etc (Poleajaeva, IA. et al . , Na ture, 2000, 407:86-90; Onishi, A. et al . , Science, 2000, 289:1188-1190; Betthauser, J. et al . , Na t . Biotechnol . , 2000, 18:1055-1059). The first transgenic cloned pig was then produced by using the somatic cells introduced a exogenous gene (Park, KW. et al., Anim . Biotechnol . , 2001, 12:173-181). Then, another cloned pig, in which a specific gene was knockout in a somatic cell, was produced (Lai, L. et al., Na t . Biotechnol . , 2002 Mar, 20 (3 ): 251-255 ) . Although somatic cell cloning with pigs has been rapidly progressed, the cloning efficiency is still very low with 1-5%, and therefore further studies are required to improve the efficiency.
Somatic cell cloning includes the steps of eliminating a nucleus from an oocyte, injecting a somatic cell into the enucleated oocyte, followed by fusing the resulting oocyte with the somatic cell to form a oocyte-somatic cell complex and activating it under electro-stimulation. The conventional method, for example microinjection of DNA has a problem of low productivity of a transgenic animal. Because GFP (green fluorescent protein) derived from fluorescent jellyfish (i.e., Aequorea ictoria ) specifically expresses strong fluorescence when this protein is excited by the ultraviolet radiation, it is widely used as a marker protein in the various fields of animals, plants and microorganisms (Takada T et al., Na t . Biotechnol . , 1997, 15:458-461; Perry AC et al., Science, 1999, 284:1180-1183; Chan AW et al., Mol . Reprod. , 1999, 52:406-413). Recently, it was confirmed that a successful development of a cloned egg was made from a pig somatic cell transfected with GFP gene (Uhm SJ et al . , Mol . Reprod. Dev. , 2000, 57:331-337; Koo D-B et al . , Mol . Reprod. Dev. , 2001, 58:15-21; Park KW et al . , Biol . Reprod. , 2001, 65:1681-1685), and the first transgenic cloned pig expressing GFP gene was produced based on that (Park KW et al . , Anim . Biotech . , 2001, 12:173-181). However, since somatic cells used for the cloning above were not clonal somatic cells, integration sites of GFP gene into chromosome were all different among offsprings. Retrovirus was used to insert GFP gene into cells, which has problems of limitation of size of a gene to be transferred to cells and difficulty in securing stability for a clinical application. Thus, in order to overcome such problems of a conventional method for producing a transgenic cloned pig, the present inventors prepared a clonal somatic cell line transfected with a foreign gene using an lipid-based reagent instead of retrovirus. And the present inventors completed this invention by confirming that a transgenic cloned pig specifically expressing a green fluorescent protein in the muscle tissues may be produced using the nuclear transfer technique of the clonal somatic cells .
Disclosure
Technical Problem It is an object of this invention to provide a method of producing a transgenic pig specifically expressing green fluorescent protein in muscle tissues . It is another object of this invention to provide the transgenic pig produced according to the method of the present invention, wherein the transgenic pig specifically expresses green fluorescent protein in muscle tissues. Technical Solution
In order to achieve the above objects, the present invention provides a method of producing a transgenic pig expressing a green fluorescent protein including the steps of; 1) separating somatic cells from a porcine fetus; 2) preparing an expression vector which includes a green fluorescent gene, followed by introducing the expression vector into the above somatic cells; 3) selecting cloned somatic cells transfected with the above expression vector and culturing those selected cells; 4) eliminating a nucleus from an oocyte of a donor pig, followed by fusing the resulting oocyte with the somatic cell to form a oocyte-somatic cell complex; and 5) transplanting the fused replicative oocyte into a surrogate pig and delivering piglets of present invention. The present invention also provides a transgenic pig specifically expressing a green fluorescent protein in muscle tissues produced by the method of the present invention. The term "cloned somatic cell lines" used herein means cell lines having the identical integration site of a foreign vector. The term "perivitelline space" means the space between zona pellucida and ooplasm of an egg. The term "oocyte-somatic cell complex" means pre-cell fusion status after eliminating a nucleus from an oocyte and injecting a somatic cell into its perivitelline space. And the term "a cloned egg" (i.e., a replicative egg) means a state that the oocyte is fused with the somatic cell by electro- stimulation . Hereinafter, the present invention is described in detail. The present invention provides a method of producing a transgenic pig expressing a green fluorescent protein, comprising the following steps: 1) Establishing somatic cells from a porcine fetus; 2) Preparing an expression vector which includes a green fluorescent gene, followed by introducing the expression vector into the above somatic cells; 3) Selecting cloned somatic cells transfected with the above expression vector and culturing those selected cells; 4) Eliminating a nucleus from an oocyte of a donor pig, followed by fusing the resulting oocyte with the somatic cell to form a oocyte-somatic cell complex; and 5) Transplanting the fused replicative oocyte into a surrogate pig and delivering piglets of present invention.
In the above step 1), a porcine fetus is preferably an embryo at day 20 - 50 after pregnancy, and is more preferably an embryo at day 30 - 40 after pregnancy. In particular, the present inventors established somatic cells from a porcine fetus at day 35 after pregnancy. To establish somatic cells from the porcine fetus, a conventional method was applied equally to this invention and, in the preferred embodiment of the present invention a small portion of a porcine fetus was removed by a razor blade, treated with trypsin, and then sub- cultured to establish somatic cells. In the above step 2), the fluorescent protein is preferably GFP, and enhanced GFP (EGFP) is more preferable, but it is not always limited thereto. All the known expression vectors containing the fluorescent protein may be used as an expression vector only if they can be expressed in pig cells. Especially in the preferred embodiment of this invention, pCX-EGFP/neo was used as an expression vector including a fluorescent protein. All the known promoters may be used without limitation as a promoter to express the fluorescent protein. Particularly in the preferred embodiment of this invention, chicken β-actin promoter, known to induce an specific expression of a fluorescent protein in muscle tissues, was used. In the above step 3), the expression vector containing a selection marker gene was used to select somatic cells transfected with the expression vector of the step 2) . As a selection marker, an antibiotics-resistant gene was preferably used. The example of an antibiotics-resistant gene includes neor, pacr, bsrr, hphr, etc and neor was preferably used in this invention. In the preferred embodiment of the present invention, somatic cells having an expression vector containing a fluorescent protein gene were selected by the treatment of G418. The present inventors inserted GFP gene into porcine somatic cells in the same manner above and then separated cloned porcine somatic cell lines expressing GFP. Then, the present inventors produced a transgenic pig specifically expressing a green fluorescent protein in muscle tissues by transferring the established somatic cell lines into enucleated oocyte.
Advantageous Effects According to recent reports, oocyte-somatic cell complex was successfully cultured to blstocyst after nuclear transfer with porcine somatic cells containing GFP (Uhm SJ et al . , Mol . Reprod. Dev. , 2000, 57:331-337; Koo D-B et al . , Mol . Reprod. Dev. , 2001, 58:15-21; Park KW et al . , Biol . Reprod. , 2001, 65:1681-1685), and a surrogate pig was transplanted with a replicative egg to give birth to a piglet expressing GFP (Park KW et al . , Anim, Biotech . , 2001, 12:173-181). The problem of the conventional research, though, was that retrovirus was used to transfer GFP into cells. In this case, the size of a gene to be transferred into a cell is limited, causing a problem of stability in clinical use. It was one of the major problems to use a retrovirus so as to produce a transgenic animal. The present inventors have overcome the problem by transferring a fluorescent protein gene into a somatic cell successfully using as a carrier a lipid instead of retrovirus. Thus, in the method of the present invention there is almost not limited to the size of a gene to be tranfered into a somatic cell.
Although earlier studies were successfully made only in introduction of a fluorescent protein gene into the somatic cell, a cloned somatic cell line has not been established. Cloned somatic cell lines have the same integration regions of the expression vector in the genome. If a cloned somatic cell line is established by transfection with the above expression vector, a fluorescent protein may be present only in the same regions of the somatic cells. According to a region on the chromosomes where a fluorescent protein gene is inserted, the expression pattern of the protein differs in each cell. So, when such cell lines was used to produce a transgenic pig, the expression patterns of a fluorescent protein was different in every pigs (Park KW et al . , Anim . Biotech . , 2001, 12:173-181). In the present invention, a cloned somatic cell line was prepared by overcoming the problem of the technique. This technique is expected to enhance production efficiency of a transgenic animal. That is, a gene encoding a protein of interest is randomly inserted into a genome of a somatic cell to obtain various cloned cell lines having different gene insertion regions. Those cell lines were used to produce cloned animals and then the expression patterns of a target protein from the cloned animals were examined to select individuals having high expression rate. It is because that the expression rate may become different in the cell lines having different insertion regions of the expression vector into a genome . In earlier studies, CMV promoter was used to express a foreign gene in whole body. On the contrary, chicken-β actin promoter was used in this method of the present invention to produce a transgenic pig, showing that a fluorescent protein was strongly expressed in muscle-related tissues (i.e., hoofs, nose, myocardium, intestines, etc.) (FIGs. 3 and 4) . The expression of a fluorescent protein was not detected in viscera, bone, and brain except an intestine. A tissue-specific expression technique is highly useful for producing a therapeutic protein by using a transgenic animal. For example, if a gene is controlled to specifically express a target protein in the mammary gland, the target protein can be obtained from milk. A transgenic pig produced by the method of the present invention expresses GFP in muscle tissues of a whole body, so that GFP cells taken from the transgenic pig can aid to understand vital phenomenon. Organs of a pig are similar to those of a human in the physiological aspect. Efforts have been made to transplant an organ of a pig into a human, which is still unsuccessful because of rejection of xenograft. Recently, a transgenic pig, in which a specific gene involved in xenograft rejection was knock out, was produced, promoting possibilities of transplanting a xenograft by using a transgenic pig (Lai L et al . , Science, 2002, 295:1089-1092; Dai Y et al., Na t . Biotechnol . , 2002, 20:251-255). If a xenograft of a pig organ may be successfully transplanted into human, a porcine somatic cell may also be introduced into human. In that case, the development and differentiation of the transplanted somatic cells in a human patient may be easily observed by using the cloned somatic cell line containing the fluorescent protein gene of the present invention.
Description of Drawings
Fig. 1 is a schematic diagram showing the cleavage mapping of pCX-EGFP/neo vector. Fig. 2 is an electrophoresis photograph showing (A) the result of PCR to investigate the presence of GFP gene in geno ic DNA of a transgenic pig, and (B) the result of FISH to analyze chromosome of a cloned pig and localize an insertion region of GFP gene on chromosome of a cloned pig. Fig. 3 is a set of photographs showing the expression patterns of GFP (green fluorescent protein) in a donor cell, an NT embryo and an offspring: A and B) Cultured fetal fibroblast cells; C and D) An NT blastocyst cultured in vi tro for 6 days; and E) a NT piglet. In particular, B, D and E are photographs examining GFP expressions by using fluorescence . Fig. 4 is a set of photographs showing the GFP expressions in the tissues of cloned NT piglets: A to U) Tissues under normal light; and A' to U' ) Tissues under fluorescent light. In Fig 4; A and A' : Hoof, B and B' : nose, C and C : Ribs with skeletal muscle, D and D' : Heart, E and E' :
Tongue, F and F' : Kidney, G and G' : Skeletal muscle (non-transgenic pig, negative control) , H and H' :
Brain, I and I' : Skin, J and J' : Thymus, K and K' :
Ovary, L and L' : Ear, M and M' : Spinal cord, N and N' : Skeletal muscle, O and O' : Umbilical cord, P and
P' : Frontal bone, Q and 0/ : Pancreas, R and R' : Liver, S and S' : Thyroid gland, T and T' : Lung, U and U' : Small intestine. FIG. 5 is a photograph showing two transgenic pigs.
Mode for Invention
Practical and presently preferred embodiments of the present invention are illustrative as shown in the following Examples. However, it will be appreciated from those skilled in the art that on consideration of this disclosure, modifications and improvements may be made within the spirit and scope of the present invention, without limiting thereto.
<Example 1> Separation of porcine somatic cells
A porcine fetus at day 35 after pregnancy was cut into small pieces with razor blades. Cells were incubated in DMEM medium (bio-whittaka . Inc) supplemented with trypsin-EDTA for 5 minutes. The cell culture was centrifuged at 1,000 rpm for 5 minutes to obtain supernatant, which was further cultured in DMEM medium supplemented with 10% FCS (Fetal Calf Serum, Hyclone . Inc) . When cells became 90% confluent, they were sub-cultured after trypsin was added. Some of them were used for nuclear transfer and the others were stored in -70 for a future use.
<Example 2> Preparation of cloned porcine cell
lines expressing GFP
To prepare a vector containing GFP gene, pCX- EGFP vector (Kato M et al . , Mol . Reprod. Dev. , 1999, 54:43-48) was connected with neo gene to construct a pCX-EGFP/neo vector (FIG. 1) . The vector pCX- EGFP/neo was constructed by separating neo gene from vector "pPNT" (Tybulewicz VL et al . , Cell . 1991, 65:1153-1163) and attaching the neo gene onto HindiII site of pCX-EGFP. The pCX-EGFP/neo vector was cut with Sail (Roche. Inc) and the target DNA was isolated by phenol-extraction. The DNA was then transfected into somatic cells by the lipid-based method (Fugene 6; Roche. Inc) according to the manufacturer's protocols (Sharon J.H. et al . , Transgenic Research , 2002, 11:143-150). The cell was cultured for 48 hours. Then, G418 (Geneticin; Gibco. Inc) was added thereto to select transfected somatic cells. Their fluorescence was observed under a fluorescent microscope (Nikon) . G418 was added to the cell culture at various concentrations to determine the optimum concentration (250 βg/ l ) . The transfected cells were treated with 250 /~g/m# of G418 for 2 weeks and then colony-forming cell lines were selected. The resulting cloned cell lines were frozen at -70 °C for nuclear transfer. GFP expression in the selected cloned cell lines were observed under a fluorescent microscope. As a result, a cell line PCX-8 expressing over 90% of GFP was selected (FIG. 3) .
The present inventors deposited the cloned cell line PCX-8 in the Korean Cell Line Research Foundation (KCLRF) on May 15, 2003 (Accession No: KCLRF-BP-00079) .
<Example 3> Nuclear transfer using the cloned
cell
<3-l> Preparation and culture of oocytes Ovaries were collected from prepubertal gilts at a local abattoir and stored in 0.9% NaCl solution at 35-39°C. Cumulus-oocyte complexes (COCs) were inspired from 2-6 mm diameter of antral follicles using an 18-gauge needle fixed to a 10 m# disposable syringe. COCs were washed with tissue culture medium. 50-60 COCs were added to a 500 μJL of culture solution, then cultured for 42-46 hours. At that time, the tissue culture medium was prepared by supplementing TCM 199 (31100035; Gibco, Grand Island, NY) with 0.1% polyvinylalcohol, 3.05 mM D-glucose, 0.91 mM sodium pyruvate, 0.57 irvM cysteine, 0.5 βg/ϊ l LH (L-5269, Sigma Chemical Co. St. Louis, MO), 0.5 μg/wl FSH (F-2293, Sigma), 10 ng/m£ epidermal growth factor (E-4127, Sigma), 75 βg/mi penicillin G and 50 ~g/ml streptomycin.
<3-2> Micromanipulation COCs obtained in the above Example <3-l> were cultured in micromanipulated medium for 5-10 minutes. The cloned cell line PCX-8, selected in the above Example 2, was added to the micromanipulated medium which was prepared by supplementing TCM 199 with 0.3% BSA and 7.5 βg/wl CB (cytochalasin B) . In order to eliminate a nucleus from an oocyte, the polar body of oocytes were fixed in the direction of 1 o'clock, and a fine glass pipette (30 μrn diameter) having 30-60° beveled grinded point was put into a polar body. The polar body and about 10- 20% of cytoplasm near the polar body were inspired by oil pressure. Most of the nuclei of oocytes were located around polar body, so that they were successfully eliminated by the inspiration above. The porcine somatic cells prepared in the above Example 2 were then inspired through the fine glass pipette, the fine glass pipette was put into the perivitelline space of enucleated oocyte, and then somatic cells were injected into the perivitelline space of the oocyte by the oil pressure.
<3-3> Cell fusion and activation
When micromanipulation was completed in the Example <3-2>, the injected oocyte was transferred into the medium consisting of 0.3 M mannitol, 1.0 mM CaCl2-H20, 0.1 mM MgCl2-6H20 and 0.5 mM HEPES, and the oocyte-somatic cell complex was placed between two platinum electrodes which are 1 mm apart in a medium. Cell fusion/activation was induced simultaneously with two successive DC pulses of 1.1 kV/cm for 30 μsec using a BTX Elector-Cell Manipulator 2001 (BTX, San Diego, CA) . As a control a native oocyte was electrically activated in the same manner described above. The level of fusion was determined at 0.5-1 hr after the DC pulses. <3-4> Cultivation/Development of replicative
embryos
20 to 30 reconstructed embryos prepared in the above Example <3-3> were selected and cultured for 6 days in a 4-well culture dish containing 500 βi of development medium. Then, all embryos were stained with Hoechst 33342 (5
Figure imgf000021_0001
) to determine the number of nuclei by epi-fluorescent microscopy. The development medium was prepared by adding 0.4% BSA to NCSU-23 medium (North Carolina State University- 23; Petters, RM. and Wells, KD., J. Reprod. Fertil . Suppl . , 1993, 48:61-73). PCX-8 cell line was used for nuclear transfer and in vitro development of each stage from external fertilization to blastocysts was investigated. As a result, fusion rate, division rate and each developmental rate up to blastocyst stage of the fused replicative embryos were 78.5%, 67.2%, and 12.9% respectively, and the number of nuclei of blastocysts was 23.8 (Table 1). Strong GFP expression was also observed in blastocysts (FIG. 3),
<Table 1> Development of Nuclear Transfer Embryos
Figure imgf000022_0001
<Example 4> Transplantation of replicative
embryos and production of transgenic piglets
The replicative embryos at day 1 or 2 after the production were transplanted into 20 surrogate pigs in estrus (DO or Dl : the beginning day of estrus cycle or the next day) by surgical operation. Ultrasonography was performed on the 28th day after the transplantation, confirming early pregnancy. And the stable maintenance of pregnancy was confirmed by using ultrasonography once a week. As a result, eleven healthy female piglets and one stillbirth were delivered from three surrogate sows. The mean birth weight was 1094 g and ranged from 680 to 1580 g (Table 2 and FIG. 5) .
<Table 2>
Cloned Piglets Derived from Nuclear Transfer.
Figure imgf000023_0001
<Example 5> Determination of transfected GFP
gene
PCR analysis was performed to investigate the expression of GFP in the piglet above and cloned cell line PCX-8. Particularly, proteinase K (Gibco Inc.) was treated to all piglets' ear skin cells, donor cells, and surrogate skin cells to decompose proteins. Then, genomic DNA was isolated by using phenol-extraction. Two PCR primers represented by SEQ ID No. 1 and No. 2 were used. PCR was performed as follows; predenaturation at 95°C for 5 minutes, denaturation at 95°C for 1 minute, annealing at 60°C for 1 minute, polymerization at 72°C for 1 minute, 30 cycles from denaturation to polymerization, and final extension at 72°C for 10 minutes. The PCR product was transferred on to agarose-gel for electrophoresis . As a result, it was confirmed that 500 bp of EGFP DNA product was present in both of piglets and PCX-8 cell lines, but not in ear tissues of surrogate pigs (FIG. 2A) . Chromosome analysis of the donor cells and cultured ear cells of piglets suggested that the chromosomes isolated from all the piglets were present in the normal state (FIG. 2B) . FISH analysis showed that the GFP gene was located close to the telomere of chromosome 7 (C7) of the piglet derived from the PCX-8 cell lines, showing that it was present in the same region as in the metaphase spreads (FIG. 2B) . Thus, the result indicated that all the produced piglets were the transgenic cloned pigs derived from the PCX-8 clonal cell line. Next, GFP image was acquired using Las 3000
(FUJI FILM, Tokyo, Japan) equipped with high sensitive cooled CCD camera. After the stillbirth (GFl-1) or the piglet GF2-4 was placed in the dark- box, gray-scale body surface image was taken.
Afterwards, GFP image was taken using blue light (460 nm) for excitation and 510DF10 filter. The piglets were dissected and various organs were removed. Specimens were also placed in the dark box and GFP expression was captured. As a result, strong expression of fluorescence was observed outside body of piglets especially in tongue, eye periphery, foreleg, lumbar region and hoof (FIG. 3). To compare the GFP expression intensity of each organ, twenty-one tissues were separated from two piglets (GFl-1, GF2-4) . GFP expression was mainly detected in muscle related tissues (hoof, nose, heart, tongue, skin, ear, skeletal muscle, and intestine), but was not detected in most internal organs except bone, bone marrow, brain, intestine, etc. (FIG. 4).
Industrial Applicability
As explained hereinbefore, the transgenic pig expressing green fluorescent protein (GFP) produced according to the method of the present invention can be useful in the fields of the basic studies of science including cell therapy and in the production of a therapeutic protein by the transgenic animals.
Sequence List Text
Nucelotide sequences represented by SEQ ID No. 1 and No. 2 are primer sequences used in the PCR reaction of Example 5.
Manifestation om the Deposited Mcxoorgarasm or Otker Biological Materials
A The Mowing manifestation is directed to the deposited microorganism or other biological materials described in the line 21 of page 8 of the present application. B. Identification of tine Microorganism An additional rrfcroorganismte) is described in the next page(s) eκcepl the one D Depository Authority Korean Ceil Line Research Foundation (RCLRP) Address (Zip code and Nationality) Korean Cell Line Research Foundation (KCLRF) Cancer Research Institute, Seoul National University College of Medicine 28 Yeøageon-dong, Jongno-gu, Seoul 110-744, Republic of K rea Date Accession number May 24 2004 IΪCLRF-BP-0007S C, Additional Manifestation (filed as "blank" if there is no disclosure) To be continued to the additional n x page D
D. Characteristic Items directed to the Manifestation
E Any Additional Manifestation (filed as "blank;" if there is no disclosure) Any of the following manifestation will be filed to the International Bureau of the World Intellectual Property Organisation later. (General Properties of the manifestation should be specificaEy described)
Items for a Receiving Authority Items for the International Bureau □ Accepted with the international D Accepted by the International Bureau application Authorised OfficiaKs) Authorized OfficiaKs)
ECX/ftO/134(Iuly 1898)

Claims

Claim 1
A method of producing a transgenic pig expressing green fluorescent protein (GFP) comprising the following steps: 1) Separating somatic cells from a porcine fetus; 2) Preparing an expression vector which includes fluorescent protein gene, followed by introducing the above expression vector into the somatic cell of the step 1); 3) Selecting cloned somatic cells transduced with the above expression vector and culturing those selected cells; 4) Eliminating a nucleus from an oocyte of a donor pig, followed by fusing the resulting oocyte with the somatic cell to form a oocyte-somatic cell complex; and 5) Transplanting the fused replicative oocyte into a surrogate pig and delivering piglets .
Claim 2
The method of producing a transgenic pig as set forth in claim 1 , wherein the porcine fetus of the step 1) is at 30 - 40 days after pregnancy.
Claim 3
The method of producing a transgenic pig as set forth in claim 2, wherein the porcine fetus is at 35 days after pregnancy.
Claim 4
The method of producing a transgenic pig as set forth in claim 1, wherein the fluorescent protein in the step 2) is enhanced green fluorescent protein (EGFP) .
Claim 5 The method of producing a transgenic pig as set forth in claim 1, wherein the expression vector in the step 2) is pCX-EGFP/neo .
Claim 6 The method of producing a transgenic pig as set forth in claim 1, wherein the cloned somatic cell in the step 3) is a PCX-8 cloned cell line (Accession No: KCLRF-BP-00079) . Claim 7
A transgenic pig specifically expressing a green fluorescent protein in muscle tissues produced according to the method of claim 1.
PCT/KR2004/001414 2003-10-08 2004-06-14 Method of producing transgenic pigs expressing green fluorescent protein WO2005032245A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR1020030070031A KR100479704B1 (en) 2003-10-08 2003-10-08 Method of producing a transgenic pig expressing green fluorescent protein
KR10-2003-0070031 2003-10-08

Publications (1)

Publication Number Publication Date
WO2005032245A1 true WO2005032245A1 (en) 2005-04-14

Family

ID=34420551

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2004/001414 WO2005032245A1 (en) 2003-10-08 2004-06-14 Method of producing transgenic pigs expressing green fluorescent protein

Country Status (2)

Country Link
KR (1) KR100479704B1 (en)
WO (1) WO2005032245A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8183307B2 (en) 2006-08-04 2012-05-22 Wacker Chemie Ag Crosslinkable substances based on organosilicon compounds
US8546643B2 (en) 2007-03-07 2013-10-01 Aarhus Universitet Pig model for atherosclerosis

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009072835A2 (en) * 2007-12-06 2009-06-11 Industry-Academic Cooperation Foundation Gyeongsang National University Transgenic cats expressing red fluorescence protein and producing method thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002102143A1 (en) * 2001-06-15 2002-12-27 Japan Science And Technology Agency Transgenic animal transformed using green fluorescent protein gene

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002102143A1 (en) * 2001-06-15 2002-12-27 Japan Science And Technology Agency Transgenic animal transformed using green fluorescent protein gene

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CHAN A.W.S. ET AL.: "Transgenic monkeys produced by retroviral gene transfer into mature oocytes", SCIENCE, vol. 291, no. 5502, 2001, pages 309 - 312 *
KOO D.B. ET AL.: "Developmental potential and transgene expression of porcine nuclear transfer embryos using somatic cells", MOL. REPROD. DEV., vol. 58, no. 1, 15 January 2001 (2001-01-15) - 21 January 2001 (2001-01-21) *
PARK K.W. ET AL.: "Production of nuclear transfer-derived swine that express the enhanced green fluorescent protein", ANIM. BIOTECHNOL., vol. 12, no. 2, November 2001 (2001-11-01), pages 173 - 181 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8183307B2 (en) 2006-08-04 2012-05-22 Wacker Chemie Ag Crosslinkable substances based on organosilicon compounds
US8546643B2 (en) 2007-03-07 2013-10-01 Aarhus Universitet Pig model for atherosclerosis

Also Published As

Publication number Publication date
KR100479704B1 (en) 2005-03-31

Similar Documents

Publication Publication Date Title
US20100115641A1 (en) Gfp-transfected clon pig, gt knock-out clon pig and methods for productions thereof
US8258369B2 (en) Method for preparing fish embryos
JP4300287B2 (en) Induction of germ line differentiation by transplantation of isolated primordial germ cells
EP1229784A1 (en) An improved method for production of porcine clone embryos via somatic cell nuclear transfer
JP4581083B2 (en) Method for breeding fish by germ cell transplantation using triploid recipients
WO2005032245A1 (en) Method of producing transgenic pigs expressing green fluorescent protein
CN105238817B (en) A method of the clone pig of building overexpression Leptin gene
KR20070101266A (en) Methods for correcting mitotic spindle defects and optimizing preimplantation embryonic developmental rates associated with somatic cell nuclear transfer in animals
DK2993234T3 (en) Vector Targeted CMP Acetylneuramic Acid Hydroxylase, Transgenic Animal for Xenotransplantation Introduced with the Vector and Method of Preparation thereof
Matsuda et al. Production of transgenic chimera rabbit fetuses using somatic cell nuclear transfer
KR100502480B1 (en) Method of producing porcine clonal somatic cell line expressing green fluorescent protein
Naito Genetic manipulation in chickens
KR101270422B1 (en) Transgenic Cats Expressing Red Fluorescence Protein and Producing Method Thereof
JP2011508600A (en) Method for producing transformed replicated dogs
JP5374389B2 (en) Method for introducing foreign gene into early embryo of primate and method for producing transgenic primate animal containing the method
WO2015163711A1 (en) Talen targeting myostatin gene and method for making animal with knockout myostatin gene using same
JP2008220222A (en) Endogalc-hdaf double transgenic pig
JP2001103867A (en) Preparation of reconstructed fertilized egg and preparation of transgenic embryo using the egg
JP4696235B2 (en) Method for producing fish embryo
JP2002238562A (en) Gene recombinant rabbit and method for making the gene recombinant rabbit using somatic nuclear transplantation
KR20230092097A (en) Transgenic cloned piglets defecting porcine GGTA1, CMAH, beta4GalNT2 and CD40 gene for xenotransplantation, and producing method thereof
KR100975350B1 (en) Transgenic cloned cow producing human lactoferricin and method for producing the same
CN111718962A (en) Method for preparing cloned cat by somatic cell cloning
Spell et al. Somatic cell cloning in the beef industry
JPWO2002022789A1 (en) Method for producing normal parenchymal cells, tissues or organs using a biological incubator

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase